Skip to main content
Log in

Role of Free Radicals in the Neurodegenerative Diseases

Therapeutic Implications for Antioxidant Treatment

  • Review Article
  • Published:
Drugs & Aging Aims and scope Submit manuscript

Abstract

Free radicals and other so-called ‘reactive species’ are constantly produced in the brain in vivo. Some arise by ‘accidents of chemistry’, an example of which may be the leakage of electrons from the mitochondrial electron transport chain to generate superoxide radical (O2 ). Others are generated for useful purposes, such as the role of nitric oxide in neurotransmission and the production of O2 by activated microglia. Because of its high ATP demand, the brain consumes O2 rapidly, and is thus susceptible to interference with mitochondrial function, which can in turn lead to increased O2 formation. The brain contains multiple antioxidant defences, of which the mitochondrial manganese-containing superoxide dismutase and reduced glutathione seem especially important. Iron is a powerful promoter of free radical damage, able to catalyse generation of highly reactive hydroxyl, alkoxyl and peroxyl radicals from hydrogen peroxide and lipid peroxides, respectively. Although most iron in the brain is stored in ferritin, ‘catalytic’ iron is readily mobilised from injured brain tissue.

Increased levels of oxidative damage to DNA, lipids and proteins have been detected by a range of assays in post-mortem tissues from patients with Parkinson’s disease, Alzheimer’s disease and amyotrophic lateral sclerosis, and at least some of these changes may occur early in disease progression. The accumulation and precipitation of proteins that occur in these diseases may be aggravated by oxidative damage, and may in turn cause more oxidative damage by interfering with the function of the proteasome. Indeed, it has been shown that proteasomal inhibition increases levels of oxidative damage not only to proteins but also to other biomolecules. Hence, there are many attempts to develop antioxidants that can cross the blood-brain barrier and decrease oxidative damage.

Natural antioxidants such as vitamin E (tocopherol), carotenoids and flavonoids do not readily enter the brain in the adult, and the lazaroid antioxidant tirilazad (U-74006F) appears to localise in the blood-brain barrier. Other antioxidants under development include modified spin traps and low molecular mass scavengers of O2 . One possible source of lead compounds is the use of traditional remedies claimed to improve brain function. Little is known about the impact of dietary antioxidants upon the development and progression of neurodegenerative diseases, especially Alzheimer’s disease. Several agents already in therapeutic use might exert some of their effects by antioxidant action, including selegiline (deprenyl), apomorphine and nitecapone.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Table I
Table II
Fig. 1

Similar content being viewed by others

References

  1. Halliwell B, Gutteridge JMC. Free radicals in biology and medicine. 3rd ed. Oxford: University Press, 1999

    Google Scholar 

  2. Sen CK, Sies H, Baeuerle PA, editors. Antioxidant and redox regulation of genes. San Diego (CA): Academic Press, 2000

    Google Scholar 

  3. Babior BM. NADPH oxidase: an update. Blood 1999; 93: 1464–76

    PubMed  CAS  Google Scholar 

  4. Bowie A, O’Neill LAJ. Oxidative stress and nuclear factor-kB activation. Biochem Pharmacol 2000; 59: 13–23

    PubMed  CAS  Google Scholar 

  5. Hobbs AJ, Higgs A, Moncada S. Inhibition of nitric oxide synthase as a potential therapeutic target. Annu Rev Pharmacol Toxicol 1999; 39: 191–220

    PubMed  CAS  Google Scholar 

  6. Fridovich I. Superoxide anion radical (O2 ), superoxide dismutases, and related matters. J Biol Chem 1997; 272: 18515–7

    PubMed  CAS  Google Scholar 

  7. Bolann BJ, Ulvik RJ. On the limited ability of superoxide to release iron from ferritin. Eur J Biochem 1990; 193: 899–904

    PubMed  CAS  Google Scholar 

  8. Liochev SL. The role of iron-sulfur clusters in in vivo hydroxyl radical production. Free Radic Res 1996; 25: 69–84

    Google Scholar 

  9. Chance B, Sies H, Boveris A. Hydroperoxide metabolism in mammalian organs. Physiol Rev 1979; 59: 527–605

    PubMed  CAS  Google Scholar 

  10. De Groot H, Littauer A. Hypoxia, reactive oxygen, and cell injury. Free Radic Biol Med 1989; 6: 541–51

    PubMed  Google Scholar 

  11. Levine RL, Berlett BS, Moskovitz J, et al. Methionine residues may protect proteins from critical oxidative damage. Mech Ageing Dev 1999; 107: 323–32

    PubMed  CAS  Google Scholar 

  12. Hampton MB, Fadeel B, Orrenius S. Redox regulation of the caspases during apoptosis. Ann NY Acad Sci 1998; 854: 328–35

    PubMed  CAS  Google Scholar 

  13. Borutaite V, Brown GC. Caspases are reversibly inactivated by hydrogen peroxide. FEBS Lett 2001; 500: 114–8

    PubMed  CAS  Google Scholar 

  14. Halliwell B, Clement MV, Long LH. Hydrogen peroxide in the human body. FEBS Lett 2000; 486: 10–13

    PubMed  CAS  Google Scholar 

  15. Long LH, Halliwell B. Coffee drinking increases levels of urinary H2O2 detected in healthy volunteers. Free Radic Res 2000; 32: 463–7

    PubMed  CAS  Google Scholar 

  16. Puppo A, Halliwell B. Formation of hydroxyl radicals in biological systems: does myoglobin stimulate hydroxyl radical formation from hydrogen peroxide? Free Radic Res Commun 1988; 4: 415–22

    PubMed  CAS  Google Scholar 

  17. Harel S, Salan MA, Kanner J. Iron release from metmyoglobin, methaemoglobin and cytochrome c by a system generating hydrogen peroxide. Free Radic Res Commun 1988; 5: 11–9

    PubMed  CAS  Google Scholar 

  18. Wasantwisut E. Nutrition and development: other micronutrients’ effect on growth and cognition. SEA J Trop Med Pub Health 1997; 2: 78–82

    Google Scholar 

  19. Halliwell B, Gutteridge JM. Oxygen toxicity, oxygen radicals, transition metals and disease. Biochem J 1984; 219: 1–14

    PubMed  CAS  Google Scholar 

  20. Halliwell B, Gutteridge JM. Role of free radicals and catalytic metal ions in human disease: an overview. Methods Enzymol 1990; 186: 1–85

    PubMed  CAS  Google Scholar 

  21. Halliwell B. Vitamin C: poison, prophylactic or panacea? Trends Biochem Sci 1999; 24: 255–9

    PubMed  CAS  Google Scholar 

  22. Von Sonntag C. The chemical basis of radiation biology. London: Taylor and Francis, 1987

    Google Scholar 

  23. Dizdaroglu M. Chemical determination of oxidative DNA damage by gas chromatography-mass spectrometry. Methods Enzymol 1994; 234: 3–16

    PubMed  CAS  Google Scholar 

  24. Kasai H. Analysis of a form of oxidative DNA damage, 8-hydroxy-2′-deoxyguanosine, as a marker of cellular oxidative stress during carcinogenesis. Mutat Res 1997; 387: 146–63

