ROS and brain diseases: the good, the bad, and the ugly

Aurel Popa-Wagner, Smaranda Mitran, Senthilkumar Sivanesan, Edwin Chang, Ana-Maria Buga, Aurel Popa-Wagner, Smaranda Mitran, Senthilkumar Sivanesan, Edwin Chang, Ana-Maria Buga

Abstract

The brain is a major metabolizer of oxygen and yet has relatively feeble protective antioxidant mechanisms. This paper reviews the Janus-faced properties of reactive oxygen species. It will describe the positive aspects of moderately induced ROS but it will also outline recent research findings concerning the impact of oxidative and nitrooxidative stress on neuronal structure and function in neuropsychiatric diseases, including major depression. A common denominator of all neuropsychiatric diseases including schizophrenia and ADHD is an increased inflammatory response of the brain caused either by an exposure to proinflammatory agents during development or an accumulation of degenerated neurons, oxidized proteins, glycated products, or lipid peroxidation in the adult brain. Therefore, modulation of the prooxidant-antioxidant balance provides a therapeutic option which can be used to improve neuroprotection in response to oxidative stress. We also discuss the neuroprotective role of the nuclear factor erythroid 2-related factor (Nrf2) in the aged brain in response to oxidative stressors and nanoparticle-mediated delivery of ROS-scavenging drugs. The antioxidant therapy is a novel therapeutic strategy. However, the available drugs have pleiotropic actions and are not fully characterized in the clinic. Additional clinical trials are needed to assess the risks and benefits of antioxidant therapies for neuropsychiatric disorders.

Figures

Figure 1
Figure 1
Schematic representation of oxidative stress-related mechanisms underlying disease development in Alzheimer's disease (AD), Parkinson disease (PD), stroke, attention deficit and hyperactivity disorders (ADHD), schizophrenia, and depression.

References

    1. Dole M. The natural history of oxygen. The Journal of General Physiology. 1965;49(1):5–27.
    1. Campbell NA, Reece JB. Biology. 7th edition. Pearson-Benjamin Cummings; 2005.
    1. Rich PR. The molecular machinery of Keilin’s respiratory chain. Biochemical Society Transactions. 2003;31(6):1095–1105.
    1. Guglielmotto M, Tamagno E, Danni O. Oxidative stress and hypoxia contribute to Alzheimer’s disease pathogenesis: two sides of the same coin. TheScientificWorldJournal. 2009;9(1):781–791.
    1. Chen S-D, Yang D-I, Lin T-K, Shaw F-Z, Liou C-W, Chuang Y-C. Roles of oxidative stress, apoptosis, PGC-1 and mitochondrial biogenesis in cerebral ischemia. International Journal of Molecular Sciences. 2011;12(10):7199–7215.
    1. Valko M, Leibfritz D, Moncol J, Cronin MTD, Mazur M, Telser J. Free radicals and antioxidants in normal physiological functions and human disease. International Journal of Biochemistry and Cell Biology. 2007;39(1):44–84.
    1. Michel TM, Pülschen D, Thome J. The role of oxidative stress in depressive disorders. Current Pharmaceutical Design. 2012;18(36):5890–5899.
    1. Wang X, Zhao X. Contribution of oxidative damage to antimicrobial lethality. Antimicrobial Agents and Chemotherapy. 2009;53(4):1395–1402.
    1. Dröge W. Free radicals in the physiological control of cell function. Physiological Reviews. 2002;82(1):47–95.
    1. Lu S-P, Lin Feng M-H, Huang H-L, Huang Y-C, Tsou W-I, Lai M-Z. Reactive oxygen species promote raft formation in T lymphocytes. Free Radical Biology & Medicine. 2007;42(7):936–944.
    1. Atkins CM, Sweatt JD. Reactive oxygen species mediate activity-dependent neuron-glia signaling in output fibers of the hippocampus. The Journal of Neuroscience. 1999;19(17):7241–7248.
    1. Hu D, Serrano F, Oury TD, Klann E. Aging-dependent alterations in synaptic plasticity and memory in mice that overexpress extracellular superoxide dismutase. The Journal of Neuroscience. 2006;26(15):3933–3941.
    1. Mehta SL, Kumari S, Mendelev N, Li PA. Selenium preserves mitochondrial function, stimulates mitochondrial biogenesis, and reduces infarct volume after focal cerebral ischemia. BMC Neuroscience. 2012;13, article 79
    1. Badjatia N, Seres D, Carpenter A, et al. Free fatty acids and delayed cerebral ischemia after subarachnoid hemorrhage. Stroke. 2012;43(3):691–696.
    1. Chen G, Jing CH, Liu PP, Ruan D, Wang L. Induction of autophagic cell death in the rat brain caused by iron. The American Journal of the Medical Sciences. 2013;345(5):369–374.
    1. McCracken E, Valeriani V, Simpson C, Jover T, McCulloch J, Dewar D. The lipid peroxidation by-product 4-hydroxynonenal is toxic to axons and oligodendrocytes. Journal of Cerebral Blood Flow and Metabolism. 2000;20(11):1529–1536.
    1. Cui J, Holmes EH, Greene TG, Liu PK. Oxidative DNA damage precedes DNA fragmentation after experimental stroke in rat brain. FASEB Journal. 2000;14(7):955–967.
    1. Panov A, Dikalov S, Shalbuyeva N, Hemendinger R, Greenamyre JT, Rosenfeld J. Species- and tissue-specific relationships between mitochondrial permeability transition and generation of ROS in brain and liver mitochondria of rats and mice. American Journal of Physiology—Cell Physiology. 2007;292(2):C708–C718.
