Antioxidant Therapies in TBI: Comparison
Please note this is a comparison between Version 2 by Vivi Li and Version 1 by Giacomo Lazzarino.

Due to a multiplicity of causes provoking traumatic brain injury (TBI), TBI is a highly heterogeneous pathology, characterized by high mortality and disability rates. TBI is an acute neurodegenerative event, potentially and unpredictably evolving into sub-chronic and chronic neurodegenerative events, with transient or permanent neurologic, cognitive, and motor deficits, for which no valid standardized therapies are available. A vast body of literature demonstrates that TBI-induced oxidative/nitrosative stress is involved in the development of both acute and chronic neurodegenerative disorders. Cellular defenses against this phenomenon are largely dependent on low molecular weight antioxidants, most of which are consumed with diet or as nutraceutical supplements. A large number of studies have evaluated the efficacy of antioxidant administration to decrease TBI-associated damage in various animal TBI models and in a limited number of clinical trials. Points of weakness of preclinical studies are represented by the large variability in the TBI model adopted, in the antioxidant tested, in the timing, dosages, and routes of administration used, and in the variety of molecular and/or neurocognitive parameters evaluated. The analysis of the very few clinical studies does not allow strong conclusions to be drawn on the real effectiveness of antioxidant administration to TBI patients. Standardizing TBI models and different experimental conditions, as well as testing the efficacy of administration of a cocktail of antioxidants rather than only one, should be mandatory. According to some promising clinical results, it appears that sports-related concussion is probably the best type of TBI to test benefits of antioxidant administration.

  • traumatic brain injury
  • oxidative/nitrosative stress
  • low molecular weight antioxidants
  • concussion
Please wait, diff process is still running!

References

  1. Maas, A.I.R.; Menon, D.K.; Adelson, P.D.; Andelic, N.; Bell, M.J.; Belli, A.; Bragge, P.; Brazinova, A.; Buki, A.; Chesnut, R.M.; et al. InTBIR Participants and Investigators. Traumatic brain injury: Integrated approaches to improve prevention, clinical care, and research. Lancet Neurol. 2017, 16, 987–1048.
  2. World Health Oraganization. Neurological Disorders: Public Health Challenges. 2006. Available online: (accessed on 19 July 2019).
  3. Teasdale, G.; Jennett, B. Assessment of coma and impaired consciousness. A practical scale. Lancet 1974, 2, 81–84.
  4. Carroll, L.J.; Cassidy, J.D.; Holm, L.; Kraus, J.; Coronado, V.G. WHO Collaborating Centre Task Force on Mild Traumatic Brain Injury. Methodological issues and research recommendations for mild traumatic brain injury: The WHO Collaborating Centre Task Force on Mild Traumatic Brain Injury. J. Rehabil. Med. 2004, 43, 113–125.
  5. Maas, A.I.; Stocchetti, N.; Bullock, R. Moderate and severe traumatic brain injury in adults. Lancet Neurol. 2008, 7, 728–741.
  6. Kochanek, P.M.; Dixon, C.E.; Mondello, S.; Wang, K.K.K.; Lafrenaye, A.; Bramlett, H.M.; Dietrich, W.D.; Hayes, R.L.; Shear, D.A.; Gilsdorf, J.S.; et al. Multi-Center Pre-clinical Consortia to Enhance Translation of Therapies and Biomarkers for Traumatic Brain Injury: Operation Brain Trauma Therapy and Beyond. Front. Neurol. 2018, 9, 640.
  7. Galgano, M.; Toshkezi, G.; Qiu, X.; Russell, T.; Chin, L.; Zhao, L.R. Traumatic Brain Injury: Current Treatment Strategies and Future Endeavors. Cell Transplant. 2017, 26, 1118–1130.
  8. Carney, N.; Totten, A.M.; O’Reilly, C.; Ullman, J.S.; Hawryluk, G.W.; Bell, M.J.; Bratton, S.L.; Chesnut, R.; Harris, O.A.; Kissoon, N.; et al. Guidelines for the Management of Severe Traumatic Brain Injury, Fourth Edition. Neurosurgery 2017, 80, 6–15.
  9. Kolias, A.G.; Guilfoyle, M.R.; Helmy, A.; Allanson, J.; Hutchinson, P.J. Traumatic brain injury in adults. Pract. Neurol. 2013, 13, 228–235.
  10. Kim, S.; Mortera, M.; Hu, X.; Krishnan, S.; Hoffecker, L.; Herrold, A.; Terhorst, L.; King, L.; Machtinger, J.; Zumsteg, J.M.; et al. Overview of pharmacological interventions after traumatic brain injuries: Impact on selected outcomes. Brain Inj. 2019, 33, 442–455.
  11. Kumar Sahel, D.; Kaira, M.; Raj, K.; Sharma, S.; Singh, S. Mitochondrial dysfunctioning and neuroinflammation: Recent highlights on the possible mechanisms involved in Traumatic Brain Injury. Neurosci. Lett. 2019, 710, 134347.
  12. Bullock, R.; Fujisawa, H. The role of glutamate antagonists for the treatment of CNS injury. J. Neurotrauma 1992, 9 (Suppl. 2), S443–S462.
  13. Tymianski, M.; Tator, C.H. Normal and abnormal calcium homeostasis in neurons: A basis for the pathophysiology of traumatic and ischemic central nervous system injury. Neurosurgery 1996, 38, 1176–1195.
  14. Hall, E.D.; Vaishnav, R.A.; Mustafa, A.G. Antioxidant therapies for traumatic brain injury. Neurotherapeutics 2010, 7, 51–61.
  15. Amorini, A.M.; Lazzarino, G.; Di Pietro, V.; Signoretti, S.; Lazzarino, G.; Belli, A.; Tavazzi, B. Severity of experimental traumatic brain injury modulates changes in concentrations of cerebral free amino acids. J. Cell. Mol. Med. 2017, 21, 530–542.
  16. Arundine, M.; Tymianski, M. Molecular mechanisms of glutamate-dependent neurodegeneration in ischemia and traumatic brain injury. Cell. Mol. Life Sci. 2004, 61, 657–668.
  17. Tavazzi, B.; Signoretti, S.; Lazzarino, G.; Amorini, A.M.; Delfini, R.; Cimatti, M.; Marmarou, A.; Vagnozzi, R. Cerebral oxidative stress and depression of energy metabolism correlate with severity of diffuse brain injury in rats. Neurosurgery 2005, 56, 582–589.