    Google Scholar 

  25. Wang D, Kreutzer DA, Essigmann JM. Mutagenicity and repair of oxidative DNA damage: insights from studies using defined lesions. Mutat Res 1998; 400: 99–115

    PubMed  CAS  Google Scholar 

  26. Arashidani K, Iwamoto-Tanaka N, Muraoka M, Kasai H. Genotoxicity of ribo- and deoxyribonucleosides of 8-hydroxyguanine, 5-hydroxycytosine, and 2-hydroxyadenine: induction of SCE in human lymphocytes and mutagenicity in Salmonella typhimurium TA100. Mutat Res 1998; 403: 223–7

    PubMed  CAS  Google Scholar 

  27. Fujikawa K, Kamiya H, Kasai H. The mutations induced by oxidatively damaged nucleotides, 5-formyl-dUTP and 5-hydroxy-dCTP, in Escherichia coli. Nucleic Acids Res 1998; 26: 4582–87

    PubMed  CAS  Google Scholar 

  28. Dean RT, Fu S, Stocker R, et al. Biochemistry and pathology of radical-mediated protein oxidation. Biochem J 1997; 324: 1–18

    PubMed  CAS  Google Scholar 

  29. Esterbauer H, Schaur RJ, Zollner H. Chemistry and biochemistry of 4-hydroxynonenal, malonaldehyde and related aldehydes. Free Radic Biol Med 1991; 11: 81–128

    PubMed  CAS  Google Scholar 

  30. Stokes AH, Hastings TG, Vrana KE. Cytotoxic and genotoxic potential of dopamine. J Neurosci Res 1999; 55: 659–65

    PubMed  CAS  Google Scholar 

  31. Graham DG, Tiffany SM, Bell Jr WR, et al. Autoxidation versus covalent binding of quinines as the mechanism of toxicity of dopamine, 6-hydroxydopamine, and related compounds toward C1300 neuroblastoma cells in vitro. Mol Pharmacol 1978; 14: 644–53

    PubMed  CAS  Google Scholar 

  32. Halliwell B. Manganese ions, oxidation reactions and the superoxide radical. Neurotoxicology 1984; 5: 113–7

    PubMed  CAS  Google Scholar 

  33. Bredt DS. Endogenous nitric oxide synthesis: biological functions and pathophysiology. Free Radic Res 1999; 31: 577–96

    PubMed  CAS  Google Scholar 

  34. Forfia PR, Hintze TH, Wolin MS, et al. Role of nitric oxide in the control of mitochondrial function. Adv Exp Med Biol 1999; 471: 381–8

    PubMed  CAS  Google Scholar 

  35. Beckman JS, Koppenol WH. Nitric oxide, superoxide, and peroxynitrite: the good, the bad, and ugly. Am J Physiol 1996; 271: C1424–37

    PubMed  CAS  Google Scholar 

  36. Halliwell B, Zhao K, Whiteman M. Nitric oxide and peroxynitrite: the ugly, the uglier and the not so good. Free Radic Res 1999; 31: 651–69

    PubMed  CAS  Google Scholar 

  37. Ischiropoulos H. Biological tyrosine nitration: a pathophysiological function of nitric oxide and reactive oxygen species. Arch Biochem Biophys 1998; 356: 1–11

    PubMed  CAS  Google Scholar 

  38. Greenacre SAB, Ischiropoulos H. Tyrosine nitration: localization, quantification, consequences for protein function and signal transduction. Free Radic Res 2001; 34: 541–81

    PubMed  CAS  Google Scholar 

  39. Gutteridge JM, Smith A. Antioxidant protection by haemopexin of haem-stimulated lipid peroxidation. Biochem J 1998; 256: 861–5

    Google Scholar 

  40. Alayash AI. Hemoglobin-based blood substitutes and the hazards of blood radicals. Free Rad Res 2000; 33: 341–8

    CAS  Google Scholar 

  41. Macdonald RL, Weir BK. A review of hemoglobin and the pathogenesis of cerebral vasospasm. Stroke 1991; 22: 971–82

    PubMed  CAS  Google Scholar 

  42. Evans PJ, Akanmu D, Halliwell B. Promotion of oxidative damage to arachidonic acid and alpha 1-antiproteinase by anti-inflammatory drugs in the presence of the haem proteins myoglobin and cytochrome c. Biochem Pharmacol 1994; 48: 2173–9

    PubMed  CAS  Google Scholar 

  43. Prasad MR, Engelman RM, Jones RM, et al. Effects of oxyradicals on oxymyoglobin: deoxygenation, haem removal and iron release. Biochem J 1989; 263: 731–6

    PubMed  CAS  Google Scholar 

  44. Matz P, Turner C, Weinstein PR, et al. Heme-oxygenase-1 induction in glia throughout rat brain following experimental subarachnoid hemorrhage. Brain Res 1996; 713: 211–22

    PubMed  CAS  Google Scholar 

  45. Lamb NJ, Quinlan GJ, Mumby S, et al. Haem oxygenase shows pro-oxidant activity in microsomal and cellular systems: implications for the release of low-molecular-mass iron. Biochem J 1999; 344: 153–8

    PubMed  CAS  Google Scholar 

  46. Lebovitz RM, Zhang H, Vogel H, et al. Neurodegeneration, myocardial injury, and perinatal death in mitochondrial superoxide dismutase-deficient mice. Proc Natl Acad Sci USA 1996; 93: 9782–7

    PubMed  CAS  Google Scholar 

  47. Melov S, Coskun P, Patel M, et al. Mitochondrial disease in superoxide dismutase 2 mutant mice. Proc Natl Acad Sci USA 1999; 96: 846–51

    PubMed  CAS  Google Scholar 

  48. Li Y, Huang TT, Carlson EJ, et al. Dilated cardiomyopathy and neonatal lethality in mutant mice lacking manganese superoxide dismutase. Nature Genet 1995; 11: 376–81

    PubMed  CAS  Google Scholar 

  49. Melov S, Schneider JA, Day BJ, et al. A novel neurological phenotype in mice lacking mitochondrial manganese superoxide dismutase. Nature Genet 1998; 18: 159–63

    PubMed  CAS  Google Scholar 

  50. Reaume AG, Elliott JL, Hoffman EK, et al. Motor neurons in Cu/Zn superoxide dismutase-deficient mice develop normally but exhibit enhanced cell death after axonal injury. Nature Genet 1996; 13: 43–7

    PubMed  CAS  Google Scholar 

  51. Huang TT, Carlson EJ, Raineri I, et al. The use of transgenic and mutant mice to study oxygen free radical metabolism. Ann NY Acad Sci 1999; 893: 95–112

    PubMed  CAS  Google Scholar 

  52. Matzuk MM, Dionne L, Guo Q, et al. Ovarian function in superoxide dismutase 1 and 2 knockout mice. Endocrinology 1998; 139: 4008–11

    PubMed  CAS  Google Scholar 

  53. Oury TD, Day BJ, Crapo JD. Extracellular superoxide dismutase: a regulator of nitric oxide bioavailability. Lab Invest 1996; 75: 617–36