    1. Hansson MJ, Månsson R, Morota S, et al. Calcium-induced generation of reactive oxygen species in brain mitochondria is mediated by permeability transition. Free Radical Biology & Medicine. 2008;45(3):284–294.
    1. Opii WO, Nukala VN, Sultana R, et al. Proteomic identification of oxidized mitochondrial proteins following experimental traumatic brain injury. Journal of Neurotrauma. 2007;24(5):772–789.
    1. Lemasters JJ, Theruvath TP, Zhong Z, Nieminen A-L. Mitochondrial calcium and the permeability transition in cell death. Biochimica et Biophysica Acta. 2009;1787(11):1395–1401.
    1. Rasola A, Sciacovelli M, Pantic B, Bernardi P. Signal transduction to the permeability transition pore. FEBS Letters. 2010;584(10):1989–1996.
    1. Halliwell B. Oxidative stress and neurodegeneration: where are we now? Journal of Neurochemistry. 2006;97(6):1634–1658.
    1. Tobe EH. Mitochondrial dysfunction, oxidative stress, and major depressive disorder. Neuropsychiatric Disease and Treatment. 2013;9:567–573.
    1. Tsaluchidu S, Cocchi M, Tonello L, Puri BK. Fatty acids and oxidative stress in psychiatric disorders. BMC Psychiatry. 2008;8(supplement 1, article S5)
    1. Masood A, Nadeem A, Mustafa SJ, O’Donnell JM. Reversal of oxidative stress-induced anxiety by inhibition of phosphodiesterase-2 in mice. Journal of Pharmacology and Experimental Therapeutics. 2008;326(2):369–379.
    1. Arranz MJ, de Leon J. Pharmacogenetics and pharmacogenomics of schizophrenia: a review of last decade of research. Molecular Psychiatry. 2007;12(8):707–747.
    1. Bouayed J, Rammal H, Younos C, Soulimani R. Positive correlation between peripheral blood granulocyte oxidative status and level of anxiety in mice. European Journal of Pharmacology. 2007;564(1–3):146–149.
    1. Bouayed J, Rammal H, Soulimani R. Oxidative stress and anxiety Relationship and cellular pathways. Oxidative Medicine and Cellular Longevity. 2009;2(2):63–72.
    1. Marazziti D, Baroni S, Picchetti M, et al. Psychiatric disorders and mitochondrial dysfunctions. European Review for Medical and Pharmacological Sciences. 2012;16(2):270–275.
    1. Hovatta I, Juhila J, Donner J. Oxidative stress in anxiety and comorbid disorders. Neuroscience Research. 2010;68(4):261–275.
    1. Ng F, Berk M, Dean O, Bush AI. Oxidative stress in psychiatric disorders: evidence base and therapeutic implications. International Journal of Neuropsychopharmacology. 2008;11(6):851–876.
    1. Auerbach BD, Osterweil EK, Bear MF. Mutations causing syndromic autism define an axis of synaptic pathophysiology. Nature. 2011;480(7375):63–68.
    1. Ben-David E, Shifman S. Networks of neuronal genes affected by common and rare variants in autism spectrum disorders. PLoS Genetics. 2012;8(3)e1002556
    1. Dagda RK, Cherra SJ, III, Kulich SM, Tandon A, Park D, Chu CT. Loss of PINK1 function promotes mitophagy through effects on oxidative stress and mitochondrial fission. The Journal of Biological Chemistry. 2009;284(20):13843–13855.
    1. Boess FG, Hendrix M, van der Staay F-J, et al. Inhibition of phosphodiesterase 2 increases neuronal cGMP, synaptic plasticity and memory performance. Neuropharmacology. 2004;47(7):1081–1092.
    1. Werner C, Raivich G, Cowen M, et al. Importance of NO/cGMP signalling via cGMP-dependent protein kinase II for controlling emotionality and neurobehavioural effects of alcohol. European Journal of Neuroscience. 2004;20(12):3498–3506.
    1. Wang X, Pinto-Duarte A, Sejnowski TJ, Behrens MM. How Nox2-containing NADPH oxidase affects cortical circuits in the NMDA receptor antagonist model of schizophrenia. Antioxidants & Redox Signaling. 2013;18(12):1444–1462.
    1. Rossignol DA, Frye RE. A review of research trends in physiological abnormalities in autism spectrum disorders: immune dysregulation, inflammation, oxidative stress, mitochondrial dysfunction and environmental toxicant exposures. Molecular Psychiatry. 2012;17(4):389–401.
    1. Gu F, Chauhan V, Kaur K, et al. Alterations in mitochondrial DNA copy number and the activities of electron transport chain complexes and pyruvate dehydrogenase in the frontal cortex from subjects with autism. Translational Psychiatry. 2013;3, article e299
    1. Johnson AW, Jaaro-Peled H, Shahani N, et al. Cognitive and motivational deficits together with prefrontal oxidative stress in a mouse model for neuropsychiatric illness. Proceedings of the National Academy of Sciences of the United States of America. 2013;110(30):12462–12467.
    1. Kano S, Colantuoni C, Han F, et al. Genome-wide profiling of multiple histone methylations in olfactory cells: further implications for cellular susceptibility to oxidative stress in schizophrenia. Molecular Psychiatry. 2012;18(7):740–742.
    1. Kubera M, Grygier B, Wrona D, et al. Stimulatory effect of antidepressant drug pretreatment on progression of B16F10 melanoma in high-active male and female C57BL/6J mice. Journal of Neuroimmunology. 2011;240-241:34–44.