  18. Cristofori, L.; Tavazzi, B.; Gambin, R.; Vagnozzi, R.; Signoretti, S.; Amorini, A.M.; Fazzina, G.; Lazzarino, G. Biochemical analysis of the cerebrospinal fluid: Evidence for catastrophic energy failure and oxidative damage preceding brain death in severe head injury: A case report. Clin. Biochem. 2005, 38, 97–100.
  19. Hall, E.D.; Springer, J.E. Neuroprotection and acute spinal cord injury: A reappraisal. NeuroRx 2004, 1, 80–100.
  20. Ozsuer, H.; Gorgulu, A.; Kiris, T.; Cobanoglu, S. The effects of memantine on lipid peroxidation following closed-head trauma in rats. Neurosurg. Rev. 2005, 28, 143–147.
  21. Zhu, Y.; Wang, H.; Fang, J.; Dai, W.; Zhou, J.; Wang, X.; Zhou, M. SS-31 Provides Neuroprotection by Reversing Mitochondrial Dysfunction After Traumatic Brain Injury. Oxid. Med. Cell. Longev. 2018, 2018, 4783602.
  22. Kang, D.; Narabayashi, H.; Sata, T.; Takeshige, K. Kinetics of superoxide formation by respiratory chain NADH- dehydrogenase of bovine heart mitochondria. J. Biochem. 1983, 94, 1301–1306.
  23. De Campos, R.P.; Siegel, J.M.; Fresta, C.G.; Caruso, G.; da Silva, J.A.; Lunte, S.M. Indirect detection of superoxide in RAW 264.7 macrophage cells using microchip electrophoresis coupled to laser-induced fluorescence. Anal. Bioanal. Chem. 2015, 407, 7003–7012.
  24. Fresta, C.G.; Hogard, M.L.; Caruso, G.; Melo Costa, E.E.; Lazzarino, G.; Lunte, S.M. Monitoring carnosine uptake by RAW 264.7 macrophage cells using microchip electrophoresis with fluorescence detection. Anal. Methods 2017, 9, 402–408.
  25. Mustafa, A.G.; Alshboul, O.A. Pathophysiology of traumatic brain injury. Neurosciences 2013, 18, 222–234.
  26. Gutteridge, J.M. Lipid peroxidation and antioxidants as biomarkers of tissue damage. Clin. Chem. 1995, 41, 1819–1828.
  27. Cobbs, C.S.; Fenoy, A.; Bredt, D.S.; Noble, L.J. Expression of nitric oxide synthase in the cerebral microvasculature after traumatic brain injury in the rat. Brain Res. 1997, 751, 336–338.
  28. Rao, V.L.; Dogan, A.; Bowen, K.K.; Dempsey, R.J. Traumatic injury to rat brain upregulates neuronal nitric oxide synthase expression and L-[3H]nitroarginine binding. J. Neurotrauma 1999, 16, 865–877.
  29. Gahm, C.; Holmin, S.; Mathiesen, T. Temporal profiles and cellular sources of three nitric oxide synthase isoforms in the brain after experimental contusion. Neurosurgery 2000, 46, 169–177.
  30. Cherian, L.; Goodman, J.C.; Robertson, C.S. Brain nitric oxide changes after controlled cortical impact injury in rats. J. Neurophysiol. 2000, 83, 2171–2178.
  31. Hlatky, R.; Lui, H.; Cherian, L.; Goodman, J.C.; O’Brien, W.E.; Contant, C.F.; Robertson, C.S. The role of endothelial nitric oxide synthase in the cerebral hemodynamics after controlled cortical impact injury in mice. J. Neurotrauma 2003, 20, 995–1006.
  32. Fresta, C.G.; Chakraborty, A.; Wijesinghe, M.B.; Amorini, A.M.; Lazzarino, G.; Lazzarino, G.; Tavazzi, B.; Lunte, S.M.; Caraci, F.; Dhar, P.; et al. Non-toxic engineered carbon nanodiamond concentrations induce oxidative/nitrosative stress, imbalance of energy metabolism, and mitochondrial dysfunction in microglial and alveolar basal epithelial cells. Cell Death Dis. 2018, 9, 245.
  33. Caruso, G.; Fresta, C.G.; Siegel, J.M.; Wijesinghe, M.B.; Lunte, S.M. Microchip electrophoresis with laser-induced fluorescence detection for the determination of the ratio of nitric oxide to superoxide production in macrophages during inflammation. Anal. Bioanal. Chem. 2017, 409, 4529–4538.
  34. Bains, M.; Hall, E.D. Antioxidant therapies in traumatic brain and spinal cord injury. Biochim. Biophys. Acta 2012, 1822, 675–684.
  35. Hall, E.D.; Andrus, P.K.; Oostveen, J.A.; Fleck, T.J.; Gurney, M.E. Relationship of oxygen radical-induced lipid peroxidative damage to disease onset and progression in a transgenic model of familial ALS. J. Neurosci. Res. 1998, 53, 66–77.
  36. Castilho, R.F.; Kowaltowski, A.J.; Meinicke, A.R.; Bechara, E.J.; Vercesi, A.E. Permeabilization of the inner mitochondrial membrane by Ca2+ ions is stimulated by t-butyl hydroperoxide and mediated by reactive oxygen species generated by mitochondria. Free Radic. Biol. Med. 1995, 18, 479–486.
  37. Jacquard, C.; Trioulier, Y.; Cosker, F.; Escartin, C.; Bizat, N.; Hantraye, P.; Cancela, J.M.; Bonvento, G.; Brouillet, E. Brain mitochondrial defects amplify intracellular [Ca2+] rise and neurodegeneration but not Ca2+ entry during NMDA receptor activation. FASEB J. 2006, 20, 1021–1023.
  38. Khan, M.; Dhammu, T.S.; Matsuda, F.; Annamalai, B.; Dhindsa, T.S.; Singh, I.; Singh, A.K. 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.
  39. Nakamura, T.; Lipton, S.A. ‘SNO’-Storms Compromise Protein Activity and Mitochondrial Metabolism in Neurodegenerative Disorders. Trends Endocrinol. Metab. 2017, 28, 879–892.
  40. Lazzarino, G.; Amorini, A.M.; Petzold, A.; Gasperini, C.; Ruggieri, S.; Quartuccio, M.E.; Lazzarino, G.; Di Stasio, E.; Tavazzi, B. Serum Compounds of Energy Metabolism Impairment Are Related to Disability, Disease Course and Neuroimaging in Multiple Sclerosis. Mol. Neurobiol. 2017, 54, 7520–7533.