    PubMed  CAS  Google Scholar 

  54. Carlsson LM, Jonsson J, Edlund T, et al. Mice lacking extracellular superoxide dismutase are more sensitive to hyperoxia. Proc Natl Acad Sci USA 1995; 92: 6264–8

    PubMed  CAS  Google Scholar 

  55. Folz RJ, Abushamaa AM, Suliman HB. Extracellular superoxide dismutase in the airways of transgenic mice reduces inflammation and attenuates lung toxicity following hyperoxia. J Clin Invest 1999; 103: 1055–66

    PubMed  CAS  Google Scholar 

  56. Sheng H, Brady TC, Pearlstein RD, et al. Extracellular superoxide dismutase deficiency worsens outcome from focal cerebral ischaemia in the mouse. Neurosci Lett 1999; 267: 13–6

    PubMed  CAS  Google Scholar 

  57. Thiels E, Urban NN, Gonzalez-Burgos, et al. Impairment of long-term potentiation and associated memory in mice that overexpress extracellular superoxide dismutase. J Neurosci 2000; 20: 7631–9

    PubMed  CAS  Google Scholar 

  58. Chae HZ, Kang SW, Rhee SG. Isoforms of mammalian peroxiredoxin that reduce peroxides in presence of thioredoxin. Methods Enzymol 1999; 300: 219–26

    PubMed  CAS  Google Scholar 

  59. Mustacich D, Powis G. Thioredoxin reductase. Biochem J 2000; 346: 1–8

    PubMed  CAS  Google Scholar 

  60. Nakamura H, Nakamura K, Yodoi J. Redox regulation of cellular activation. Annu Rev Immunol 1997; 15: 351–69

    PubMed  CAS  Google Scholar 

  61. Lash LH, Visarius TM, Sall JM, et al. Cellular and subcellular heterogeneity of glutathione metabolism and transport in rat kidney cells. Toxicology 1998; 130: 1–15

    PubMed  CAS  Google Scholar 

  62. Esposito LA, Kokoszka JE, Waymire KG, et al. Mitochondrial oxidative stress in mice lacking the glutathione peroxidase-1 gene. Free Radic Biol Med 2000; 28: 754–66

    PubMed  CAS  Google Scholar 

  63. Kang SW, Chae HZ, Seo MS, et al. Mammalian peroxiredoxin isoforms can reduce hydrogen peroxide generated in response to growth factors and tumor necrosis factor-alpha. J Biol Chem 1998; 273: 6297–302

    PubMed  CAS  Google Scholar 

  64. Chen Z, Putt DA, Lash LH. Enrichment and functional reconstitution of glutathione transport activity from rabbit kidney mitochondria: further evidence for the role of the dicarboxylate and 2-oxoglutarate carriers in mitochondrial glutathione transport. Arch Biochem Biophys 2000; 373: 193–202

    PubMed  CAS  Google Scholar 

  65. Halliwell B, Gutteridge JM. The antioxidants of human extracellular fluids. Arch Biochem Biophys 1990; 280: 1–8

    PubMed  CAS  Google Scholar 

  66. Evans PJ, Bomford A, Halliwell B. Non-caeruloplasmin copper and ferroxidase activity in mammalian serum: ferroxidase activity and phenanthroline-detectable copper in human serum in Wilson’s disease. Free Radic Res Commun 1989; 7: 55–62

    PubMed  CAS  Google Scholar 

  67. Chasteen ND, Harrison PM. Mineralization in ferritin: an efficient means of iron storage. J Struct Biol 1999; 126: 182–94

    PubMed  CAS  Google Scholar 

  68. Dameron CT, Harrison MD. Mechanisms for protection against copper toxicity. Am J Clin Nutr 1998; 67: 1091S–7S

    PubMed  CAS  Google Scholar 

  69. Breuer W, Epsztejn S, Cabantchik ZI. Dynamics of the cytosolic chelatable iron pool of K562 cells. FEBS Lett 1996; 382: 304–8

    PubMed  CAS  Google Scholar 

  70. Konijn AM, Glickstein H, Vaisman B, et al. The cellular labile iron pool and intracellular ferritin in K562 cells. Blood, 1999; 94: 2128–34

    PubMed  CAS  Google Scholar 

  71. Petrat F, Rauen U, de Groot H. Determination of the chelatable iron pool of isolated rat hepatocytes by digital fluorescence microscopy using the fluorescent probe, phen green SK. Hepatology 1999; 29: 1171–9

    PubMed  CAS  Google Scholar 

  72. Gutteridge JM, Cao W, Chevion M. Bleomycin-detectable iron in brain tissue. Free Radic Res Commun 1991; 11: 317–20

    PubMed  CAS  Google Scholar 

  73. Spencer JP, Jenner A, Aruoma OI, et al. Intense oxidative DNA damage promoted by L-dopa and its metabolites: implications for neurodegenerative disease. FEBS Lett 1994; 353: 246–50

    PubMed  CAS  Google Scholar 

  74. Stocks J, Gutteridge JM, Sharp RJ, et al. Assay using brain homogenate for measuring the antioxidant activity of biological fluids. Clin Sci Mol Med 1974; 47: 215–22

    PubMed  CAS  Google Scholar 

  75. Zaleska, MM, Floyd, RA. Regional lipid peroxidation in rat brain in vitro: possible role of endogenous iron. Neurochem Res 1985; 10: 397–410

    PubMed  CAS  Google Scholar 

  76. Traber MG. Biokinetics of vitamin E. In: Cadenas E, Packer L, editors. Handbook of antioxidants. New York (NY): Marcel Dekker Inc, 1996: 43–61

    Google Scholar 

  77. Vatassery GT, Brin MF, Fahn S, et al. Effect of high doses of dietary vitamin E on the concentrations of vitamin E in several brain regions, plasma, liver, and adipose tissue of rats. J Neurochem 1988; 51: 621–3

    PubMed  CAS  Google Scholar 

  78. Kagan VE, Tyurina YY, Witt E. Role of coenzyme Q and superoxide in vitamin E cycling. Sub-Cell Biochem 1998; 30: 491–507

    CAS  Google Scholar 

  79. Krinsky NI. The antioxidant and biological properties of the carotenoids. Ann NY Acad Sci 1998; 854: 443–7

    PubMed  CAS  Google Scholar 

  80. Giovannucci E. Tomatoes, tomato-based products, lycopene, and cancer: review of the epidemiologic literature. J Natl Cancer Inst 1999; 91: 317–31

    PubMed  CAS  Google Scholar 

  81. Hammond Jr BR, Wooten BR, Snodderly DM. Density of the human crystalline lens is related to the macular pigment carotenoids, lutein and zeaxanthin. Optom Vis Sci 1997; 74: 499–504

    PubMed  Google Scholar 

  82. Di Mascio P, Kaiser S, Sies H. Lycopene as the most efficient biological carotenoids singlet oxygen quencher. Arch Biochem Biophys 1989; 274: 532–8