    1. Maes M, Fišar Z, Medina M, Scapagnini G, Nowak G, Berk M. New drug targets in depression: inflammatory, cell-mediated immune, oxidative and nitrosative stress, mitochondrial, antioxidant, and neuroprogressive pathways. And new drug candidates—Nrf2 activators and GSK-3 inhibitors. Inflammopharmacology. 2012;20(3):127–150.
    1. Wang X, Lo EH. Triggers and mediators of hemorrhagic transformation in cerebral ischemia. Molecular Neurobiology. 2003;28(3):229–244.
    1. Godínez-Rubí M, Rojas-Mayorquín AE, Ortuño-Sahagún D. Nitric oxide donors as neuroprotective agents after an ischemic stroke-related inflammatory reaction. Oxidative Medicine and Cellular Longevity. 2013;2013:16 pages.297357
    1. Love S. Oxidative stress in brain ischemia. Brain Pathology. 1999;9(1):119–131.
    1. Lakhan SE, Kirchgessner A, Hofer M. Inflammatory mechanisms in ischemic stroke: therapeutic approaches. Journal of Translational Medicine. 2009;7, article 97
    1. del Zoppo GJ, Hallenbeck JM. Advances in the vascular pathophysiology of ischemic stroke. Thrombosis Research. 2000;98(3):V73–V81.
    1. Harari OA, Liao JK. NF-κB and innate immunity in ischemic stroke. Annals of the New York Academy of Sciences. 2010;1207:32–40.
    1. Nichols TC. NF-κB and reperfusion injury. Drug News & Perspectives. 2004;17(2):99–104.
    1. Wei Z, Chigurupati S, Arumugam TV, Jo D-G, Li H, Chan SL. Notch activation enhances the microglia-mediated inflammatory response associated with focal cerebral ischemia. Stroke. 2011;42(9):2589–2594.
    1. Pradillo JM, Fernández-López D, García-Yébenes I, et al. Toll-like receptor 4 is involved in neuroprotection afforded by ischemic preconditioning. Journal of Neurochemistry. 2009;109(1):287–294.
    1. Caso JR, Pradillo JM, Hurtado O, Lorenzo P, Moro MA, Lizasoain I. Toll-like receptor 4 is involved in brain damage and inflammation after experimental stroke. Circulation. 2007;115(12):1599–1608.
    1. Caso JR, Pradillo JM, Hurtado O, Leza JC, Moro MA, Lizasoain I. Toll-like receptor 4 is involved in subacute stress-induced neuroinflammation and in the worsening of experimental stroke. Stroke. 2008;39(4):1314–1320.
    1. Gill R, Tsung A, Billiar T. Linking oxidative stress to inflammation: toll-like receptors. Free Radical Biology & Medicine. 2010;48(9):1121–1132.
    1. Suzuki Y, Hattori K, Hamanaka J, et al. Pharmacological inhibition of TLR4-NOX4 signal protects against neuronal death in transient focal ischemia. Surface Science Reports. 2012;2, article 896
    1. Jenner P, Olanow CW. The pathogenesis of cell death in Parkinson’s disease. Neurology. 2006;66(10, supplement 4):S24–S36.
    1. Hirsch EC, Hunot S. Neuroinflammation in Parkinson’s disease: a target for neuroprotection? The Lancet Neurology. 2009;8(4):382–397.
    1. Gallagher DA, Schapira AHV. Etiopathogenesis and treatment of Parkinson’s disease. Current Topics in Medicinal Chemistry. 2009;9(10):860–868.
    1. Wu D-C, Teismann P, Tieu K, et al. NADPH oxidase mediates oxidative stress in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine model of Parkinson’s disease. Proceedings of the National Academy of Sciences of the United States of America. 2003;100(10):6145–6150.
    1. Blum-Degena D, Müller T, Kuhn W, Gerlach M, Przuntek H, Riederer P. Interleukin-1β and interleukin-6 are elevated in the cerebrospinal fluid of Alzheimer’s and de novo Parkinson’s disease patients. Neuroscience Letters. 1995;202(1-2):17–20.
    1. González-Scarano F, Baltuch G. Microglia as mediators of inflammatory and degenerative diseases. Annual Review of Neuroscience. 1999;22:219–240.
    1. Pabon MM, Bachstetter AD, Hudson CE, Gemma C, Bickford PC. CX3CL1 reduces neurotoxicity and microglial activation in a rat model of Parkinson’s disease. Journal of Neuroinflammation. 2011;8, article 9
    1. Joglar B, Rodriguez-Pallares J, Rodriguez-Perez AI, Rey P, Guerra MJ, Labandeira-Garcia JL. The inflammatory response in the MPTP model of Parkinson’s disease is mediated by brain angiotensin: relevance to progression of the disease. Journal of Neurochemistry. 2009;109(2):656–669.
    1. Reynolds AD, Stone DK, Mosley RL, Gendelman HE. Nitrated α-synuclein-induced alterations in microglial immunity are regulated by CD4+ T cell subsets. Journal of Immunology. 2009;182(7):4137–4149.
    1. Markesbery WR, Carney JM. Oxidative alterations in Alzheimer’s disease. Brain Pathology. 1999;9(1):133–146.
    1. Verri M, Pastoris O, Dossena M, et al. Mitochondrial alterations, oxidative stress and neuroinflammation in Alzheimer’s disease. International Journal of Immunopathology and Pharmacology. 2012;25(2):345–353.
    1. Leuner K, Müller WE, Reichert AS. From mitochondrial dysfunction to amyloid beta formation: novel insights into the pathogenesis of Alzheimer's disease. Molecular Neurobiology. 2012;46(1):186–193.
