268
Views
9
CrossRef citations to date
0
Altmetric
Perspective

Nutraceutical induction and mimicry of heme oxygenase activity as a strategy for controlling excitotoxicity in brain trauma and ischemic stroke: focus on oxidative stress

& ORCID Icon
Pages 157-168 | Received 31 Oct 2020, Accepted 07 Dec 2020, Published online: 28 Dec 2020

References

  • Abdoli A, Rahimi-Bashar F, Torabian S, et al. Efficacy of simultaneous administration of nimodipine, progesterone, and magnesium sulfate in patients with severe traumatic brain injury: a randomized controlled trial. Bull Emerg Trauma. 2019;7:124–129.
  • Beez T, Steiger HJ, Etminan N. Pharmacological targeting of secondary brain damage following ischemic or hemorrhagic stroke, traumatic brain injury, and bacterial meningitis - a systematic review and meta-analysis. BMC Neurol. 2017;17:209–220.
  • Santos SC, Pelosi P, Leme SP, et al. Immunomodulation after ischemic stroke: potential mechanisms and implications for therapy. Crit Care. 2016;20:391–399.
  • Nishizawa Y. Glutamate release and neuronal damage in ischemia. Life Sci. 2001;69:369–381.
  • Lee SY, Kim JH. Mechanisms underlying presynaptic Ca2+ transient and vesicular glutamate release at a CNS nerve terminal during in vitro ischaemia. J Physiol. 2015;593:2793–2806.
  • Kosugi T, Kawahara K. Reversed actrocytic GLT-1 during ischemia is crucial to excitotoxic death of neurons, but contributes to the survival of astrocytes themselves. Neurochem Res. 2006;31:933–943.
  • Lai TW, Zhang S, Wang YT. Excitotoxicity and stroke: identifying novel targets for neuroprotection. Prog Neurobiol. 2014;115:157–188.
  • Rothman SM. Synaptic activity mediates death of hypoxic neurons. Science. 1983;220:536–537.
  • Rothman S. Synaptic release of excitatory amino acid neurotransmitter mediates anoxic neuronal death. J Neurosci. 1984;4:1884–1891.
  • Clark GD, Rothman SM. Blockade of excitatory amino acid receptors protects anoxic hippocampal slices. Neuroscience. 1987;21(3):665–671. .
  • 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(7):857–863. .
  • 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.
  • Girouard H, Wang G, Gallo EF, et al. NMDA receptor activation increases free radical production through nitric oxide and NOX2. J Neurosci. 2009;29(8):2545–2552. .
  • 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(4):e580.
  • 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(4):525–538. .
  • Christopherson KS, Hillier BJ, Lim WA, et al. PSD-95 assembles a ternary complex with the N-methyl-D-aspartic acid receptor and a bivalent neuronal NO synthase PDZ domain. J Biol Chem. 1999;274:27467–27473.
  • Zhou L, Li F, Xu HB, et al. Treatment of cerebral ischemia by disrupting ischemia-induced interaction of nNOS with PSD-95. Nat Med. 2010;16:1439–1443.
  • Schulz JB, Matthews RT, Jenkins BG, et al. Blockade of neuronal nitric oxide synthase protects against excitotoxicity in vivo. J Neurosci. 1995;15:8419–8429.
  • Ayata C, Ayata G, Hara H, et al. Mechanisms of reduced striatal NMDA excitotoxicity in type I nitric oxide synthase knock-out mice. J Neurosci. 1997;17:6908–6917.
  • Gunasekar PG, Kanthasamy AG, Borowitz JL, et al. NMDA receptor activation produces concurrent generation of nitric oxide and reactive oxygen species: implication for cell death. J Neurochem. 1995;65:2016–2021.
  • Ayata C, Ayata G, Hara H, et al. Mechanisms of reduced striatal NMDA excitotoxicity in type I nitric oxide synthase knock-out mice. J Neurosci. 1997;17:6908–6917.
  • Eliasson MJ, Huang Z, Ferrante RJ, et al. Neuronal nitric oxide synthase activation and peroxynitrite formation in ischemic stroke linked to neural damage. J Neurosci. 1999;19:5910–5918.