    PubMed  Google Scholar 

  83. Berneburg M, Grether-Beck S, Kurten V, et al. Singlet oxygen mediates the UVA-induced generation of the photoaging-associated mitochondrial common deletion. J Biol Chem 1999; 274: 15345–9

    PubMed  CAS  Google Scholar 

  84. Mortensen A, Skibsted LH, Sampson J, et al. Comparative mechanisms and rates of free radical scavenging by carotenoid antioxidants. FEBS Lett 1997; 418: 91–7

    PubMed  CAS  Google Scholar 

  85. Rice-Evans C, editor. Wake up to flavonoids. London: Royal Society of Medicine Press, 2000

    Google Scholar 

  86. Halliwell B, Zhao K, Whiteman M. The gastrointestinal tract: a major site of antioxidant action? Free Radic Res 2000; 33: 819–30

    PubMed  CAS  Google Scholar 

  87. Halliwell B. Can oxidative DNA damage be used as a biomarker of cancer risk in humans? Problems, resolutions and preliminary results from nutritional supplementation studies. Free Radic Res 1998; 29: 469–86

    PubMed  CAS  Google Scholar 

  88. Young JF, Nielsen SE, Haraldsdottir J, et al. Polyphenolic antioxidants in fruit juice; urinary excretion and effects on biological markers for antioxidative status. Ugeskrift for Laeger 2000; 162: 1388–92

    PubMed  CAS  Google Scholar 

  89. Morrow JD, Roberts LJ. The isoprostanes: unique bioactive products of lipid peroxidation. Prog Lipid Res 1997; 36: 1–21

    PubMed  CAS  Google Scholar 

  90. Lawson JA, Rokach J, FitzGerald GA. Isoprostanes: formation, analysis and use as indices of lipid peroxidation in vivo. J Biol Chem 1999; 247: 24441–4

    Google Scholar 

  91. Pegg AE. DNA repair pathways and cancer prevention. Adv Exp Med Biol 1999; 472: 253–67

    PubMed  CAS  Google Scholar 

  92. Grune T, Reinheckel T, Davies KJ. Degradation of oxidized proteins in mammalian cells. FASEB J 1997; 11: 526–34

    PubMed  CAS  Google Scholar 

  93. Brigelius-Flohe R. Tissue-specific functions of individual glutathione peroxidases. Free Radic Biol Med 1999; 27: 951–65

    PubMed  CAS  Google Scholar 

  94. Chen QM, Bartholomew JC, Campisi J, et al. Molecular analysis of H2O2-induced senescent-growth arrest in normal human fibroblasts: p53 and Rb control G1 arrest but not cell replication. Biochem J 1998; 332: 43–50

    PubMed  CAS  Google Scholar 

  95. Halliwell B. Antioxidant defence mechanisms: from the beginning to the end (of the beginning). Free Radic Res 1999; 31: 261–72

    PubMed  CAS  Google Scholar 

  96. Roxborough HE, Burton GW, Kelly FJ. Inter- and intra-individual variation in plasma and red blood cell vitamin E after supplementation. Free Radic Res 2000; 33: 437–45

    PubMed  CAS  Google Scholar 

  97. Hulla JE, Miller MS, Taylor JA, et al. Symposium overview: the role of genetic polymorphism and repair deficiencies in environmental disease. Toxicol Sci 1999; 47: 135–43

    PubMed  CAS  Google Scholar 

  98. Fan F, Liu C, Tavare S, et al. Polymorphisms in the human DNA repair gene XPF. Mutat Res 1999; 406: 115–20

    PubMed  CAS  Google Scholar 

  99. Hayes JD, McLellan LI. Glutathione and glutathione-dependent enzymes represent a co-ordinately regulated defence against oxidative stress. Free Radic Res 1999; 31: 273–300

    PubMed  CAS  Google Scholar 

  100. Forsberg L, de Faive U, Morgenstern R. Oxidative stress, human genetic variation, and disease. Arch Biochem Biophys 2001; 389: 84–93

    PubMed  CAS  Google Scholar 

  101. England T, Beatty E, Rehman A, et al. The steady-state levels of oxidative DNA damage and of lipid peroxidation F2-isoprostanes are not correlated in healthy human subjects. Free Radic Res 1999; 32: 355–62

    Google Scholar 

  102. Orr WC, Sohal RS. Extension of life-span by overexpression of superoxide dismutase and catalase in Drosophila melanogaster. Science 1994; 263: 1128–30

    PubMed  CAS  Google Scholar 

  103. Honda Y, Honda S. The daf-2 gene network for longevity regulates oxidative stress resistance and Mn-superoxide dismutase gene expression in Caenorhabditis elegans. FASEB J 1999; 13: 1385–93

    PubMed  CAS  Google Scholar 

  104. Vanfleteren JR, De Vreese A. The gerontogenes age-1 and daf-2 determine metabolic rate potential in aging Caenorhabditis elegans. FASEB J 1995; 9: 1355–61

    PubMed  CAS  Google Scholar 

  105. Larsen PL. Aging and resistance to oxidative damage in Caenorhabditis elegans. Proc Natl Acad Sci USA 1993; 90: 8905–9

    PubMed  CAS  Google Scholar 

  106. Melov S, Ravenscroft J, Maletr S, et al. Extension of life-span with superoxide dismutase/catalase mimetics. Science 2000; 289: 1567–9

    PubMed  CAS  Google Scholar 

  107. Sohal RS, Weindruch R. Oxidative stress, caloric restriction, and aging. Science 1996; 273: 59–63

    PubMed  CAS  Google Scholar 

  108. Leeuwenburgh C, Wagner P, Holloszy JO, et al. Caloric restriction attenuates dityrosine cross-linking of cardiac and skeletal muscle proteins in aging mice. Arch Biochem Biophys 1997; 346: 74–80

    PubMed  CAS  Google Scholar 

  109. Campisi J. Aging, chromatin and food restriction: connecting the dots. Science 2000; 289: 2062–3

    PubMed  CAS  Google Scholar 

  110. Dubey A, Forster MJ, Lal H, et al. Effect of age and caloric intake on protein oxidation in different brain regions and on behavioral functions of the mouse. Arch Biochem Biophys 1996; 333: 189–97

    PubMed  CAS  Google Scholar 

  111. Sastre J, Pallardo FV, Garcia de la Asuncion J, et al. Mitochondria, oxidative stress and aging. Free Radic Res 2000; 32: 189–98

    PubMed  CAS  Google Scholar 

  112. Atamna H, Cheung I, Ames BN. A method for detecting abasic sites in living cells: age-dependent changes in base excision repair. Proc Natl Acad Sci USA 2000; 97: 686–91

    PubMed  CAS  Google Scholar 

  113. Rao KS. DNA-damage and DNA-repair in aging brain. Indian J Med Res 1997; 106: 423–37

    PubMed  CAS  Google Scholar 

  114. Michikawa Y, Mazzucchelli F, Bresolin N, et al. Aging-dependent large accumulation of point mutations in the human mtDNA control region for replication. Science 1999; 286: 774–9