    1. Younessi P, Yoonessi A. Advanced glycation end-products and their receptor-mediated roles: inflammation and oxidative stress. Iranian Journal of Medical Sciences. 2011;36(3):154–166.
    1. Kalaria RN, Harshbarger-Kelly M, Cohen DL, Premkumar DRD. Molecular aspects of inflammatory and immune responses in Alzheimer’s disease. Neurobiology of Aging. 1996;17(5):687–693.
    1. Bornemann KD, Wiederhold K-H, Pauli C, et al. Aβ-induced inflammatory processes in microglia cells of APP23 transgenic mice. American Journal of Pathology. 2001;158(1):63–73.
    1. Kitazawa M, Yamasaki TR, LaFerla FM. Microglia as a potential bridge between the amyloid β-peptide and tau. Annals of the New York Academy of Sciences. 2004;1035:85–103.
    1. Cribbs DH, Berchtold NC, Perreau V, et al. Extensive innate immune gene activation accompanies brain aging, increasing vulnerability to cognitive decline and neurodegeneration: a microarray study. Journal of Neuroinflammation. 2012;9, article 179
    1. Zhang W, Wang L-Z, Yu J-T, Chi Z-F, Tan L. Increased expressions of TLR2 and TLR4 on peripheral blood mononuclear cells from patients with Alzheimer’s disease. Journal of the Neurological Sciences. 2012;315(1-2):67–71.
    1. Jin J-J, Kim H-D, Maxwell JA, Li L, Fukuchi K-I. Toll-like receptor 4-dependent upregulation of cytokines in a transgenic mouse model of Alzheimer’s disease. Journal of Neuroinflammation. 2008;5, article 23
    1. Tang S-C, Lathia JD, Selvaraj PK, et al. Toll-like receptor-4 mediates neuronal apoptosis induced by amyloid β-peptide and the membrane lipid peroxidation product 4-hydroxynonenal. Experimental Neurology. 2008;213(1):114–121.
    1. Balistreri CR, Colonna-Romano G, Lio D, Candore G, Caruso C. TLR4 polymorphisms and ageing: Implications for the pathophysiology of age-related diseases. Journal of Clinical Immunology. 2009;29(4):406–415.
    1. Drechsel DA, Estévez AG, Barbeito L, Beckman JS. Nitric oxide-mediated oxidative damage and the progressive demise of motor neurons in ALS. Neurotoxicity Research. 2012;22(4):251–264.
    1. Gonsette RE. Neurodegeneration in multiple sclerosis: the role of oxidative stress and excitotoxicity. Journal of the Neurological Sciences. 2008;274(1-2):48–53.
    1. Zeis T, Probst A, Steck AJ, Stadelmann C, Brück W, Schaeren-Wiemers N. Molecular changes in white matter adjacent to an active demyelinating lesion in early multiple sclerosis: molecular changes in MS periplaque white matter. Brain Pathology. 2009;19(3):459–466.
    1. Fischer MT, Sharma R, Lim JL, et al. NADPH oxidase expression in active multiple sclerosis lesions in relation to oxidative tissue damage and mitochondrial injury. Brain. 2012;135(3):886–899.
    1. Ratanayake U, Quinn T, Walker DW, Dickinson H. Cytokines and the neurodevelopmental basis of mental illness. Frontiers in Neuroscience. 2013;17(7, article 180)
    1. Meyer U, Nyffeler M, Engler A, et al. The time of prenatal immune challenge determines the specificity of inflammation-mediated brain and behavioral pathology. The Journal of Neuroscience. 2006;26(18):4752–4762.
    1. Gilmore JH, Jarskog LF, Vadlamudi S, Lauder JM. Prenatal infection and risk for schizophrenia: IL-1β, IL-6, and TNFα inhibit cortical neuron dendrite development. Neuropsychopharmacology. 2004;29(7):1221–1229.
    1. Chez MG, Dowling T, Patel PB, Khanna P, Kominsky M. Elevation of tumor necrosis factor-alpha in cerebrospinal fluid of autistic childre. Pediatric Neurology. 2007;36(6):361–365.
    1. Potvin S, Stip E, Sepehry AA, Gendron A, Bah R, Kouassi E. Inflammatory cytokine alterations in schizophrenia: a systematic quantitative review. Biological Psychiatry. 2008;63(8):801–808.
    1. Ashwood P, Krakowiak P, Hertz-Picciotto I, Hansen R, Pessah I, Van de Water J. Elevated plasma cytokines in autism spectrum disorders provide evidence of immune dysfunction and are associated with impaired behavioral outcome. Brain, Behavior, and Immunity. 2011;25(1):40–45.
    1. Bilbo SD, Schwarz JM. Early-life programming of later-life brain and behavior: a critical role for the immune system. Frontiers in Behavioral Neuroscience. 2009;3, article 14
    1. Bennet L, Gunn A. The fetal origins of adult mental illness. In: Wintour-Coghlan M, Owens J, editors. Early Life Origins of Health and Disease (Advances in Experimental Medicine and Biology) New York, NY, USA: Springer; 2006. pp. 204–211.
    1. Morgan JT, Chana G, Pardo CA, et al. Microglial activation and increased microglial density observed in the dorsolateral prefrontal cortex in autism. Biological Psychiatry. 2010;68(4):368–376.
    1. Fredericks CA, Drabant EM, Edge MD, et al. Healthy young women with serotonin transporter SS polymorphism show a pro-inflammatory bias under resting and stress conditions. Brain, Behavior, and Immunity. 2010;24(3):350–357.