  41. Aquilano, K.; Baldelli, S.; Rotilio, G.; Ciriolo, M.R. Role of nitric oxide synthases in Parkinson’s disease: A review on the antioxidant and anti-inflammatory activity of polyphenols. Neurochem. Res. 2008, 33, 2416–2426.
  42. Lazzarino, G.; Listorti, I.; Muzii, L.; Amorini, A.M.; Longo, S.; Di Stasio, E.; Caruso, G.; D’Urso, S.; Puglia, I.; Pisani, G.; et al. Low-molecular weight compounds in human seminal plasma as potential biomarkers of male infertility. Hum. Reprod. 2018, 33, 1817–1828.
  43. Lazzarino, G.; Listorti, I.; Bilotta, G.; Capozzolo, T.; Amorini, A.M.; Longo, S.; Caruso, G.; Lazzarino, G.; Tavazzi, B.; Bilotta, P. Water- and Fat-Soluble Antioxidants in Human Seminal Plasma and Serum of Fertile Males. Antioxidants 2019, 8, 96.
  44. Perez-Gregorio, R.; Simal-Gandara, J. A Critical Review of Bioactive Food Components, and of their Functional Mechanisms, Biological Effects and Health Outcomes. Curr. Pharm. Des. 2017, 23, 2731–2741.
  45. Hasadsri, L.; Wang, B.H.; Lee, J.V.; Erdman, J.W.; Llano, D.A.; Barbey, A.K.; Wszalek, T.; Sharrock, M.F.; Wang, H.J. Omega-3 fatty acids as a putative treatment for traumatic brain injury. J. Neurotrauma 2013, 30, 897–906.
  46. Rigg, J.L.; Elovic, E.P.; Greenwald, B.D. A review of the effectiveness of antioxidant therapy to reduce neuronal damage in acute traumatic brain injury. J. Head Trauma Rehabil. 2005, 20, 389–391.
  47. Vonder Haar, C.; Peterson, T.C.; Martens, K.M.; Hoane, M.R. Vitamins and nutrients as primary treatments in experimental brain injury: Clinical implications for nutraceutical therapies. Brain Res. 2016, 1640, 114–129.
  48. Granger, M.; Eck, P. Dietary Vitamin C in Human Health. Adv. Food Nutr. Res. 2018, 83, 281–310.
  49. Castiglione, D.; Platania, A.; Conti, A.; Falla, M.; D’Urso, M.; Marranzano, M. Dietary Micronutrient and Mineral Intake in the Mediterranean Healthy Eating, Ageing, and Lifestyle (MEAL) Study. Antioxidants 2018, 7, 79.
  50. Burzle, M.; Suzuki, Y.; Ackermann, D.; Miyazaki, H.; Maeda, N.; Clemencon, B.; Burrier, R.; Hediger, M.A. The sodium-dependent ascorbic acid transporter family SLC23. Mol. Asp. Med. 2013, 34, 436–454.
  51. Savini, I.; Rossi, A.; Pierro, C.; Avigliano, L.; Catani, M.V. SVCT1 and SVCT2: Key proteins for vitamin C uptake. Amino Acids 2008, 34, 347–355.
  52. Di Pietro, V.; Lazzarino, G.; Amorini, A.M.; Tavazzi, B.; D’Urso, S.; Longo, S.; Vagnozzi, R.; Signoretti, S.; Clementi, E.; Giardina, B.; et al. Neuroglobin expression and oxidant/antioxidant balance after graded traumatic brain injury in the rat. Free Radic. Biol. Med. 2014, 69, 258–264.
  53. Grunewald, R.A. Ascorbic acid in the brain. Brain Res. Rev. 1993, 18, 123–133.
  54. Rice, M.E. Ascorbate regulation and its neuroprotective role in the brain. Trends Neurosci. 2000, 23, 209–216.
  55. Parker, W.H.; Qu, Z.C.; May, J.M. Ascorbic acid transport in brain microvascular pericytes. Biochem. Biophys. Res. Commun. 2015, 458, 262–267.
  56. Angulo, C.; Castro, M.A.; Rivas, C.I.; Segretain, D.; Maldonado, R.; Yanez, A.J.; Slebe, J.C.; Vera, J.C.; Concha, I.I. Molecular identification and functional characterization of the vitamin C transporters expressed by Sertoli cells. J. Cell Physiol. 2008, 217, 708–716.
  57. Awasthi, D.; Church, D.F.; Torbati, D.; Carey, M.E.; Pryor, W.A. Oxidative stress following traumatic brain injury in rats. Surg. Neurol. 1997, 47, 575–581.
  58. Tyurin, V.A.; Tyurina, Y.Y.; Borisenko, G.G.; Sokolova, T.V.; Ritov, V.B.; Quinn, P.J.; Rose, M.; Kochanek, P.; Graham, S.H.; Kagan, V.E. Oxidative stress following traumatic brain injury in rats: Quantitation of biomarkers and detection of free radical intermediates. J. Neurochem. 2000, 75, 2178–2189.
  59. Ishaq, G.M.; Saidu, Y.; Bilbis, L.S.; Muhammad, S.A.; Jinjir, N.; Shehu, B.B. Effects of alpha-tocopherol and ascorbic acid in the severity and management of traumatic brain injury in albino rats. J. Neurosci. Rural Pract. 2013, 4, 292–297.
  60. Razmkon, A.; Sadidi, A.; Sherafat-Kazemzadeh, E.; Mehrafshan, A.; Jamali, M.; Malekpour, B.; Saghafinia, M. Administration of vitamin C and vitamin E in severe head injury: A randomized double-blind controlled trial. Clin. Neurosurg. 2011, 58, 133–137.
  61. Lv, H.; Zhen, C.; Liu, J.; Yang, P.; Hu, L.; Shang, P. Unraveling the Potential Role of Glutathione in Multiple Forms of Cell Death in Cancer Therapy. Oxid. Med. Cell. Longev. 2019, 2019, 3150145.
  62. Limongi, D.; Baldelli, S.; Checconi, P.; Marcocci, M.E.; De Chiara, G.; Fraternale, A.; Magnani, M.; Ciriolo, M.R.; Palamara, A.T. GSH-C4 Acts as Anti-inflammatory Drug in Different Models of Canonical and Cell Autonomous Inflammation Through NFκB Inhibition. Front. Immunol. 2019, 10, 155.
  63. Steullet, P.; Neijt, H.C.; Cuenod, M.; Do, K.Q. Synaptic plasticity impairment and hypofunction of NMDA receptors induced by glutathione deficit: Relevance to schizophrenia. Neuroscience 2006, 137, 807–819.