    PubMed  CAS  Google Scholar 

  115. Ponnappan U, Zhong M, Trebilcock GU. Decreased proteasome-mediated degradation in T cells from the elderly: a role in immune senescence. Cell Immunol 1999; 192: 167–74

    PubMed  CAS  Google Scholar 

  116. Keller JN, Hanni KB, Markesbery WR. Possible involvement of proteasome inhibition in aging: implications for oxidative stress. Mech Aging Dev 2000; 113: 61–70

    PubMed  CAS  Google Scholar 

  117. Hayashi T, Goto S. Age-related changes in the 20S and 26S proteasome activities in the liver of male F344 rats. Mech Aging Dev 1998; 102: 55–66

    PubMed  CAS  Google Scholar 

  118. Mecocci P, Fano G, Fulle S, et al. Age-dependent increases in oxidative damage to DNA, lipids, and proteins in human skeletal muscle. Free Radic Biol Med 1999; 26: 303–8

    PubMed  CAS  Google Scholar 

  119. Halliwell B. Proteasomal dysfunction: a primary event in neurodegeneration that leads to nitrative and oxidative stress and subsequent cell death. Ann N Y Acad Sci. In press

  120. Forster MJ, Dubey A, Dawson KM, et al. Age-related losses of cognitive function and motor skills in mice are associated with oxidative protein damage in the brain. Proc Natl Acad Sci USA 1996; 93: 4765–9

    PubMed  CAS  Google Scholar 

  121. Smith CD, Carney JM, Starke-Reed Pe, et al. Excess brain protein oxidation and enzyme dysfunction in normal aging and in Alzheimer disease. Proc Natl Acad Sci USA 1991; 88: 10540–3

    PubMed  CAS  Google Scholar 

  122. Lee HC, Lim ML, Lu CY, et al. Concurrent increase of oxidative DNA damage and lipid peroxidation together with mitochondrial DNA mutation in human lung tissues during aging: smoking enhances oxidative stress on the aged tissues. Arch Biochem Biophys 1999; 362: 309–16

    PubMed  CAS  Google Scholar 

  123. Kaneko T, Tahara S, Matsuo M. Retarding effect of dietary restriction on the accumulation of 8-hydroxy-2′-deoxyguanosine in organs of Fisher 344 rats during aging. Free Radic Biol Med 1997; 23: 76–81

    PubMed  CAS  Google Scholar 

  124. Kramer PJ, Caldwell J, Hofmann A, et al. Neurotoxicity risk assessment of MPTP (N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) as a synthetic impurity of drugs. Hum Exp Toxicol 1998; 17: 283–93

    PubMed  CAS  Google Scholar 

  125. Yamamoto H, Tang HW. Antagonistic effect of melatonin against cyanide-induced seizures and acute lethality in mice. Toxicol Lett 1996; 87: 19–24

    PubMed  CAS  Google Scholar 

  126. Dautry C, Conde F, Brouillet E, et al. Serial 1H-NMR spectroscopy study of metabolic impairment in primates chronically treated with the succinate dehydrogenase inhibitor 3-nitropropionic acid. Neurobiol Disease 1999; 6: 259–68

    CAS  Google Scholar 

  127. Betarbet R, Sherer TB, Mackenzie G, et al. Chronic systemic pesticide exposure reproduces features of Parkinson’s disease. Nature Neurosci 2000; 3: 1301–6

    PubMed  CAS  Google Scholar 

  128. Schulz JB, Mathews RT, Klockgether T, et al. The role of mitochondrial dysfunction and neuronal nitric oxide in animal models of neurodegenerative diseases. Mol Cell Biochem 1997; 174: 193–7

    PubMed  CAS  Google Scholar 

  129. Schulz JB, Huang PL, Matthews RT, et al. Striatal malonate lesions are attenuated in neuronal nitric oxide synthase knockout mice. J Neurochem 1996; 67: 430–3

    PubMed  CAS  Google Scholar 

  130. Molina JA, Jimenez-Jimenex FJ, Orti-Pareja M, et al. The role of nitric oxide in neurodegeneration: potential for pharmacological intervention. Drugs Aging 1998; 12: 251–9

    PubMed  CAS  Google Scholar 

  131. Mark RJ. Common mechanisms of oxidative damage in chronic neurodegenerative conditions: potential points of therapeutic intervention. Exp Opin Ther Patents 1999; 9: 1339–46

    CAS  Google Scholar 

  132. Markesbery WR, Carney JM. Oxidative alterations in Alzheimer’s disease. Brain Pathol 1999; 9: 133–46

    PubMed  CAS  Google Scholar 

  133. Spencer JP, Jenner P, Daniel SE, et al. Conjugates of catecholamines with cysteine and GSH in Parkinson’s disease: possible mechanisms of formation involving reactive oxygen species. J Neurochem 1998; 71: 2112–22

    PubMed  CAS  Google Scholar 

  134. Anden NE, Hfuxe K, Hamberger B, et al. A quantitative study on the nigro-neostriatal dopamine neuron system in the rat. Acta Physiol Scandinavica 1966; 67: 306–12

    CAS  Google Scholar 

  135. Spina MB, Cohen G. Exposure of striatal synaptosomes to L-dopa increases levels of oxidized glutathione. J Pharmacol Exp Ther 1989; 248: 478: 502–7

    Google Scholar 

  136. Cheepsunthorn P, Palmer C, Connor JR. Cellular distribution of ferritin subunits in postnatal rat brain. J Compar Neurol 1998; 400: 73–86

    CAS  Google Scholar 

  137. Hulet SW, Powers S, Connor JR. Distribution of transferrin and ferritin binding in normal and multiple sclerotic human brains. J Neurol Sci 1999; 165: 48–55

    PubMed  CAS  Google Scholar 

  138. Focht SJ, Synder BS, Beard JL, et al. Regional distribution of iron, transferrin, ferritin and oxidatively-modified proteins in young and aged Fischer 344 rat brains. Neuroscience 1997; 79: 255–61

    PubMed  CAS  Google Scholar 

  139. Kabuto H, Yokoi I, Habu H, et al. Reduction in nitric synthase activity with development of an epileptogenic focus induced by ferric chloride in the rat brain. Epilepsy Res 1996; 25: 65–8

    PubMed  CAS  Google Scholar 

  140. Gutteridge JM. Iron and oxygen radicals in brain. Ann Neurol 1992; 32 Suppl: S16–21

    PubMed  CAS  Google Scholar 

  141. Moos T, Morgan EH. Evidence for low molecular weight, non-transferrin-bound iron in rat brain and cerebrospinal fluid. J Neurosci Res 1998; 54: 486–94

    PubMed  CAS  Google Scholar 

  142. Pinero DJ, Li NQ, Connor JR, et al. Variations in dietary iron alter brain iron metabolism in developing rats. J Nutr 2000; 130: 254–63

    PubMed  CAS  Google Scholar 

  143. Moos T, Morgan EH. Transferrin and transferrin receptor function in brain barrier systems. Cell Mol Neurobiol 2000; 20: 77–95