  • Mandir AS, Poitras MF, Berliner AR, et al. NMDA but not non-NMDA excitotoxicity is mediated by Poly(ADP-ribose) polymerase. J Neurosci. 2000;20:8005–8011.
  • Forder JP, Tymianski M. Postsynaptic mechanisms of excitotoxicity: involvement of postsynaptic density proteins, radicals, and oxidant molecules. Neuroscience. 2009;158:293–300.
  • Al-Gayyar MM, Abdelsaid MA, Matragoon S, et al. Neurovascular protective effect of FeTPPs in N-methyl-D-aspartate model: similarities to diabetes. Am J Pathol. 2010;177:1187–1197.
  • Biegon A, Fry PA, Paden CM, et al. Dynamic changes in N-methyl-D-aspartate receptors after closed head injury in mice: implications for treatment of neurological and cognitive deficits. Proc Natl Acad Sci U S A. 2004;101:5117–5122.
  • Biegon A, Liraz-Zaltsman S, Shohami E. Stimulation of N-methyl-D-aspartate receptors by exogenous and endogenous ligands improves outcome of brain injury. Curr Opin Neurol. 2018;31:687–692.
  • 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.
  • Rama Rao KV, Iring S, Younger D, et al. A single primary blast-induced traumatic brain injury in a rodent model causes cell-type dependent increase in nicotinamide adenine dinucleotide phosphate oxidase isoforms in vulnerable brain regions. J Neurotrauma. 2018;35:2077–2090.
  • Zhang HF, Li TB, Liu B, et al. Inhibition of myosin light chain kinase reduces NADPH oxidase-mediated oxidative injury in rat brain following cerebral ischemia/reperfusion. Naunyn Schmiedebergs Arch Pharmacol. 2015;388:953–963.
  • Zhang YS, Liu B, Luo XJ, et al. Nuclear cardiac myosin light chain 2 modulates NADPH oxidase 2 expression in myocardium: a novel function beyond muscle contraction. Basic Res Cardiol. 2015;110:38. DOI: 10.1007/s00395-015-0494
  • Zhang YS, Tang LJ, Tu H, et al. Fasudil ameliorates the ischemia/reperfusion oxidative injury in rat hearts through suppression of myosin regulatory light chain/NADPH oxidase 2 pathway. Eur J Pharmacol. 2018;822:1–12.
  • Jin L, Ying Z, Webb RC. Activation of Rho/Rho kinase signaling pathway by reactive oxygen species in rat aorta. Am J Physiol Heart Circ Physiol. 2004;287:H1495–H1500.
  • Jin L, Ying Z, Hilgers RH, et al. Increased RhoA/Rho-kinase signaling mediates spontaneous tone in aorta from angiotensin II-induced hypertensive rats. J Pharmacol Exp Ther. 2006;318:288–295.
  • Somlyo AP, Somlyo AV. Signal transduction by G-proteins, rho-kinase and protein phosphatase to smooth muscle and non-muscle myosin II. J Physiol. 2000;522(Pt 2):177–185.
  • Kim HW, Cho KJ, Lee SK, et al. Apoptosis signal-regulating kinase 1 (Ask1) targeted small interfering RNA on ischemic neuronal cell death. Brain Res. 2011;1412:73–78.
  • Barros-Minones L, Orejana L, Goni-Allo B, et al. Modulation of the ASK1-MKK3/6-p38/MAPK signalling pathway mediates sildenafil protection against chemical hypoxia caused by malonate. Br J Pharmacol. 2013;168:1820–1834.
  • An S, Kuang Y, Shen T, et al. Brain-targeting delivery for RNAi neuroprotection against cerebral ischemia reperfusion injury. Biomaterials. 2013;34:8949–8959.
  • Saitoh M, Nishitoh H, Fujii M, et al. Mammalian thioredoxin is a direct inhibitor of apoptosis signal-regulating kinase (ASK) 1. Embo J. 1998;17:2596–2606.