    1. Macchi F, Homberg JR, Calabrese F, et al. Altered inflammatory responsiveness in serotonin transporter mutant rats. Journal of Neuroinflammation. 2013;10, article 116
    1. Monji A, Kato TA, Mizoguchi Y, et al. Neuroinflammation in schizophrenia especially focused on the role of microglia. Progress in Neuro-Psychopharmacology & Biological Psychiatry. 2013;42:115–121.
    1. Liaury K, Miyaoka T, Tsumori T, et al. Morphological features of microglial cells in the hippocampal dentate gyrus of Gunn rat: a possible schizophrenia animal model. Journal of Neuroinflammation. 2012;9, article 56
    1. Post RM. Kindling and sensitization as models for affective episode recurrence, cyclicity, and tolerance phenomena. Neuroscience and Biobehavioral Reviews. 2007;31(6):858–873.
    1. Berk M, Kapczinski F, Andreazza AC, et al. Pathways underlying neuroprogression in bipolar disorder: focus on inflammation, oxidative stress and neurotrophic factors. Neuroscience and Biobehavioral Reviews. 2011;35(3):804–817.
    1. Iyengar RL, Gandhi S, Aneja A, et al. NSAIDs are associated with lower depression scores in patients with osteoarthritis. American Journal of Medicine. 2013;126(11):1017.e11–1017.e18.
    1. Harris JL, Yeh HW, Choi IY, et al. Altered neurochemical profile after traumatic brain injury: 1H-MRS biomarkers of pathological mechanisms. Journal of Cerebral Blood Flow & Metabolism. 2012;32(12):2122–2134.
    1. Rossignol DA, Frye RE. A review of research trends in physiological abnormalities in autism spectrum disorders: immune dysregulation, inflammation, oxidative stress, mitochondrial dysfunction and environmental toxicant exposures. Molecular Psychiatry. 2012;17(4):389–401.
    1. Ming X, Brimacombe M, Chaaban J, Zimmerman-Bier B, Wagner GC. Autism spectrum disorders: concurrent clinical disorders. Journal of Child Neurology. 2008;23(1):6–13.
    1. Frye RE, Delatorre R, Taylor H, et al. Redox metabolism abnormalities in autistic children associated with mitochondrial disease. Translational Psychiatry. 2013;3, article e273
    1. Rose S, Melnyk S, Pavliv O, et al. Evidence of oxidative damage and inflammation associated with low glutathione redox status in the autism brain. Translational Psychiatry. 2012;10(2, article e134)
    1. Kulak A, Steullet P, Cabungcal JH, et al. Redox dysregulation in the pathophysiology of schizophrenia and bipolar disorder: insights from animal models. Antioxidants & Redox Signaling. 2013;18(12):1428–1443.
    1. Jorgensen A, Krogh J, Miskowiak K, et al. Systemic oxidatively generated DNA/RNA damage in clinical depression: associations to symptom severity and response to electroconvulsive therapy. Journal of Affective Disorders. 2013;149(1–3):355–362.
    1. Durand T, De Felice C, Signorini C, et al. F(2)-Dihomo-isoprostanes and brain white matter damage in stage 1 Rett syndrome. Biochimie. 2013;95:86–90.
    1. Flatow J, Buckley P, Miller BJ. Meta-analysis of oxidative stress in schizophrenia. Biological Psychiatry. 2013;74(6):400–409.
    1. Bulut M, Selek S, Gergerlioglu HS, et al. Malondialdehyde levels in adult attention-deficit hyperactivity disorder. Journal of Psychiatry and Neuroscience. 2007;32(6):435–438.
    1. Dwivedi Y, Rizavi HS, Conley RR, Roberts RC, Tamminga CA, Pandey GN. Altered gene expression of brain-derived neurotrophic factor and receptor tyrosine kinase B in postmortem brain of suicide subjects. Archives of General Psychiatry. 2003;60(8):804–815.
    1. Sklar P, Gabriel SB, McInnis MG, et al. Family-based association study of 76 candidate genes in bipolar disorder: BDNF is a potential risk locus. Molecular Psychiatry. 2002;7(6):579–593.
    1. Sen S, Nesse RM, Stoltenberg SF, et al. A BDNF coding variant is associated with the NEO personality inventory domain neuroticism, a risk factor for depression. Neuropsychopharmacology. 2003;28(2):397–401.
    1. Kaufman J, Yang B-Z, Douglas-Palumberi H, et al. Brain-derived neurotrophic factor-5-HTTLPR gene interactions and environmental modifiers of depression in children. Biological Psychiatry. 2006;59(8):673–680.
    1. Chen Z-Y, Jing D, Bath KG, et al. Genetic variant BDNF (Val66Met) polymorphism alters anxiety-related behavior. Science. 2006;314(5796):140–143.
    1. Sen S, Duman R, Sanacora G. Serum brain-derived neurotrophic factor, depression, and antidepressant medications: meta-analyses and implications. Biological Psychiatry. 2008;64(6):527–532.
    1. Versace A, Andreazza AC, Young LT, et al. Elevated serum measures of lipid peroxidation and abnormal prefrontal white matter in euthymic bipolar adults: toward peripheral biomarkers of bipolar disorder. Molecular Psychiatry. 2013
    1. Najjar S, Pearlman DM, Hirsch S, et al. Brain biopsy findings link major depressive disorder to neuroinflammation, oxidative stress, and neurovascular dysfunction: a case report. Biological Psychiatry. 2013
    1. Kosenko EA, Aliev G, Tikhonova LA, Li Y, Poghosyan AC, Kaminsky YG. Antioxidant status and energy state of erythrocytes in Alzheimer dementia: probing for markers. CNS & Neurological Disorders Drug Targets. 2012;11(7):926–932.