  64. Varga, V.; Jenei, Z.; Janaky, R.; Saransaari, P.; Oja, S.S. Glutathione is an endogenous ligand of rat brain N-methyl-D-aspartate (NMDA) and 2-amino-3-hydroxy-5-methyl-4-isoxazolepropionate (AMPA) receptors. Neurochem. Res. 1997, 22, 1165–1171.
  65. Oja, S.S.; Janaky, R.; Varga, V.; Saransaari, P. Modulation of glutamate receptor functions by glutathione. Neurochem. Int. 2000, 37, 299–306.
  66. Barnett, S.D.; Buxton, I.L.O. The role of S-nitrosoglutathione reductase (GSNOR) in human disease and therapy. Crit. Rev. Biochem. Mol. Biol. 2017, 52, 340–354.
  67. Ansari, M.A.; Roberts, K.N.; Scheff, S.W. Oxidative stress and modification of synaptic proteins in hippocampus after traumatic brain injury. Free Radic. Biol. Med. 2008, 45, 443–452.
  68. Bayir, H.; Kagan, V.E.; Tyurina, Y.Y.; Tyurin, V.; Ruppel, R.A.; Adelson, P.D.; Graham, S.H.; Janesko, K.; Clark, R.S.; Kochanek, P.M. Assessment of antioxidant reserves and oxidative stress in cerebrospinal fluid after severe traumatic brain injury in infants and children. Pediatr. Res. 2002, 51, 571–578.
  69. Dash, P.K.; Hergenroeder, G.W.; Jeter, C.B.; Choi, H.A.; Kobori, N.; Moore, A.N. Traumatic Brain Injury Alters Methionine Metabolism: Implications for Pathophysiology. Front. Syst. Neurosci. 2016, 10, 36.
  70. Koza, L.; Linseman, D.A. Glutathione precursors shield the brain from trauma. Neural Regen. Res. 2019, 14, 1701–1702.
  71. Reed, T.T.; Owen, J.; Pierce, W.M.; Sebastian, A.; Sullivan, P.G.; Butterfield, D.A. Proteomic identification of nitrated brain proteins in traumatic brain-injured rats treated postinjury with gamma-glutamylcysteine ethyl ester: Insights into the role of elevation of glutathione as a potential therapeutic strategy for traumatic brain injury. J. Neurosci. Res. 2009, 87, 408–417.
  72. Hicdonmez, T.; Kanter, M.; Tiryaki, M.; Parsak, T.; Cobanoglu, S. Neuroprotective effects of N-acetylcysteine on experimental closed head trauma in rats. Neurochem. Res. 2006, 31, 473–481.
  73. Sangobowale, M.; Nikulina, E.; Bergold, P.J. Minocycline plus N-acetylcysteine protect oligodendrocytes when first dosed 12 h after closed head injury in mice. Neurosci. Lett. 2018, 682, 16–20.
  74. Senol, N.; Naziroglu, M.; Yuruker, V. N-acetylcysteine and selenium modulate oxidative stress, antioxidant vitamin and cytokine values in traumatic brain injury-induced rats. Neurochem. Res. 2014, 39, 685–692.
  75. Santos-Buelga, C.; Feliciano, A.S. Flavonoids: From Structure to Health Issues. Molecules 2017, 22, 477.
  76. Izzi, V.; Masuelli, L.; Tresoldi, I.; Sacchetti, P.; Modesti, A.; Galvano, F.; Bei, R. The effects of dietary flavonoids on the regulation of redox inflammatory networks. Front. Biosci. (Landmark Ed.) 2012, 17, 2396–2418.
  77. Theadom, A.; Mahon, S.; Barker-Collo, S.; McPherson, K.; Rush, E.; Vandal, A.C.; Feigin, V.L. Enzogenol for cognitive functioning in traumatic brain injury: A pilot placebo-controlled RCT. Eur. J. Neurol. 2013, 20, 1135–1144.
  78. Yang, X.; Li, X.; Ren, J. From French Paradox to cancer treatment: Anti-cancer activities and mechanisms of resveratrol. Anticancer Agents Med. Chem. 2014, 14, 806–825.
  79. Tsai, H.Y.; Ho, C.T.; Chen, Y.K. Biological actions and molecular effects of resveratrol, pterostilbene, and 3′-hydroxypterostilbene. J. Food Drug Anal. 2017, 25, 134–147.
  80. Ates, O.; Cayli, S.; Altinoz, E.; Gurses, I.; Yucel, N.; Sener, M.; Kocak, A.; Yologlu, S. Neuroprotection by resveratrol against traumatic brain injury in rats. Mol. Cell. Biochem. 2007, 294, 137–144.
  81. Lin, C.J.; Chen, T.H.; Yang, L.Y.; Shih, C.M. Resveratrol protects astrocytes against traumatic brain injury through inhibiting apoptotic and autophagic cell death. Cell Death Dis. 2014, 5, e1147.
  82. Gatson, J.W.; Liu, M.M.; Abdelfattah, K.; Wigginton, J.G.; Smith, S.; Wolf, S.; Minei, J.P. Resveratrol decreases inflammation in the brain of mice with mild traumatic brain injury. J. Trauma Acute Care Surg. 2013, 74, 470–474.
  83. Irias-Mata, A.; Stuetz, W.; Sus, N.; Hammann, S.; Gralla, K.; Cordero-Solano, A.; Vetter, W.; Frank, J. Tocopherols, Tocomonoenols, and Tocotrienols in Oils of Costa Rican Palm Fruits: A Comparison between Six Varieties and Chemical versus Mechanical Extraction. J. Agric. Food Chem. 2017, 65, 7476–7482.
  84. Comitato, R.; Ambra, R.; Virgili, F. Tocotrienols: A Family of Molecules with Specific Biological Activities. Antioxidants 2017, 6, 93.
  85. Inci, S.; Ozcan, O.E.; Kilinc, K. Time-level relationship for lipid peroxidation and the protective effect of alpha-tocopherol in experimental mild and severe brain injury. Neurosurgery 1998, 43, 330–335.
  86. Yang, J.; Han, Y.; Ye, W.; Liu, F.; Zhuang, K.; Wu, G. Alpha tocopherol treatment reduces the expression of Nogo-A and NgR in rat brain after traumatic brain injury. J. Surg. Res. 2013, 182, e69–e77.
  87. Clifton, G.L.; Lyeth, B.G.; Jenkins, L.W.; Taft, W.C.; DeLorenzo, R.J.; Hayes, R.L. Effect of D, alpha-tocopheryl succinate and polyethylene glycol on performance tests after fluid percussion brain injury. J. Neurotrauma 1989, 6, 71–81.