  • Song JJ, Lee YJ. Catalase, but not MnSOD, inhibits glucose deprivation-activated ASK1-MEK-MAPK signal transduction pathway and prevents relocalization of Daxx: hydrogen peroxide as a major second messenger of metabolic oxidative stress. J Cell Biochem. 2003;90:304–314.
  • Kylarova S, Kosek D, Petrvalska O, et al. Cysteine residues mediate high-affinity binding of thioredoxin to ASK1. Febs J. 2016;283:3821–3838.
  • Yin T, Hou R, Liu S, et al. Nitrative inactivation of thioredoxin-1 increases vulnerability of diabetic hearts to ischemia/reperfusion injury. J Mol Cell Cardiol. 2010;49:354–361.
  • Al-Gayyar MM, Abdelsaid MA, Matragoon S, et al. Neurovascular protective effect of FeTPPs in N-methyl-D-aspartate model: similarities to diabetes. Am J Pathol. 2010;177:1187–1197.
  • Liu Y, Qu Y, Wang R, et al. The alternative crosstalk between RAGE and nitrative thioredoxin inactivation during diabetic myocardial ischemia-reperfusion injury. Am J Physiol Endocrinol Metab. 2012;303:E841–E852.
  • Wang K, Zhang J, Wang X, et al. Thioredoxin reductase was nitrated in the aging heart after myocardial ischemia/reperfusion. Rejuvenation Res. 2013;16:377–385.
  • Xu J, Kurup P, Zhang Y, et al. Extrasynaptic NMDA receptors couple preferentially to excitotoxicity via calpain-mediated cleavage of STEP. J Neurosci. 2009;29:9330–9343.
  • Li S, Jin M, Koeglsperger T, et al. Soluble Abeta oligomers inhibit long-term potentiation through a mechanism involving excessive activation of extrasynaptic NR2B-containing NMDA receptors. J Neurosci. 2011;31:6627–6638.
  • Wang Y, Briz V, Chishti A, et al. Distinct roles for mu-calpain and m-calpain in synaptic NMDAR-mediated neuroprotection and extrasynaptic NMDAR-mediated neurodegeneration. J Neurosci. 2013;33:18880–18892.
  • Komjáti K, Besson VC, Poly SC. (adp-ribose) polymerase inhibitors as potential therapeutic agents in stroke and neurotrauma. Curr Drug Targets CNS Neurol Disord. 2005;4:179–194.
  • Haddad M, Rhinn H, Bloquel C, et al. Anti-inflammatory effects of PJ34, a poly(ADP-ribose) polymerase inhibitor, in transient focal cerebral ischemia in mice. Br J Pharmacol. 2006;149:23–30.
  • Moroni F. Poly(ADP-ribose)polymerase 1 (PARP-1) and postischemic brain damage. Curr Opin Pharmacol. 2008;8:96–103.
  • Goto S, Xue R, Sugo N, et al. Poly(ADP-ribose) polymerase impairs early and long-term experimental stroke recovery. Stroke. 2002;33:1101–1106.
  • Whalen MJ, Clark RS, Dixon CE, et al. Reduction of cognitive and motor deficits after traumatic brain injury in mice deficient in poly(ADP-ribose) polymerase. J Cereb Blood Flow Metab. 1999;19:835–842.
  • Stoica BA, Loane DJ, Zhao Z, et al. PARP-1 inhibition attenuates neuronal loss, microglia activation and neurological deficits after traumatic brain injury. J Neurotrauma. 2014;31:758–772.
  • Kim SH, Kim H. Inhibitory Effect of Astaxanthin on Oxidative Stress-Induced Mitochondrial Dysfunction-A Mini-Review. Nutrients. 2018;10:9.
  • Baburina Y, Krestinin R, Odinokova I, et al. Astaxanthin Inhibits Mitochondrial Permeability Transition Pore Opening in Rat Heart Mitochondria. In: Antioxidants (Basel). 2019. p. 8. DOI: 10.3390/antiox8120576
  • Shah ZA, Li RC, Ahmad AS, et al. The flavanol (-)-epicatechin prevents stroke damage through the Nrf2/HO1 pathway. J Cereb Blood Flow Metab. 2010;30:1951–1961.