    PubMed  CAS  Google Scholar 

  144. Nourooz-Zadeh J, Liu EH, Yhlen B, et al. F4-isoprostanes as specific marker of docosahexaenoic acid peroxidation in Alzheimer’s disease. J Neurochem 1999; 72: 734–40

    PubMed  CAS  Google Scholar 

  145. Mark RJ, Lovell MA, Markesbery WR, et al. A role for 4-hydroxynonenal, an aldehyde product of lipid peroxidation, in disruption of ion homeostasis and neuronal death induced by amyloid beta-peptide. J Neurochem 1997; 68: 225–64

    Google Scholar 

  146. Kruman I, Bruce-Keller AJ, Bredesen, et al. Evidence that 4-hydroxynonenal mediates oxidative stress-induced neuronal apoptosis. J Neurosci 1997; 17: 5089–100

    PubMed  CAS  Google Scholar 

  147. Bruce-Keller AJ, Li YJ, Lovell MA, et al. 4-Hydroxynonenal, a product of lipid peroxidation, damages cholinergic neurons and impairs visuospatial mempory in rats. J Neuropathol Exp Neurol 1998; 57: 257–67

    PubMed  CAS  Google Scholar 

  148. Montine KS, Reich E, Neely MD, et al. Distribution of reducible 4-hydroxynonenal adduct immunoreactivity in Alzheimer disease is associated with APOE genotype. J Neuropathol Exp Neurol 1998; 57: 415–25

    PubMed  CAS  Google Scholar 

  149. Smith RG, Henry YK, Mattson MP, et al. Presence of 4-hydroxynonenal in cerebrospinal fluid of patients with sporadic amyotrophic lateral sclerosis. Ann Neurol 1998; 44: 696–9

    PubMed  CAS  Google Scholar 

  150. Keller JN, Mattson MP. Roles of lipid peroxidation in modulation of cellular signaling pathways, cell dysfunction, and death in the nervous system. Rev Neurosci 1998; 9: 105–16

    PubMed  CAS  Google Scholar 

  151. Sayre LM, Zelasko DA, Harris PL, et al. 4-Hydroxynonenal-derived advanced lipid peroxidation end products are increased in Alzheimer’s disease. J Neurochem 1997; 68: 2092–7

    PubMed  CAS  Google Scholar 

  152. Ong WY, Lu XR, Hu CY, et al. Distribution of hydroxynonenal-modified proteins in the kainate-lesioned rat hippocampus: evidence that hydroxynonenal formation precedes neuronal cell death. Free Radic Biol Med 2000; 28: 1214–21

    PubMed  CAS  Google Scholar 

  153. Sheu KF, Blass JP. The alpha-ketoglutarate dehydrogenase complex. Ann NY Acad Sci 1999; 893: 61–78

    PubMed  CAS  Google Scholar 

  154. Spina MB, Cohen G. Dopamine turnover and glutathione oxidation: implications for Parkinson disease. Proc Natl Acad Sci USA 1989; 86: 1398–400

    PubMed  CAS  Google Scholar 

  155. Cohen G, Spina MB. Deprenyl suppresses the oxidant stress associated with increased dopamine turnover. Ann Neurol 1989; 26: 689–90

    PubMed  CAS  Google Scholar 

  156. Shih JC, Chen K, Ridd MJ. Role of MAO A and B in neurotransmitter metabolism and behavior. Polish J Pharmacol 1999; 51: 25–9

    CAS  Google Scholar 

  157. Aksenov MY, Aksenova MV, Carney JM, et al. Oxidative modification of glutamine synthetase by amyloid beta peptide. Free Radic Res 1997; 27: 267–81

    PubMed  CAS  Google Scholar 

  158. Brannan TS, Maker HS, Raes IP. Regional distribution of catalase in the adult rat brain. J Neurochem 1981; 36: 307–9

    PubMed  CAS  Google Scholar 

  159. McKenna O, Arnold G, Holtzman E. Microperoxisome distribution in the central nervous system of the rat. Brain Res 1976; 117: 191–4

    Google Scholar 

  160. Sinet PM, Heikkila RE, Cohen G. Hydrogen peroxide production by rat brain in vivo. J Neurochem 1980; 34: 1421–8

    PubMed  CAS  Google Scholar 

  161. Boje KM, Arora PK. Microglial-produced nitric oxide and reactive nitrogen oxides mediate neuronal cell death. Brain Res 1992; 587: 250–6

    PubMed  CAS  Google Scholar 

  162. Colton C, Wilt S, Gilbert D, et al. Species differences in the generation of reactive oxygen species by microglia. Mol Chem Neuropathol 1996; 28: 15–20

    PubMed  CAS  Google Scholar 

  163. Uysal G, Yuksel G, Sinav B, et al. Cerebrospinal fluid nitric oxide levels in childhood bacterial meningitis. Scand J Infect Dis 1999; 31: 518–20

    PubMed  CAS  Google Scholar 

  164. Svenningsson A, Petersson AS, Andersen O, et al. Nitric oxide metabolites in CSF of patients with MS are related to clinical disease course. Neurology 1999; 53: 1880–2

    PubMed  CAS  Google Scholar 

  165. Ravindranath V. Metabolism of xenobiotics in the central nervous system: implications and challenges. Biochem Pharmacol 1998; 56: 547–51

    PubMed  CAS  Google Scholar 

  166. Hansson T, Tindberg N, Ingelman-Sundberg M, et al. Regional distribution of ethanol-inducible cytochrome P450 IIE 1 in the rat central nervous system. Neuroscience 1990; 34: 451–63

    PubMed  CAS  Google Scholar 

  167. Upadhya SC, Tirumalai PS, Boyd MR, et al. Cytochrome P4502E (CYP2E) in brain: constitutive expression, induction by ethanol and localization by fluorescence in situ hybridization. Arch Biochem Biophys 2000; 373: 23–34

    PubMed  CAS  Google Scholar 

  168. Andrews AM, Ladenheim B, Epstein CJ, et al. Transgenic mice with high levels of superoxide dismutase activity are protected from the neurotoxic effects of 2′-NH2-MPTP on serotonergic and noradrenergic nerve terminals. Mol Pharmacol 1996; 50: 1511–9

    PubMed  CAS  Google Scholar 

  169. Mikawa S, Kinouchi H, Kamii H, et al. Attenuation of acute and chronic damage following traumatic brain injury in copper, zinc-superoxide dismutase transgenic mice. J Neurosurgery 1996; 85: 885–91

    CAS  Google Scholar 

  170. Chan PH, Kawase M, Murakami K, et al. Overexpression of SOD1 in transgenic rats protects vulnerable neurons against ischemic damage after global cerebral ischaemia and reperfusion. J Neurosci 1998; 18: 8292–9

    PubMed  CAS  Google Scholar 

  171. Jayanthi S, Ladenheim B, Andrews AM, et al. Overexpression of human copper/zinc superoxide dismutase in transgenic mice attenuates oxidative stress caused by methylenedioxy-methamphetamine (Ecstasy). Neuroscience 1999; 91: 1379–87