  88. Aiguo, W.; Zhe, Y.; Gomez-Pinilla, F. Vitamin E protects against oxidative damage and learning disability after mild traumatic brain injury in rats. Neurorehabil. Neural Repair 2010, 24, 290–298.
  89. Artuch, R.; Salviati, L.; Jackson, S.; Hirano, M.; Navas, P. Coenzyme Q10 deficiencies in neuromuscular diseases. Adv. Exp. Med. Biol. 2009, 652, 117–128.
  90. Jorat, M.V.; Tabrizi, R.; Kolahdooz, F.; Akbari, M.; Salami, M.; Heydari, S.T.; Asemi, Z. The effects of coenzyme Q10 supplementation on biomarkers of inflammation and oxidative stress in among coronary artery disease: A systematic review and meta-analysis of randomized controlled trials. Inflammopharmacology 2019, 27, 233–248.
  91. Wang, Y.; Hekimi, S. Understanding Ubiquinone. Trends Cell Biol. 2016, 26, 367–378.
  92. Kalayci, M.; Unal, M.M.; Gul, S.; Acikgoz, S.; Kandemir, N.; Hanci, V.; Edebali, N.; Acikgoz, B. Effect of coenzyme Q10 on ischemia and neuronal damage in an experimental traumatic brain-injury model in rats. BMC Neurosci. 2011, 12, 75.
  93. Santoro, M.M. The Antioxidant Role of Non-mitochondrial CoQ10: Mystery Solved! Cell Metab. 2020, 31, 13–15.
  94. Pierce, J.D.; Shen, Q.; Peltzer, J.; Thimmesch, A.; Hiebert, J.B. A pilot study exploring the effects of ubiquinol on brain genomics after traumatic brain injury. Nurs. Outlook 2017, 65, S44–S52.
  95. Pierce, J.D.; Gupte, R.; Thimmesch, A.; Shen, Q.; Hiebert, J.B.; Brooks, W.M.; Clancy, R.L.; Diaz, F.J.; Harris, J.L. Ubiquinol treatment for TBI in male rats: Effects on mitochondrial integrity, injury severity, and neurometabolism. J. Neurosci. Res. 2018, 96, 1080–1092.
  96. Ross, A.C.; Caballero, B.H.; Cousins, R.J.; Tucker, K.L.; Ziegler, T.R. Modern Nutrition in Health and Disease, 11th ed.; Wolters Kluwer Health: Philadelphia, PA, USA, 2012.
  97. Cho, K.S.; Shin, M.; Kim, S.; Lee, S.B. Recent Advances in Studies on the Therapeutic Potential of Dietary Carotenoids in Neurodegenerative Diseases. Oxid. Med. Cell. Longev. 2018, 2018, 4120458.
  98. Zhang, M.; Cui, Z.; Cui, H.; Wang, Y.; Zhong, C. Astaxanthin protects astrocytes against trauma-induced apoptosis through inhibition of NKCC1 expression via the NF-kappaB signaling pathway. BMC Neurosci. 2017, 18, 42.
  99. Zhang, M.; Cui, Z.; Cui, H.; Cao, Y.; Zhong, C.; Wang, Y. Astaxanthin alleviates cerebral edema by modulating NKCC1 and AQP4 expression after traumatic brain injury in mice. BMC Neurosci. 2016, 17, 60.
  100. Ji, X.; Peng, D.; Zhang, Y.; Zhang, J.; Wang, Y.; Gao, Y.; Lu, N.; Tang, P. Astaxanthin improves cognitive performance in mice following mild traumatic brain injury. Brain Res. 2017, 1659, 88–95.
  101. Zhang, L.; Wang, H.; Fan, Y.; Gao, Y.; Li, X.; Hu, Z.; Ding, K.; Wang, Y.; Wang, X. Fucoxanthin provides neuroprotection in models of traumatic brain injury via the Nrf2-ARE and Nrf2-autophagy pathways. Sci. Rep. 2017, 7, 46763.
  102. Zhong, J.; Jiang, L.; Huang, Z.; Zhang, H.; Cheng, C.; Liu, H.; He, J.; Wu, J.; Darwazeh, R.; Wu, Y.; et al. The long non-coding RNA Neat1 is an important mediator of the therapeutic effect of bexarotene on traumatic brain injury in mice. Brain Behav. Immun. 2017, 65, 183–194.
  103. Zhong, J.; Cheng, C.; Liu, H.; Huang, Z.; Wu, Y.; Teng, Z.; He, J.; Zhang, H.; Wu, J.; Cao, F.; et al. Bexarotene protects against traumatic brain injury in mice partially through apolipoprotein E. Neuroscience 2017, 343, 434–448.
  104. Wang, K.; Zhang, L.; Rao, W.; Su, N.; Hui, H.; Wang, L.; Peng, C.; Tu, Y.; Zhang, S.; Fei, Z. Neuroprotective effects of crocin against traumatic brain injury in mice: Involvement of notch signaling pathway. Neurosci. Lett. 2015, 591, 53–58.
  105. Tan, D.; Yu, X.; Chen, M.; Chen, J.; Xu, J. Lutein protects against severe traumatic brain injury through antiinflammation and antioxidative effects via ICAM1/Nrf2. Mol. Med. Rep. 2017, 16, 4235–4240.
  106. Lauritzen, L.; Hansen, H.S.; Jorgensen, M.H.; Michaelsen, K.F. The essentiality of long chain n-3 fatty acids in relation to development and function of the brain and retina. Prog. Lipid Res. 2001, 40, 1–94.
  107. Matsuoka, Y.; Nishi, D.; Tanima, Y.; Itakura, M.; Kojima, M.; Hamazaki, K.; Noguchi, H.; Hamazaki, T. Serum pro-BDNF/BDNF as a treatment biomarker for response to docosahexaenoic acid in traumatized people vulnerable to developing psychological distress: A randomized controlled trial. Transl. Psychiatry 2015, 5, e596.
  108. Noguchi, H.; Nishi, D.; Matsumura, K.; Hamazaki, K.; Hamazaki, T.; Matsuoka, Y.J. Limited effect of omega-3 fatty acids on the quality of life in survivors of traumatic injury: A randomized, placebo-controlled trial. Prostaglandins Leukot. Essent. Fatty Acids 2017, 127, 1–5.
  109. Gross, B.W.; Gillio, M.; Rinehart, C.D.; Lynch, C.A.; Rogers, F.B. Omega-3 Fatty Acid Supplementation and Warfarin: A Lethal Combination in Traumatic Brain Injury. J. Trauma Nurs. 2017, 24, 15–18.