  • Zhao JJ, Song JQ, Pan SY, et al. Treatment with Isorhamnetin Protects the Brain Against Ischemic Injury in Mice. Neurochem Res. 2016;41:1939–1948.
  • Li CY, Liu XY, Bu H, et al. Prevention of glutamate excitotoxicity in motor neurons by 5,6-dihydrocyclopenta-1,2-dithiole-3-thione: implication to the development of neuroprotective drugs. Cell Mol Life Sci. 2007;64:1861–1869.
  • Liu XY, Li CY, Bu H, et al. The neuroprotective potential of phase II enzyme inducer on motor neuron survival in traumatic spinal cord injury in vitro. Cell Mol Neurobiol. 2008;28:769–779.
  • Son TG, Kawamoto EM, Yu QS, et al. Naphthazarin protects against glutamate-induced neuronal death via activation of the Nrf2/ARE pathway. Biochem Biophys Res Commun. 2013;433:602–606.
  • Assis LC, Straliotto MR, Engel D, et al. beta-Caryophyllene protects the C6 glioma cells against glutamate-induced excitotoxicity through the Nrf2 pathway. Neuroscience. 2014;279:220–231.
  • Uddin MS, Mamun AA, Jakaria M, et al. Emerging promise of sulforaphane-mediated Nrf2 signaling cascade against neurological disorders, Sci Total Environ, 2020;;707:135624.
  • Dash PK, Zhao J, Orsi SA, et al. Sulforaphane improves cognitive function administered following traumatic brain injury. Neurosci Lett. 2009;460:103–107.
  • Xu J, Wang H, Ding K, et al. Luteolin provides neuroprotection in models of traumatic brain injury via the Nrf2-ARE pathway. Free Radic Biol Med. 2014;71:186–195.
  • Zhang L, Wang H. Targeting the NF-E2-related factor 2 pathway: a novel strategy for traumatic brain injury. Mol Neurobiol. 2018;55:1773–1785.
  • Fang J, Wang H, Zhou J, et al. Baicalin provides neuroprotection in traumatic brain injury mice model through Akt/Nrf2 pathway. Drug Des Devel Ther. 2018;12:2497–2508.
  • Zhang L, Wang H, Zhou Y, et al. Fisetin alleviates oxidative stress after traumatic brain injury via the Nrf2-ARE pathway. Neurochem Int. 2018;118:304–313.
  • Liu Y, Zhang L, Liang J. Activation of the Nrf2 defense pathway contributes to neuroprotective effects of phloretin on oxidative stress injury after cerebral ischemia/reperfusion in rats. J Neurol Sci. 2015;351:88–92.
  • Lv C, Maharjan S, Wang Q, et al. Alpha-lipoic acid promotes neurological recovery after ischemic stroke by activating the nrf2/ho-1 pathway to attenuate oxidative damage. Cell Physiol Biochem. 2017;43:1273–1287.
  • Soares MP, Bach FH. Heme oxygenase-1: from biology to therapeutic potential. Trends Mol Med. 2009;15:50–58.
  • 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, Dusting GJ. 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.
  • Basuroy S, Bhattacharya S, Leffler CW, et al. NADPH oxidase mediates oxidative stress and apoptosis caused by TNF-alpha in cerebral vascular endothelial cells. Am J Physiol Cell Physiol. 2009;296:C422–C432.
  • 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.
  • Deguchi K, Hayashi T, Nagotani S, et al. Reduction of cerebral infarction in rats by biliverdin associated with amelioration of oxidative stress. Brain Res. 2008;1188:1–8.
  • Li JJ, Zou ZY, Liu J, et al. Biliverdin administration ameliorates cerebral ischemia reperfusion injury in rats and is associated with proinflammatory factor downregulation. Exp Ther Med. 2017;14:671–679.
  • Kitamura Y, Ishida Y, Takata K, et al. Hyperbilirubinemia protects against focal ischemia in rats. J Neurosci Res. 2003;71:544–550.
  • 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.
  • 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.
  • Pavon-Fuentes N, Marin-Prida J, Llopiz-Arzuaga A, et al. Phycocyanobilin reduces brain injury after endothelin-1- induced focal cerebral ischaemia. Clin Exp Pharmacol Physiol. 2020;47:383-392.