    PubMed  CAS  Google Scholar 

  172. Nakao N, Frodl EM, Widner H, et al. Overexpressing Cu/Zn superoxidase dismutase enhances survival of transplanted neurons in a rat model of Parkinson’s disease. Nature Med 1995; 1: 226–31

    PubMed  CAS  Google Scholar 

  173. Lee MH, Hyun DH, Jenner P, et al. Effect of overexpression of wild-type and mutant CuZnSODs on oxidative stress and cell-death induced by H2O2, 4-HNE or serum deprivation: potentiation of injury by ALS-related mutant SODs and protection by bcl-2. J Neurochem 2001; 78: 209–20

    PubMed  CAS  Google Scholar 

  174. Troy CM, Shelanski ML. Down-regulation of Cu/Zn-superoxide dismutase causes apoptotic death in PC12 neuronal cells. Proc Natl Acad Sci USA 1994; 91: 6384–7

    PubMed  CAS  Google Scholar 

  175. Jain A, Martensson J, Stole E, et al. Glutathione deficiency leads to mitochondrial damage in brain. Proc Natl Acad Sci USA 1991; 88: 1913–7

    PubMed  CAS  Google Scholar 

  176. Weisbrot-Lefkowitz M, Reuhl K, Perry B, et al. Overexpression of human glutathione peroxidase protects transgenic mice against focal cerebral ischaemia/reperfusion damage. Brain Res 1998; 53: 333–8

    CAS  Google Scholar 

  177. Klivenyi P, Andreassen OA, Ferrante RJ, et al. Mice deficient in cellular glutathione peroxidase show increased vulnerability to malonate 3-nitropropionic acid, and MPTP. J Neurosci 2000; 20: 1–7

    PubMed  CAS  Google Scholar 

  178. Sarafian TA, Verity MA, Vinters HV, et al. Differential expression of peroxiredoxin subtypes in human brain cell types. J Neurosci Res 1999; 56: 206–12

    PubMed  CAS  Google Scholar 

  179. Takagi Y, Mitsui A, Nishiyama A, et al. Overexpression of thioredoxin in transgenic mice attenuates focal ischemic brain damage. Proc Natl Acad Sci USA 1999; 96: 4131–6

    PubMed  CAS  Google Scholar 

  180. Ceballos-Picot I, Nicole A, Sinet PM. Cellular clones and transgenic mice overexpressing copper-zinc superoxide dismutase: models for the study of free radical metabolism and aging. EXS 1992; 62: 89–98

    PubMed  CAS  Google Scholar 

  181. Kedziora J, Bartosz G. Down’s syndrome: a pathology involving the lack of balance of reactive oxygen species. Free Radic Biol Med 1988; 4: 317–30

    PubMed  CAS  Google Scholar 

  182. Groner Y, Elroy-Stein O, Avraham KB, et al. Cell damage by excess CuZnSOD and Down’s syndrome. Biomed Pharmacother 1994; 48: 231–40

    PubMed  CAS  Google Scholar 

  183. Lee MH, Hyun DH, Jenner P, et al. Effect of overexpression of wild-type and mutant CuZnSODs on oxidative damage and antioxidant defences: relevance to Down’s syndrome and familial ALS. J Neurochem 2001; 76: 957–65

    PubMed  CAS  Google Scholar 

  184. Levin ED, Brady TC, Hochrein EC, et al. Molecular manipulations of extracellular superoxide dismutase: functional importance for learning. Behavior Genet 1998; 28: 381–90

    CAS  Google Scholar 

  185. Dringen R, Gutterer JM, Hirrlinger J. Glutathione metabolism in brain. Eur J Biochem 2000; 267: 4912–6

    PubMed  CAS  Google Scholar 

  186. Dringen R, Kussmaul L, Gutterer JM, et al. The glutathione system of peroxide detoxification is less efficient in neurons than in astroglial cells. J Neurochem 1999; 72: 2523–30

    PubMed  CAS  Google Scholar 

  187. Ong WY, Hu CY, Hjelle OP, et al. Changes in glutathione in the hippocampus of rats injected with kainate: depletion in neurons and upregulation in glia. Exp Brain Res 2000; 132: 510–6

    PubMed  CAS  Google Scholar 

  188. Mathisen GA, Fonnum F, Paulsen RE. Contributing mechanisms for cysteine excitotoxicity in cultured cerebellar granule cells. Neurochem Res 1996; 21: 293–8

    PubMed  CAS  Google Scholar 

  189. Puka-Sundvall M, Eriksson P, Nilsson M, et al. Neurotoxicity of cysteine: interaction with glutamate. Brain Res 1995; 705: 65–70

    PubMed  CAS  Google Scholar 

  190. Spector R, Spector AZ, Snodgrass SR. Model for transport in the central nervous system. Am J Physiol 1977; 232: R73–9

    PubMed  CAS  Google Scholar 

  191. Lonnrot K, Metsa-Ketela T, Molnar G, et al. The effect of ascorbate and ubiquinone supplementation on plasma and CSF total antioxidant capacity. Free Radic Biol Med 1996; 21: 211–7

    PubMed  CAS  Google Scholar 

  192. Siushansian R, Wilson JX. Ascorbate transport and intracellular concentration in cerebral astrocytes. J Neurochem 1995; 65: 41–9

    PubMed  CAS  Google Scholar 

  193. Rice-Evans C, Okunade G, Khan R. The suppression of iron release from activated myoglobin by physiological electron donors and by desferrioxamine. Free Radic Res Commun 1989; 7: 45–54

    PubMed  CAS  Google Scholar 

  194. Podda M, Weber C, Traber MG, et al. Simultaneous determination of tissue tocopherols, tocotrienols, ubiquinols, and ubiquinones. J Lipid Res 1996; 37: 893–901

    PubMed  CAS  Google Scholar 

  195. Muller DP, Goss-Sampson MA. Neurochemical, neurophysiological, and neuropathological studies in vitamin E deficiency. Crit Rev Neurobiol 1990; 5: 239–63

    PubMed  CAS  Google Scholar 

  196. Dexter DT, Brooks DJ, Harding AE, et al. Nigrostriatal function in vitamin E deficiency: clinical, experimental, and positron emisson tomographic studies. Ann Neurol 1994; 35: 298–303

    PubMed  CAS  Google Scholar 

  197. Pillai SR, Traber MG, Steiss JE, et al. Alpha-tocopherol concentrations of the nervous system and selected tissues of adult dogs fed three levels of vitamin E. Lipids 1993; 28: 1101–5

    PubMed  CAS  Google Scholar 

  198. Dexter DT, Nanayakkara I, Goss-Sampson MA, et al. Nigral dopaminergic cell loss in vitamin E deficient rats. Neuroreport 1994; 5: 1773–6

    PubMed  CAS  Google Scholar 

  199. Metcalfe T, Bowen DM, Muller DP. Vitamin E concentrations in human brain of patients with Alzheimer’s disease, fetuses with Down’s syndrome, centenarians, and controls. Neurochem Res 1989; 14: 1209–12

    PubMed  CAS  Google Scholar 

  200. Mecocci P, Polidori MC, Trolano L, et al. Plasma antioxidants and longevity: a study on healthy centenarians. Free Radic Biol Med 2000; 28: 1243–8