    1. Cardoso SM, Proença MT, Santos S, Santana I, Oliveira CR. Cytochrome c oxidase is decreased in Alzheimer’s disease platelets. Neurobiology of Aging. 2004;25(1):105–110.
    1. Bosetti F, Brizzi F, Barogi S, et al. Cytochrome c oxidase and mitochondrial F1F0-ATPase (ATP synthase) activities in platelets and brain from patients with Alzheimer’s disease. Neurobiology of Aging. 2002;23(3):371–376.
    1. Mancuso M, Filosto M, Bosetti F, et al. Decreased platelet cytochrome c oxidase activity is accompanied by increased blood lactate concentration during exercise in patients with Alzheimer disease. Experimental Neurology. 2003;182(2):421–426.
    1. Leuner K, Schulz K, Schütt T, et al. Peripheral mitochondrial dysfunction in Alzheimer's disease: focus on lymphocytes. Molecular Neurobiology. 2012;46(1):194–204.
    1. Puertas MC, Martínez-Martos JM, Cobo MP, Carrera MP, Mayas MD, Ramírez-Expósito MJ. Plasma oxidative stress parameters in men and women with early stage Alzheimer type dementia. Experimental Gerontology. 2012;47(8):625–630.
    1. Okun E, Arumugam TV, Tang S-C, et al. The organotellurium compound ammonium trichloro(dioxoethylene-0,0′) tellurate enhances neuronal survival and improves functional outcome in an ischemic stroke model in mice. Journal of Neurochemistry. 2007;102(4):1232–1241.
    1. Nagotani S, Hayashi T, Sato K, et al. Reduction of cerebral infarction in stroke-prone spontaneously hypertensive rats by statins associated with amelioration of oxidative stress. Stroke. 2005;36(3):670–672.
    1. Lee WC, Wong HY, Chai YY, et al. Lipid peroxidation dysregulation in ischemic stroke: plasma 4-HNE as a potential biomarker? Biochemical and Biophysical Research Communications. 2012;425(4):842–847.
    1. Selley ML. (E)-4-Hydroxy-2-nonenal may be involved in the pathogenesis of Parkinson’s disease. Free Radical Biology & Medicine. 1998;25(2):169–174.
    1. Ilic TV, Jovanovic M, Jovicic A, Tomovic M. Oxidative stress indicators are elevated in de novo Parkinson’s disease patients. Functional Neurology. 1999;14(3):141–147.
    1. Bulut M, Selek S, Bez Y, et al. Lipid peroxidation markers in adult attention deficit hyperactivity disorder: new findings for oxidative stress. Psychiatry Research. 2013;209(3):638–642.
    1. Akyol Ö, Herken H, Uz E, et al. The indices of endogenous oxidative and antioxidative processes in plasma from schizophrenic patients: the possible role of oxidant/antioxidant imbalance. Progress in Neuro-Psychopharmacology & Biological Psychiatry. 2002;26(5):995–1005.
    1. Ceylan M, Sener S, Bayraktar AC, Kavutcu M. Oxidative imbalance in child and adolescent patients with attention-deficit/hyperactivity disorder. Progress in Neuro-Psychopharmacology & Biological Psychiatry. 2010;34(8):1491–1494.
    1. Sanders LH, Timothy Greenamyre J. Oxidative damage to macromolecules in human Parkinson disease and the rotenone model. Free Radical Biology & Medicine. 2013;62:111–120.
    1. Gulec M, Ozkol H, Selvi Y, et al. Oxidative stress in patients with primary insomnia. Progress in Neuro-Psychopharmacology & Biological Psychiatry. 2012;37(2):247–251.
    1. Kalra J, Rajput AH, Mantha SV, Chaudhary AK, Prasad K. Oxygen free radical producing activity of polymorphonuclear leukocytes in patients with Parkinson’s disease. Molecular and Cellular Biochemistry. 1992;112(2):181–186.
    1. Cristalli DO, Arnal N, Marra FA, de Alaniz MJT, Marra CA. Peripheral markers in neurodegenerative patients and their first-degree relatives. Journal of the Neurological Sciences. 2012;314(1-2):48–56.
    1. Kouti L, Noroozian M, Akhondzadeh S, et al. Nitric oxide and peroxynitrite serum levels in Parkinson's disease: correlation of oxidative stress and the severity of the disease. European Review for Medical and Pharmacological Sciences. 2013;17(7):964–970.
    1. Haxaire C, Turpin FR, Potier B, et al. Reversal of age-related oxidative stress prevents hippocampal synaptic plasticity deficits by protecting d-serine-dependent NMDA receptor activation. Aging Cell. 2012;11(2):336–344.
    1. Qin L, Liu Y, Hong JS, Crews FT. NADPH oxidase and aging drive microglial activation, oxidative stress, and dopaminergic neurodegeneration following systemic LPS administration. Glia. 2013;61(6):855–868.
    1. Milani P, Ambrosi G, Gammoh O, Blandini F, Cereda C. SOD1 and DJ-1 converge at Nrf2 pathway: a clue for antioxidant therapeutic potential in neurodegeneration. Oxidative Medicine and Cellular Longevity. 2013;2013:12 pages.836760
    1. Cheng MY, Lee I-P, Jin M, et al. An insult-inducible vector system activated by hypoxia and oxidative stress for neuronal gene therapy. Translational Stroke Research. 2011;2(1):92–100.