  • Penton-Rol G, Marin-Prida J, C-Phycocyanin F-CV Phycocyanobilin as remyelination therapies for enhancing recovery in multiple sclerosis and ischemic stroke: a preclinical perspective. In: Behav sci (Basel). and; 2018;8:15-31
  • Cervantes-Llanos M, Lagumersindez-Denis N, Marin-Prida J, et al. Beneficial effects of oral administration of C-Phycocyanin and Phycocyanobilin in rodent models of experimental autoimmune encephalomyelitis. Life Sci. 2018;194:130–138.
  • Gammella E, Recalcati S, Cairo G. Dual role of ros as signal and stress agents: iron tips the balance in favor of toxic effects. In Oxid med cell longev. 2016;;2016:8629024.
  • Rodriguez AI, Gangopadhyay A, Kelley EE, et al. HO-1 and CO decrease platelet-derived growth factor-induced vascular smooth muscle cell migration via inhibition of Nox1. Arterioscler Thromb Vasc Biol. 2010;30:98–104.
  • Verma A, Hirsch DJ, Glatt CE, et al. Carbon monoxide: a putative neural messenger. Science. 1993;259:381–384.
  • Christodoulides N, Durante W, Kroll MH, et al. Vascular smooth muscle cell heme oxygenases generate guanylyl cyclase-stimulatory carbon monoxide. Circulation. 1995;91:2306–2309.
  • Ha KS, Kim KM, Kwon YG, et al. Nitric oxide prevents 6-hydroxydopamine-induced apoptosis in PC12 cells through cGMP-dependent PI3 kinase/Akt activation. Faseb J. 2003;17:1036–1047.
  • Mejia-Garcia TA, Portugal CC, Encarnacao TG, et al. Nitric oxide regulates AKT phosphorylation and nuclear translocation in cultured retinal cells. Cell Signal. 2013;25:2424–2439.
  • Culmsee C, Gerling N, Landshamer S, et al. Nitric oxide donors induce neurotrophin-like survival signaling and protect neurons against apoptosis. Mol Pharmacol. 2005;68:1006–1017.
  • Akassoglou K. Nerve growth factor-independent neuronal survival: a role for NO donors. Mol Pharmacol. 2005;68:952–955.
  • Wang L, Gang ZZ, Lan ZR, et al. Activation of the PI3-K/Akt pathway mediates cGMP enhanced-neurogenesis in the adult progenitor cells derived from the subventricular zone. J Cereb Blood Flow Metab. 2005;25:1150–1158.
  • Kim AH, Khursigara G, Sun X, et al. Akt phosphorylates and negatively regulates apoptosis signal-regulating kinase 1. Mol Cell Biol. 2001;21:893–901.
  • Zhang R, Luo D, Miao R, et al. Hsp90-Akt phosphorylates ASK1 and inhibits ASK1-mediated apoptosis. Oncogene. 2005;24:3954–3963.
  • Datta SR, Dudek H, Tao X, et al. Akt phosphorylation of BAD couples survival signals to the cell-intrinsic death machinery. Cell. 1997;91:231–241.
  • Downward J. PI 3-kinase, Akt and cell survival. Semin Cell Dev Biol. 2004;15:177–182.
  • Sauzeau V, Le JH, Cario-Toumaniantz C, et al. Cyclic GMP-dependent protein kinase signaling pathway inhibits RhoA-induced Ca2+ sensitization of contraction in vascular smooth muscle. J Biol Chem. 2000;275:21722–21729.
  • Rolli-Derkinderen M, Sauzeau V, Boyer L, et al. Phosphorylation of serine 188 protects RhoA from ubiquitin/proteasome-mediated degradation in vascular smooth muscle cells. Circ Res. 2005;96(11):1152–1160. .
  • Follmann M, Griebenow N, Hahn MG, et al. The chemistry and biology of soluble guanylate cyclase stimulators and activators. Angew Chem Int Ed Engl. 2013;52(36):9442–9462. .