  110. McCrory, P.; Feddermann-Demont, N.; Dvorak, J.; Cassidy, J.D.; McIntosh, A.; Vos, P.E.; Echemendia, R.J.; Meeuwisse, W.; Tarnutzer, A.A. What is the definition of sports-related concussion: A systematic review. Br. J. Sports Med. 2017, 51, 877–887.
  111. McCrory, P.; Meeuwisse, W.; Dvorak, J.; Aubry, M.; Bailes, J.; Broglio, S.; Cantu, R.C.; Cassidy, D.; Echemendia, R.J.; Castellani, R.J.; et al. Consensus statement on concussion in sport-the 5(th) international conference on concussion in sport held in Berlin, October 2016. Br. J. Sports Med. 2017, 51, 838–847.
  112. Signoretti, S.; Tavazzi, B.; Lazzarino, G.; Vagnozzi, R. The Pathophysiology of Concussive Brain Injury. In Concussion and Traumatic Encephalopathy: Causes, Diagnosis and Management; Victoroff, J., Bigler, E.D., Eds.; Cambridge University Press: Cambridge, UK, 2019; pp. 138–152.
  113. Giza, C.C.; Hovda, D.A. The Neurometabolic Cascade of Concussion. J. Athl. Train. 2001, 36, 228–235.
  114. Giza, C.C.; Hovda, D.A. The new neurometabolic cascade of concussion. Neurosurgery 2014, 75 (Suppl. 4), S24–S33.
  115. Amorini, A.M.; Lazzarino, G.; Di Pietro, V.; Signoretti, S.; Lazzarino, G.; Belli, A.; Tavazzi, B. Metabolic, enzymatic and gene involvement in cerebral glucose dysmetabolism after traumatic brain injury. Biochim. Biophys. Acta 2016, 1862, 679–687.
  116. Vagnozzi, R.; Tavazzi, B.; Signoretti, S.; Amorini, A.M.; Belli, A.; Cimatti, M.; Delfini, R.; Di Pietro, V.; Finocchiaro, A.; Lazzarino, G. Temporal window of metabolic brain vulnerability to concussions: Mitochondrial-related impairment—Part I. Neurosurgery 2007, 61, 379–388.
  117. Di Pietro, V.; Lazzarino, G.; Amorini, A.M.; Signoretti, S.; Hill, L.J.; Porto, E.; Tavazzi, B.; Lazzarino, G.; Belli, A. Fusion or Fission: The Destiny of Mitochondria In Traumatic Brain Injury of Different Severities. Sci. Rep. 2017, 7, 9189.
  118. Vagnozzi, R.; Signoretti, S.; Floris, R.; Marziali, S.; Manara, M.; Amorini, A.M.; Belli, A.; Di Pietro, V.; D’Urso, S.; Pastore, F.S.; et al. Decrease in N-acetylaspartate following concussion may be coupled to decrease in creatine. J. Head Trauma Rehabil. 2013, 28, 284–292.
  119. Vagnozzi, R.; Signoretti, S.; Tavazzi, B.; Floris, R.; Ludovici, A.; Marziali, S.; Tarascio, G.; Amorini, A.M.; Di Pietro, V.; Delfini, R.; et al. Temporal window of metabolic brain vulnerability to concussion: A pilot 1H-magnetic resonance spectroscopic study in concussed athletes—Part III. Neurosurgery 2008, 62, 1286–1295.
  120. Tavazzi, B.; Vagnozzi, R.; Signoretti, S.; Amorini, A.M.; Belli, A.; Cimatti, M.; Delfini, R.; Di Pietro, V.; Finocchiaro, A.; Lazzarino, G. Temporal window of metabolic brain vulnerability to concussions: Oxidative and nitrosative stresses—Part II. Neurosurgery 2007, 61, 390–395.
  121. Mez, J.; Daneshvar, D.H.; Kiernan, P.T.; Abdolmohammadi, B.; Alvarez, V.E.; Huber, B.R.; Alosco, M.L.; Solomon, T.M.; Nowinski, C.J.; McHale, L.; et al. Clinicopathological Evaluation of Chronic Traumatic Encephalopathy in Players of American Football. JAMA 2017, 318, 360–370.
  122. Hoffer, M.E.; Balaban, C.; Slade, M.D.; Tsao, J.W.; Hoffer, B. Amelioration of acute sequelae of blast induced mild traumatic brain injury by N-acetyl cysteine: A double-blind, placebo controlled study. PLoS ONE 2013, 8, e54163.
  123. Oliver, J.M.; Jones, M.T.; Kirk, K.M.; Gable, D.A.; Repshas, J.T.; Johnson, T.A.; Andreasson, U.; Norgren, N.; Blennow, K.; Zetterberg, H. Effect of Docosahexaenoic Acid on a Biomarker of Head Trauma in American Football. Med. Sci. Sports Exerc. 2016, 48, 974–982.
  124. Walter, A.; Finelli, K.; Bai, X.; Arnett, P.; Bream, T.; Seidenberg, P.; Lynch, S.; Johnson, B.; Slobounov, S. Effect of Enzogenol® Supplementation on Cognitive, Executive, and Vestibular/Balance Functioning in Chronic Phase of Concussion. Dev. Neuropsychol. 2017, 42, 93–103.
  125. Smith, S.L.; Andrus, P.K.; Zhang, J.R.; Hall, E.D. Direct measurement of hydroxyl radicals, lipid peroxidation, and blood-brain barrier disruption following unilateral cortical impact head injury in the rat. J. Neurotrauma 1994, 11, 393–404.
  126. Vagnozzi, R.; Marmarou, A.; Tavazzi, B.; Signoretti, S.; Di Pierro, D.; del Bolgia, F.; Amorini, A.M.; Fazzina, G.; Sherkat, S.; Lazzarino, G. Changes of cerebral energy metabolism and lipid peroxidation in rats leading to mitochondrial dysfunction after diffuse brain injury. J. Neurotrauma 1999, 16, 903–913.
  127. Singh, I.N.; Sullivan, P.G.; Deng, Y.; Mbye, L.H.; Hall, E.D. Time course of post-traumatic mitochondrial oxidative damage and dysfunction in a mouse model of focal traumatic brain injury: Implications for neuroprotective therapy. J. Cereb. Blood Flow Metab. 2006, 26, 1407–1418.