    1. Li Y, Paonessa JD, Zhang Y. Mechanism of chemical activation of Nrf2. PLoS One. 2012;7(4)e35122
    1. Gan L, Vargas MR, Johnson DA, Johnson JA. Astrocyte-specific overexpression of Nrf2 delays motor pathology and synuclein aggregation throughout the CNS in the alpha-synuclein mutant (A53T) mouse model. The Journal of Neuroscience. 2012;32(49):17775–17787.
    1. Son TG, Kawamoto EM, Yu QS, Greig NH, Mattson MP, Camandola S. Naphthazarin protects against glutamate-induced neuronal death via activation of the Nrf2/ARE pathway. Biochemical and Biophysical Research Communications. 2013;433(4):602–606.
    1. Dinkova-Kostova AT, Holtzclaw WD, Cole RN, et al. Direct evidence that sulfhydryl groups of Keap1 are the sensors regulating induction of phase 2 enzymes that protect against carcinogens and oxidants. Proceedings of the National Academy of Sciences of the United States of America. 2002;99(18):11908–11913.
    1. Hong F, Freeman ML, Liebler DC. Identification of sensor cysteines in human Keap1 modified by the cancer chemopreventive agent sulforaphane. Chemical Research in Toxicology. 2005;18(12):1917–1926.
    1. Jiang H, Tian X, Guo Y, Duan W, Bu H, Li C. Activation of nuclear factor erythroid 2-related factor 2 cytoprotective signaling by curcumin protect primary spinal cord astrocytes against oxidative toxicity. Biological and Pharmaceutical Bulletin. 2011;34(8):1194–1197.
    1. Kaidery NA, Banerjee R, Yang L, et al. Targeting Nrf2-mediated gene transcription by extremely potent synthetic triterpenoids attenuate dopaminergic neurotoxicity in the MPTP mouse model of Parkinson's disease. Antioxidants & Redox Signaling. 2013;18(2):139–157.
    1. L’Episcopo F, Tirolo C, Testa N, et al. Aging-induced Nrf2-ARE pathway disruption in the subventricular zone drives neurogenic impairment in Parkinsonian mice via PI3K-Wnt/β-Catenin dysregulation. The Journal of Neuroscience. 2013;33(4):1462–1485.
    1. Pérez-de-Puig I, Martín A, Gorina R, de la Rosa X, Martinez E, Planas AM. Induction of hemeoxygenase-1 expression after inhibition of hemeoxygenase activity promotes inflammation and worsens ischemic brain damage in mice. Neuroscience. 2013;243:22–32.
    1. Buga AM, Scholz CJ, Kumar S, et al. Identification of new therapeutic targets by genome-wide analysis of gene expression in the ipsilateral cortex of aged rats after stroke. PLoS One. 2012;7e50985
    1. Bousquet M, Calon F, Cicchetti F. Impact of omega-3 fatty acids in Parkinson’s disease. Ageing Research Reviews. 2011;10(4):453–463.
    1. Joshi K, Lad S, Kale M, et al. Supplementation with flax oil and vitamin C improves the outcome of Attention Deficit Hyperactivity Disorder (ADHD) Prostaglandins Leukotrienes and Essential Fatty Acids. 2006;74(1):17–21.
    1. Richardson AJ, Montgomery P. The Oxford-Durham Study: a randomized, controlled trial of dietary supplementation with fatty acids in children with developmental coordination disorder. Pediatrics. 2005;115(5):1360–1366.
    1. Kamat CD, Gadal S, Mhatre M, Williamson KS, Pye QN, Hensley K. Antioxidants in central nervous system diseases: preclinical promise and translational challenges. Journal of Alzheimer’s Disease. 2008;15(3):473–493.
    1. Bjelakovic G, Nikolova D, Gluud LL, Simonetti RG, Gluud C. Mortality in randomized trials of antioxidant supplements for primary and secondary prevention: systematic review and meta-analysis. The Journal of the American Medical Association. 2007;297(8):842–857.
    1. Høyer-Hansen M, Nordbrandt SPS, Jäättelä M. Autophagy as a basis for the health-promoting effects of vitamin D. Trends in Molecular Medicine. 2010;16(7):295–302.
    1. Tuohimaa P, Keisala T, Minasyan A, Cachat J, Kalueff A. Vitamin D, nervous system and aging. Psychoneuroendocrinology. 2009;34(1):S278–S286.
    1. Kaundal RK, Sharma SS. Peroxisome proliferator-activated receptor γ agonists as neuroprotective agents. Drug News & Perspectives. 2010;23(4):241–256.
    1. Polvani S, Tarocchi M, Galli A. PPAR and oxidative stress: Con(β) catenating NRF2 and FOXO. PPAR Research. 2012;2012:15 pages.641087
    1. Haraguchi T, Iwasaki K, Takasaki K, et al. Telmisartan, a partial agonist of peroxisome proliferator-activated receptor γ, improves impairment of spatial memory and hippocampal apoptosis in rats treated with repeated cerebral ischemia. Brain Research. 2010;1353(1):125–132.
    1. Washida K, Ihara M, Nishio K, et al. Nonhypotensive dose of telmisartan attenuates cognitive impairment partially due to peroxisome proliferator-activated receptor-γ activation in mice with chronic cerebral hypoperfusion. Stroke. 2010;41(8):1798–1806.
    1. Kishi T, Hirooka Y, Sunagawa K. Telmisartan protects against cognitive decline via up-regulation of brain-derived neurotrophic factor/tropomyosin-related kinase B in hippocampus of hypertensive rats. Journal of Cardiology. 2012;60(6):489–494.