  • Xiao S, Li Q, Hu L. Soluble Guanylate Cyclase Stimulators and Activators: where are We and Where to Go? Mini Rev Med Chem. 2019;19(18):1544–1557.
  • Vesely DL. Biotin enhances guanylate cyclase activity. Science. 1982;216(4552):1329–1330. .
  • Vilches-Flores A, Tovar AR, Marin-Hernandez A, et al. Biotin increases glucokinase expression via soluble guanylate cyclase/protein kinase G, adenosine triphosphate production and autocrine action of insulin in pancreatic rat islets. J Nutr Biochem. 2010;21(7):606–612. .
  • Watanabe-Kamiyama M, Kamiyama S, Horiuchi K, et al., Antihypertensive effect of biotin in stroke-prone spontaneously hypertensive rats. Br J Nutr. 99(4): 756–763. 2008. .
  • Mock DM. Biotin: from Nutrition to Therapeutics. J Nutr. 2017;147(8):1487–1492.
  • Tourbah A, Lebrun-Frenay C, Edan G, et al. MD1003 (high-dose biotin) for the treatment of progressive multiple sclerosis: A randomised, double-blind, placebo-controlled study. Mult Scler. 2016;22(13):1719–1731. .
  • McCarty MF, DiNicolantonio JJ. Neuroprotective potential of high-dose biotin. Med Hypotheses. 2017;109:145–149.
  • Chen XM, Wang NN, Zhang TY, et al. Neuroprotection by sildenafil: neuronal networks potentiation in acute experimental stroke. CNS Neurosci Ther. 2014;20:40–49.
  • Barros-Minones L, Martin-de-Saavedra D, Perez-Alvarez S, et al. Inhibition of calpain-regulated p35/cdk5 plays a central role in sildenafil-induced protection against chemical hypoxia produced by malonate. Biochim Biophys Acta. 2013;1832(6):705–717. .
  • Barros-Minones L, Orejana L, Goni-Allo B, et al. Modulation of the ASK1-MKK3/6-p38/MAPK signalling pathway mediates sildenafil protection against chemical hypoxia caused by malonate. Br J Pharmacol. 2013;168(8):1820–1834. .
  • Ozdegirmenci O, Kucukozkan T, Akdag E, et al. Effects of sildenafil and tadalafil on ischemia/reperfusion injury in fetal rat brain. J Matern Fetal Neonatal Med. 2011;24(2):317–323. .
  • Garcia-Barroso C, Ricobaraza A, Pascual-Lucas M, et al. Tadalafil crosses the blood-brain barrier and reverses cognitive dysfunction in a mouse model of AD. Neuropharmacology. 2013;64:114–123.
  • Eardley I, Cartledge J. Tadalafil (Cialis) for men with erectile dysfunction. Int J Clin Pract. 2002;56:300–304.
  • Zhou Z, Martin E, Sharina I, et al. Regulation of soluble guanylyl cyclase redox state by hydrogen sulfide. Pharmacol Res. 2016;111:556–562.
  • Szabo C. Hydrogen sulfide, an enhancer of vascular nitric oxide signaling: mechanisms and implications. Am J Physiol Cell Physiol. 2017;312(1):C3–C15.
  • 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.
  • Wolf AM, Asoh S, Hiranuma H, et al. Astaxanthin protects mitochondrial redox state and functional integrity against oxidative stress. J Nutr Biochem. 2010;21(5):381–389. .
  • Liu X, Osawa T. Astaxanthin protects neuronal cells against oxidative damage and is a potent candidate for brain food. Forum Nutr. 2009;61:129–135.
  • Liu X, Shibata T, Hisaka S, et al. Astaxanthin inhibits reactive oxygen species-mediated cellular toxicity in dopaminergic SH-SY5Y cells via mitochondria-targeted protective mechanism. Brain Res. 2009;1254:18–27.
  • Nakajima Y, Inokuchi Y, Shimazawa M, et al. Astaxanthin, a dietary carotenoid, protects retinal cells against oxidative stress in-vitro and in mice in-vivo. J Pharm Pharmacol. 2008;60(10):1365–1374. .