  128. Cristofori, L.; Tavazzi, B.; Gambin, R.; Vagnozzi, R.; Vivenza, C.; Amorini, A.M.; Di Pierro, D.; Fazzina, G.; Lazzarino, G. Early onset of lipid peroxidation after human traumatic brain injury: A fatal limitation for the free radical scavenger pharmacological therapy? J. Investig. Med. 2001, 49, 450–458.
  129. Rashno, M.; Sarkaki, A.; Farbood, Y.; Rashno, M.; Khorsandi, L.; Naseri, M.K.G.; Dianat, M. Therapeutic effects of chrysin in a rat model of traumatic brain injury: A behavioral, biochemical, and histological study. Life Sci. 2019, 228, 285–294.
  130. Umemoto, Y.; Patel, A.; Huynh, T.; Chitravanshi, V.C. Wogonin attenuates the deleterious effects of traumatic brain injury in anesthetized Wistar rats. Eur. J. Pharmacol. 2019, 848, 121–130.
  131. Zhang, M.; Huang, L.L.; Teng, C.H.; Wu, F.F.; Ge, L.Y.; Shi, Y.J.; He, Z.L.; Liu, L.; Jiang, C.J.; Hou, R.N.; et al. Isoliquiritigenin Provides Protection and Attenuates Oxidative Stress-Induced Injuries via the Nrf2-ARE Signaling Pathway After Traumatic Brain Injury. Neurochem. Res. 2018, 43, 2435–2445.
  132. Kosari-Nasab, M.; Shokouhi, G.; Ghorbanihaghjo, A.; Abbasi, M.M.; Salari, A.A. Hesperidin attenuates depression-related symptoms in mice with mild traumatic brain injury. Life Sci. 2018, 213, 198–205.
  133. Joo, H.; Bae, J.; Lee, J.S.; Bang, Y.; Lee, B.J.; Park, J.W.; Lee, K.; Cho, J.H.; Bu, Y. Icariin Improves Functional Behavior in a Mouse Model of Traumatic Brain Injury and Promotes Synaptic Plasticity Markers. Planta Med. 2019, 85, 231–238.
  134. Fang, J.; Wang, H.; Zhou, J.; Dai, W.; Zhu, Y.; Zhou, Y.; Wang, X.; Zhou, M. Baicalin provides neuroprotection in traumatic brain injury mice model through Akt/Nrf2 pathway. Drug Des. Dev. Ther. 2018, 12, 2497–2508.
  135. Li, Z.; Zeng, G.; Zheng, X.; Wang, W.; Ling, Y.; Tang, H.; Zhang, J. Neuroprotective effect of formononetin against TBI in rats via suppressing inflammatory reaction in cortical neurons. Biomed. Pharmacother. 2018, 106, 349–354.
  136. Zhao, H.; Liu, Y.; Zeng, J.; Li, D.; Huang, Y. Troxerutin cerebroprotein hydrolysate injection ameliorates neurovascular injury induced by traumatic brain injury—Via endothelial nitric oxide synthase pathway regulation. Int. J. Neurosci. 2018, 128, 1118–1127.
  137. Du, G.; Zhao, Z.; Chen, Y.; Li, Z.; Tian, Y.; Liu, Z.; Liu, B.; Song, J. Quercetin protects rat cortical neurons against traumatic brain injury. Mol. Med. Rep. 2018, 17, 7859–7865.
  138. Shi, Z.; Qiu, W.; Xiao, G.; Cheng, J.; Zhang, N. Resveratrol Attenuates Cognitive Deficits of Traumatic Brain Injury by Activating p38 Signaling in the Brain. Med. Sci. Monit. 2018, 24, 1097–1103.
  139. Kosari-Nasab, M.; Shokouhi, G.; Ghorbanihaghjo, A.; Abbasi, M.M.; Salari, A.A. Anxiolytic- and antidepressant-like effects of Silymarin compared to diazepam and fluoxetine in a mouse model of mild traumatic brain injury. Toxicol. Appl. Pharmacol. 2018, 338, 159–173.
  140. Mirshekar, M.A.; Fanaei, H.; Keikhaei, F.; Javan, F.S. Diosmin improved cognitive deficit and amplified brain electrical activity in the rat model of traumatic brain injury. Biomed. Pharmacother. 2017, 93, 1220–1229.
  141. Jiang, Z.; Zhang, J.; Cai, Y.; Huang, J.; You, L. Catechin attenuates traumatic brain injury-induced blood-brain barrier damage and improves longer-term neurological outcomes in rats. Exp. Physiol. 2017, 102, 1269–1277.
  142. Krishna, G.; Agrawal, R.; Zhuang, Y.; Ying, Z.; Paydar, A.; Harris, N.G.; Royes, L.F.; Gomez-Pinilla, F. 7,8-Dihydroxyflavone facilitates the action exercise to restore plasticity and functionality: Implications for early brain trauma recovery. Biochim. Biophys. Acta Mol. Basis Dis. 2017, 1863, 1204–1213.
  143. Scheff, S.W.; Roberts, K.N. Cognitive assessment of pycnogenol therapy following traumatic brain injury. Neurosci. Lett. 2016, 634, 126–131.
  144. Jiang, L.; Xia, Q.J.; Dong, X.J.; Hu, Y.; Chen, Z.W.; Chen, K.; Wang, K.H.; Liu, J.; Wang, T.H. Neuroprotective effect of breviscapine on traumatic brain injury in rats associated with the inhibition of GSK3beta signaling pathway. Brain Res. 2017, 1660, 1–9.
  145. Wang, Y.; Zhang, C.; Peng, W.; Xia, Z.; Gan, P.; Huang, W.; Shi, Y.; Fan, R. Hydroxysaf fl or yellow A exerts antioxidant effects in a rat model of traumatic brain injury. Mol. Med. Rep. 2016, 14, 3690–3696.
  146. Soltani, Z.; Khaksari, M.; Jafari, E.; Iranpour, M.; Shahrokhi, N. Is genistein neuroprotective in traumatic brain injury? Physiol. Behav. 2015, 152, 26–31.
  147. Cheng, T.; Wang, W.; Li, Q.; Han, X.; Xing, J.; Qi, C.; Lan, X.; Wan, J.; Potts, A.; Guan, F.; et al. Cerebroprotection of flavanol (-)-epicatechin after traumatic brain injury via Nrf2-dependent and -independent pathways. Free Radic. Biol. Med. 2016, 92, 15–28.
  148. Mao, X.; Hao, S.; Zhu, Z.; Zhang, H.; Wu, W.; Xu, F.; Liu, B. Procyanidins protects against oxidative damage and cognitive deficits after traumatic brain injury. Brain Inj. 2015, 29, 86–92.