    1. Xu F, Li J, Ni W, Shen YW, Zhang XP. Peroxisome proliferator-activated receptor-γ agonist 15d-prostaglandin J2 mediates neuronal autophagy after cerebral ischemia-reperfusion injury. PLoS One. 2013;8(1)e55080
    1. Tureyen K, Kapadia R, Bowen KK, et al. Peroxisome proliferator-activated receptor-γ agonists induce neuroprotection following transient focal ischemia in normotensive, normoglycemic as well as hypertensive and type-2 diabetic rodents. Journal of Neurochemistry. 2007;101(1):41–56.
    1. Park S-W, Yi J-H, Miranpuri G, et al. Thiazolidinedione class of peroxisome proliferator-activated receptor γ agonists prevents neuronal damage, motor dysfunction, myelin loss, neuropathic pain, and inflammation after spinal cord injury in adult rats. Journal of Pharmacology and Experimental Therapeutics. 2007;320(3):1002–1012.
    1. Pereira MP, Hurtado O, Cárdenas A, et al. The nonthiazolidinedione PPARγ agonist L-796,449 is neuroprotective in experimental stroke. Journal of Neuropathology and Experimental Neurology. 2005;64(9):797–805.
    1. Chungpaibulpatana J, Sumpatanarax T, Thadakul N, Chantharatreerat C, Konkaew M, Aroonlimsawas M. Hyperbaric oxygen therapy in Thai autistic children. Journal of the Medical Association of Thailand. 2008;91(8):1232–1238.
    1. Rossignol DA, Rossignol LW. Hyperbaric oxygen therapy may improve symptoms in autistic children. Medical Hypotheses. 2006;67(2):216–228.
    1. Joseph C, Buga AM, Vintilescu R, et al. Prolonged gaseous hypothermia prevents the upregulation of phagocytosis-specific protein annexin 1 and causes low-amplitude EEG activity in the aged rat brain after cerebral ischemia. Journal of Cerebral Blood Flow & Metabolism. 2012;32(8):1632–1642.
    1. Ji Q, Hui K, Zhang L, Sun X, Li W, Duan M. The effect of hydrogen-rich saline on the brain of rats with transient ischemia. Journal of Surgical Research. 2011;168(1):e95–e101.
    1. Li J, Dong Y, Chen H, et al. Protective effects of hydrogen-rich saline in a rat model of permanent focal cerebral ischemia via reducing oxidative stress and inflammatory cytokines. Brain Research. 2012;1486(1):103–111.
    1. Khairova R, Pawar R, Salvadore G, et al. Effects of lithium on oxidative stress parameters in healthy subjects. Molecular Medicine Reports. 2012;5(3):680–682.
    1. Chuang DM, Wang Z, Chiu CT. GSK-3 as a target for lithium-induced neuroprotection against excitotoxicity in neuronal cultures and animal models of ischemic stroke. Frontiers in Molecular Neuroscience. 2011;4, article 15
    1. Pignitter M, Gorren ACF, Nedeianu S, Schmidt K, Mayer B. Inefficient spin trapping of superoxide in the presence of nitric-oxide: implications for studies on nitric-oxide synthase uncoupling. Free Radical Biology & Medicine. 2006;41(3):455–463.
    1. Talarowska M, Gałecki P, Maes M, et al. Nitric oxide plasma concentration associated with cognitive impairment in patients with recurrent depressive disorder. Neuroscience Letters. 2012;510(2):127–131.
    1. Yoshitomi T, Hirayama A, Nagasaki Y. The ROS scavenging and renal protective effects of pH-responsive nitroxide radical-containing nanoparticles. Biomaterials. 2011;32(31):8021–8028.
    1. Michel TM, Sheldrick AJ, Camara S, Grnblatt E, Schneider F, Riederer P. Alteration of the pro-oxidant xanthine oxidase (XO) in the thalamus and occipital cortex of patients with schizophrenia. World Journal of Biological Psychiatry. 2011;12(8):588–597.
    1. Hu X, Zhou H, Zhang D, et al. Clozapine protects dopaminergic neurons from inflammation-induced damage by inhibiting microglial overactivation. Journal of NeuroImmune Pharmacology. 2012;7(1):187–201.
    1. Pandya CD, Howell KR, Pillai A. Antioxidants as potential therapeutics for neuropsychiatric disorders. Progress in Neuropsychopharmacology & Biological Psychiatry. 2013;46:214–223.
    1. Dodd S, Maes M, Anderson G, Dean OM, Moylan S, Berk M. Putative neuroprotective agents in neuropsychiatric disorders. Progress in Neuro-Psychopharmacology & Biological Psychiatry. 2013;42:135–145.
    1. Grillo RW, Ottoni GL, Leke R, Souza DO, Portela LV, Lara DR. Reduced serum BDNF levels in schizophrenic patients on clozapine or typical antipsychotics. Journal of Psychiatric Research. 2007;41(1-2):31–35.
    1. Pedrini M, Chendo I, Grande I, et al. Serum brain-derived neurotrophic factor and clozapine daily dose in patients with schizophrenia: a positive correlation. Neuroscience Letters. 2011;491(3):207–210.
    1. Lim C-M, Kim S-W, Park J-Y, Kim C, Yoon SH, Lee J-K. Fluoxetine affords robust neuroprotection in the postischemic brain via its anti-inflammatory effect. Journal of Neuroscience Research. 2009;87(4):1037–1045.
    1. Wehner NG, Skov M, Shopp G, Rocca MS, Clarke J. Effects of natalizumab, an α4 integrin inhibitor, on fertility in male and female guinea pigs. Birth Defects Research B. 2009;86(2):108–116.

Source: PubMed

3
订阅