  • Zhang M, Cui Z, Cui H, et al. Astaxanthin alleviates cerebral edema by modulating NKCC1 and AQP4 expression after traumatic brain injury in mice. BMC Neurosci. 2016;17:60.
  • Ji X, Peng D, Zhang Y, et al. Astaxanthin improves cognitive performance in mice following mild traumatic brain injury. Brain Res. 2017;1659:88–95.
  • Xue Y, Qu Z, Fu J, et al. The protective effect of astaxanthin on learning and memory deficits and oxidative stress in a mouse model of repeated cerebral ischemia/reperfusion. Brain Res Bull. 2017;131:221–228.
  • Cui H, Hayashi A, Sun HS, et al. PDZ protein interactions underlying NMDA receptor-mediated excitotoxicity and neuroprotection by PSD-95 inhibitors. J Neurosci. 2007;27:9901–9915.
  • Mo SF, Liao GY, Yang J, et al. Protection of neuronal cells from excitotoxicity by disrupting nNOS-PSD95 interaction with a small molecule SCR-4026. Brain Res. 2016;1648:(Pt A):250–256.
  • Tang W, Sun X, Fang JS, et al. Flavonoids from Radix Scutellariae as potential stroke therapeutic agents by targeting the second postsynaptic density 95 (PSD-95)/disc large/zonula occludens-1 (PDZ) domain of PSD-95. Phytomedicine. 2004;11:277–284.
  • Liu LY, Wei EQ, Zhao YM, et al. Protective effects of baicalin on oxygen/glucose deprivation- and NMDA-induced injuries in rat hippocampal slices. J Pharm Pharmacol. 2005;57:1019–1026.
  • Yang J, Wu X, Yu H, et al. NMDA receptor-mediated neuroprotective effect of the Scutellaria baicalensis Georgi extract on the excitotoxic neuronal cell death in primary rat cortical cell cultures. ScientificWorldJournal. 2014;2014:459549.
  • Li N, Feng L, Tan Y, et al. 23. Characterization, Pharmacokinetics and Biodistribution of Baicalin-Loaded Liposome on Cerebral Ischemia-Reperfusion after i.v. Administration in Rats. Molecules; 2018.
  • Chen H, Guan B, Chen X, et al. Baicalin Attenuates Blood-Brain Barrier Disruption and Hemorrhagic Transformation and Improves Neurological Outcome in Ischemic Stroke Rats with Delayed t-PA Treatment: involvement of ONOO(-)-MMP-9 Pathway. Transl Stroke Res. 2018;9:515–529.
  • Xu M, Chen X, Gu Y, et al. Baicalin can scavenge peroxynitrite and ameliorate endogenous peroxynitrite-mediated neurotoxicity in cerebral ischemia-reperfusion injury. J Ethnopharmacol. 2013;150(1):116–124. .
  • Hlatky R, Lui H, Cherian L, et al. The role of endothelial nitric oxide synthase in the cerebral hemodynamics after controlled cortical impact injury in mice. J Neurotrauma. 2003;20(10):995–1006. .
  • Ahn MJ, Sherwood ER, Prough DS, et al. The effects of traumatic brain injury on cerebral blood flow and brain tissue nitric oxide levels and cytokine expression. J Neurotrauma. 2004;21:1431–1442.
  • Garry PS, Ezra M, Rowland MJ, et al. The role of the nitric oxide pathway in brain injury and its treatment–from bench to bedside. Exp Neurol. 2015;263:235–243.
  • Villalba N, Sackheim AM, Nunez IA, et al. Traumatic brain injury causes endothelial dysfunction in the systemic microcirculation through arginase-1-dependent uncoupling of endothelial nitric oxide synthase. J Neurotrauma. 2017;34:192–203.
  • Cherian L, Robertson CS. L-arginine and free radical scavengers increase cerebral blood flow and brain tissue nitric oxide concentrations after controlled cortical impact injury in rats. J Neurotrauma. 2003;20(1):77–85.
  • Cherian L, Hlatky R, Robertson CS. Comparison of tetrahydrobiopterin and L-arginine on cerebral blood flow after controlled cortical impact injury in rats. J Neurotrauma. 2004;21(9):1196–1203. .