  149. Erdem, Y.; Tekiner, A.; Erkoc, Y.S.; Yilmaz, M.B.; Celik, H.; Yildirim, A.E.; Tekiner, A.S.; Bayar, M.A. Antiedema effects of proanthocyanidin on experimental traumatic brain edema. Turk. Neurosurg. 2015, 25, 85–89.
  150. Itoh, T.; Tabuchi, M.; Mizuguchi, N.; Imano, M.; Tsubaki, M.; Nishida, S.; Hashimoto, S.; Matsuo, K.; Nakayama, T.; Ito, A.; et al. Neuroprotective effect of (-)-epigallocatechin-3-gallate in rats when administered pre- or post-traumatic brain injury. J. Neural Transm. 2013, 120, 767–783.
  151. Wang, J.W.; Wang, H.D.; Cong, Z.X.; Zhou, X.M.; Xu, J.G.; Jia, Y.; Ding, Y. Puerarin ameliorates oxidative stress in a rodent model of traumatic brain injury. J. Surg. Res. 2014, 186, 328–337.
  152. Xu, J.; Wang, H.; Ding, K.; Zhang, L.; Wang, C.; Li, T.; Wei, W.; Lu, X. Luteolin provides neuroprotection in models of traumatic brain injury via the Nrf2-ARE pathway. Free Radic. Biol. Med. 2014, 71, 186–195.
  153. Cui, Q.J.; Wang, L.Y.; Wei, Z.X.; Qu, W.S. Continual naringin treatment benefits the recovery of traumatic brain injury in rats through reducing oxidative and inflammatory alterations. Neurochem. Res. 2014, 39, 1254–1262.
  154. Chen, S.F.; Hsu, C.W.; Huang, W.H.; Wang, J.Y. Post-injury baicalein improves histological and functional outcomes and reduces inflammatory cytokines after experimental traumatic brain injury. Br. J. Pharmacol. 2008, 155, 1279–1296.
  155. Thau-Zuchman, O.; Ingram, R.; Harvey, G.G.; Cooke, T.; Palmas, F.; Pallier, P.N.; Brook, J.; Priestley, J.V.; Dalli, J.; Lopez-Tremoleda, J.; et al. A single injection of docosahexaenoic acid induces a pro-resolving lipid mediator profile in the injured tissue and a long-lasting reduction in neurological deficit after traumatic brain injury in mice. J. Neurotrauma 2020, 37, 66–79.
  156. Schober, M.E.; Requena, D.F.; Casper, T.C.; Velhorst, A.K.; Lolofie, A.; McFarlane, K.E.; Otto, T.E.; Terry, C.; Gensel, J.C. Docosahexaenoic acid decreased neuroinflammation in rat pups after controlled cortical impact. Exp. Neurol. 2019, 320, 112971.
  157. Berg, R.W.V.; Davidsson, J.; Lidin, E.; Angeria, M.; Risling, M.; Gunther, M. Brain tissue saving effects by single-dose intralesional administration of Neuroprotectin D1 on experimental focal penetrating brain injury in rats. J. Clin. Neurosci. 2019, 64, 227–233.
  158. Zhu, W.; Ding, Y.; Kong, W.; Li, T.; Chen, H. Docosahexaenoic Acid (DHA) Provides Neuroprotection in Traumatic Brain Injury Models via Activating Nrf2-ARE Signaling. Inflammation 2018, 41, 1182–1193.
  159. Tang, R.; Lin, Y.M.; Liu, H.X.; Wang, E.S. 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.
  160. Zhu, W.; Chi, N.; Zou, P.; Chen, H.; Tang, G.; Zhao, W. Effect of docosahexaenoic acid on traumatic brain injury in rats. Exp. Ther. Med. 2017, 14, 4411–4416.
  161. Salberg, S.; Yamakawa, G.; Christensen, J.; Kolb, B.; Mychasiuk, R. Assessment of a nutritional supplement containing resveratrol, prebiotic fiber, and omega-3 fatty acids for the prevention and treatment of mild traumatic brain injury in rats. Neuroscience 2017, 365, 146–157.
  162. Harvey, L.D.; Yin, Y.; Attarwala, I.Y.; Begum, G.; Deng, J.; Yan, H.Q.; Dixon, C.E.; 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.
  163. Schober, M.E.; Requena, D.F.; Abdullah, O.M.; Casper, T.C.; Beachy, J.; Malleske, D.; Pauly, J.R. Dietary Docosahexaenoic Acid Improves Cognitive Function, Tissue Sparing, and Magnetic Resonance Imaging Indices of Edema and White Matter Injury in the Immature Rat after Traumatic Brain Injury. J. Neurotrauma 2016, 33, 390–402.
  164. 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-kappaB pathway following experimental traumatic brain injury. J. Neuroinflamm. 2017, 14, 143.
  165. Yin, Y.; Li, E.; Sun, G.; Yan, H.Q.; Foley, L.M.; Andrzejczuk, L.A.; Attarwala, I.Y.; Hitchens, T.K.; Kiselyov, K.; Dixon, C.E.; et al. Effects of DHA on Hippocampal Autophagy and Lysosome Function After Traumatic Brain Injury. Mol. Neurobiol. 2018, 55, 2454–2470.
  166. Ren, H.; Yang, Z.; Luo, C.; Zeng, H.; Li, P.; Kang, J.X.; Wan, J.B.; He, C.; Su, H. Enriched Endogenous Omega-3 Fatty Acids in Mice Ameliorate Parenchymal Cell Death After Traumatic Brain Injury. Mol. Neurobiol. 2017, 54, 3317–3326.
  167. Wu, A.; Ying, Z.; Gomez-Pinilla, F. Dietary strategy to repair plasma membrane after brain trauma: Implications for plasticity and cognition. Neurorehabil. Neural Repair 2014, 28, 75–84.
  168. Wu, A.; Ying, Z.; Gomez-Pinilla, F. Exercise facilitates the action of dietary DHA on functional recovery after brain trauma. Neuroscience 2013, 248, 655–663.
  169. Russell, K.L.; Berman, N.E.; Levant, B. Low brain DHA content worsens sensorimotor outcomes after TBI and decreases TBI-induced Timp1 expression in juvenile rats. Prostaglandins Leukot. Essent. Fatty Acids 2013, 89, 97–105.
  170. Mills, J.D.; Hadley, K.; Bailes, J.E. Dietary supplementation with the omega-3 fatty acid docosahexaenoic acid in traumatic brain injury. Neurosurgery 2011, 68, 474–481.
More