  • Cherian L, Chacko G, Goodman C, et al. Neuroprotective effects of L-arginine administration after cortical impact injury in rats: dose response and time window. J Pharmacol Exp Ther. 2003;304:617–623.
  • Schwedhelm E, Maas R, Freese R, et al. Pharmacokinetic and pharmacodynamic properties of oral L-citrulline and L-arginine: impact on nitric oxide metabolism. Br J Clin Pharmacol. 2008;65:51–59.
  • Terpolilli NA, Kim SW, Thal SC, et al. Inhaled nitric oxide reduces secondary brain damage after traumatic brain injury in mice. J Cereb Blood Flow Metab. 2013;33:311–318.
  • Morikawa E, Moskowitz MA, Huang Z, et al. L-arginine infusion promotes nitric oxide-dependent vasodilation, increases regional cerebral blood flow, and reduces infarction volume in the rat. Stroke. 1994;25:429–435.
  • Terpolilli NA, Kim SW, Thal SC, et al. Inhalation of nitric oxide prevents ischemic brain damage in experimental stroke by selective dilatation of collateral arterioles. Circ Res. 2012;110:727–738.
  • Yu L, Zhang Y, Ma R, et al. Potent protection of ferulic acid against excitotoxic effects of maternal intragastric administration of monosodium glutamate at a late stage of pregnancy on developing mouse fetal brain. Eur Neuropsychopharmacol. 2006;16(3):170–177. .
  • Koh PO. Ferulic acid prevents the cerebral ischemic injury-induced decrease of Akt and Bad phosphorylation. Neurosci Lett. 2012;507:156–160.
  • Sung JH, Cho EH, Cho JH, et al. Identification of proteins regulated by ferulic acid in a middle cerebral artery occlusion animal model-a proteomics approach. J Vet Med Sci. 2012;74:1401–1407.
  • Koh PO. Ferulic acid modulates nitric oxide synthase expression in focal cerebral ischemia. Lab Anim Res. 2012;28:273–278.
  • Koh PO. Ferulic acid attenuates focal cerebral ischemia-induced decreases in p70S6 kinase and S6 phosphorylation. Neurosci Lett. 2013;555:7–11.
  • Koh PO. Ferulic acid attenuates the down-regulation of MEK/ERK/p90RSK signaling pathway in focal cerebral ischemic injury. Neurosci Lett. 2015;588:18–23.
  • 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;13:419–427.
  • 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.
  • Wang Q, Chen SY, Xiong LZ, et al. Neuroprotective effect of sodium ferulate on transient focal cerebral ischemia by weakening activation of postsynaptic density-95 in rats. Chin J Traumatol. 2005;8:297–302.
  • Cheng CY, Ho TY, Lee EJ, et al. Ferulic acid reduces cerebral infarct through its antioxidative and anti-inflammatory effects following transient focal cerebral ischemia in rats. Am J Chin Med. 2008;36:1105–1119.
  • Cheng CY, Su SY, Tang NY, et al. Ferulic acid inhibits nitric oxide-induced apoptosis by enhancing GABA(B1) receptor expression in transient focal cerebral ischemia in rats. Acta Pharmacol Sin. 2010;31:889–899.
  • Wang BH, Ou-Yang JP. Pharmacological actions of sodium ferulate in cardiovascular system. Cardiovasc Drug Rev. 2005;23:161–172.
  • 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.
  • Parastan RH, Christopher M, Torrys YS, et al. Combined therapy potential of apocynin and tert-butylhydroquinone as a therapeutic agent to prevent secondary progression to traumatic brain injury. Asian J Neurosurg. 2020;15:10–15.
  • Chandran R, Kim T, Mehta SL, et al. A combination antioxidant therapy to inhibit NOX2 and activate Nrf2 decreases secondary brain damage and improves functional recovery after traumatic brain injury. J Cereb Blood Flow Metab. 2018;38:1818–1827.
  • 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.
  • 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.
  • 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.
  • 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;14:143-154.
  • 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;15:116-130.
  • 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:(Pt A):26–31.
  • 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.
  • 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.

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.