    PubMed  CAS  Google Scholar 

  201. Fernandez-Calle P, Molina JA, Jimenez-Jimenez FJ, et al. Serum levels of α-tocopherol (vitamin E) in Parkinson’s disease. Neurology 1992; 42: 1064–6

    PubMed  CAS  Google Scholar 

  202. Dexter DT, Ward RJ, Wells FR, et al. Alpha-tocopherol levels in brain are not altered in Parkinson’s disease. Ann Neurol 1992; 32: 591–593

    PubMed  CAS  Google Scholar 

  203. Rötig A, Appelkvist EL, Geromel V, et al. Quinone-responsive multiple respiratory-chain dysfunction due to widespread coenzyme Q10 deficiency. Lancet 2000; 356: 391–5

    PubMed  Google Scholar 

  204. Beal ME. Coenzyme Q10 administration and its potential for treatment of neurodegenerative diseases. Biofactors 1999; 9: 261–6

    PubMed  CAS  Google Scholar 

  205. Ludi R, Hart PE, Rajagopalon B, et al. Antioxidant treatment improves in vivo cardiac and skeletal muscle bioenergetics in patients with Friedrich’s ataxia. Ann Neurol 2001; 49: 590–6

    Google Scholar 

  206. Stahl W, Schwarz W, Sundquist AR, et al. Cis-trans isomers of lycopene and beta-carotene in human serum and tissues. Arch Biochem Biophys 1992; 294: 173–7

    PubMed  CAS  Google Scholar 

  207. Serbinova E, Choo M, Packer L. Distribution and antioxidant activity of a palm oil carotene fraction in rats. Biochem Int 1992; 28: 881–6

    PubMed  CAS  Google Scholar 

  208. Schroder-van der Elst JP, van der Heide D, Rokos H, et al. Synthetic flavonoids cross the placenta in the rat and are found in fetal brain. Am J Physiol 1998; 274: E253–6

    PubMed  CAS  Google Scholar 

  209. Wettstein A. Cholinesterase inhibitors and Gingko extract: are they comparable in the treatment of dementia? Comparison of published placebo-controlled efficacy studies of at least six months’ duration. Phytomedicine 2000; 6: 393–401

    PubMed  CAS  Google Scholar 

  210. Oyama Y, Fuchs PA, Katayama N, et al. Myricetin and quercetin, the flavonoid constituents of Ginkgo biloba extract, greatly reduce oxidative metabolism in both resting and Ca2+loaded brain neurons. Brain Res 1994; 635: 125–9

    PubMed  CAS  Google Scholar 

  211. Watanabe CH, Wolffram S, Ader P, et al. The in vivo neuromodulatory effects of the herbal medicine ginkgo biloba. Proc Natl Acad U S A 2001; 98: 6577–80

    CAS  Google Scholar 

  212. Yan JJ, Cho JY, Kim HS, et al. Protection against β-amyloid peptide toxicity in vivo with long-term administration of ferolic acid. Br J Pharmacol 2001; 133: 89–96

    PubMed  CAS  Google Scholar 

  213. Klomp LW, Gitlin JD. Expression of the ceruloplasmin gene in the human retina and brain: implications for a pathogenic model in aceruloplasminemia. Hum Mol Genet 1996; 5: 1989–96

    PubMed  CAS  Google Scholar 

  214. Yoshida K, Kaneko K, Miyajima H, et al. Increased lipid peroxidation in the brains of aceruloplasminemia patients. J Neurol Sci 2000; 175: 91–5

    PubMed  CAS  Google Scholar 

  215. Elovaara I, Icen A, Palo J, et al. CSF in Alzheimer’s disease: studies on blood-brain barrier function and intrathecal protein synthesis. J Neurol Sci 1985; 70: 73–80

    PubMed  CAS  Google Scholar 

  216. Panter SS, Sadrzadeh SM, Hallaway PE, et al. Hypohaptoglobinemia associated with familial epilepsy. J Exp Med 1985; 161: 748–54

    PubMed  CAS  Google Scholar 

  217. Hidalgo J, Carrasco J. Regulation of the synthesis of brain metallothioneins. Neurotoxicology 1998; 19: 661–6

    PubMed  CAS  Google Scholar 

  218. Penkowa M, Carrasco J, Giralt M, et al. CNS wound healing is severely depressed in metallothionein I- and II-deficient mice. J Neurosci 1999; 19: 2535–45

    PubMed  CAS  Google Scholar 

  219. Ebadi M, Iversen PL, Hao R, et al. Expression and regulation of brain metallothionein. Neurochem Int 1995; 27: 1–22

    PubMed  CAS  Google Scholar 

  220. Huang XD, Cuajungco MP, Atwood CS, et al. Alzheimer’s disease, beta-amyloid proteinand zinc. J Nutr 2000; 130: 1488S–92S

    PubMed  CAS  Google Scholar 

  221. Elbirt KK, Bonkovsky HL. Heme oxygenase: recent advances in understanding its regulation and role. Proc Assoc Am Phys 1999; 111: 438–47

    PubMed  CAS  Google Scholar 

  222. Markesbery WR. Oxidative stress hypothesis in Alzheimer’s disease. Free Radic Biol Med 1997; 23: 134–47

    PubMed  CAS  Google Scholar 

  223. Panathian N, Yoshiura M, Maines MD. Overexpression of heme oxygenase-1 is neuroprotective in a model of permanent middle cerebral artery occlusion in transgenic mice. J Neurochem 1999; 72: 1187–203

    Google Scholar 

  224. Takeda A, Smith MA, Avilá J, et al. In Alzheimer’s disease, heme oxygenase is coincident with Alz50, an epitope of k induced by 4-hydroxy-2-nonenal modification. J Neurochem 2000; 75: 1234–41

    PubMed  CAS  Google Scholar 

  225. Van Bergen P, Rauhala P, Spooner CM, et al. Hemoglobin and iron-evoked oxidative stress in the brain: protection by bile pigments, manganese and S-nitrosoglutathione. Free Radic Res 1999; 31: 631–40

    PubMed  Google Scholar 

  226. Akaishi T, Shiomi T, Sawada H, et al. Purification and properties of the 26S proteasome from the rat brain: evidence for its degradation of myelin basic protein in a ubiquitin-dependent manner. Brain Res 1996; 722: 139–44

    PubMed  CAS  Google Scholar 

  227. Lovell MA, Xie C, Markesbery WR. Decreased base excision repair and increased helicase activity in Alzheimer’s disease brain. Brain Res 2000; 855: 116–23

    PubMed  CAS  Google Scholar 

  228. Bonfanti L, Peretto P, De Marchis S, et al. Carnosine-related dipeptides in the mammalian brain. Prog Neurobiol 1999; 59: 333–53

    PubMed  CAS  Google Scholar 

  229. Aruoma OI, Laughton MJ, Halliwell B. Carnosine, homocarnosine and anserine: could they act as antioxidants in vivo? Biochem J 1989; 264: 863–9

    PubMed  CAS  Google Scholar 

  230. Zhou S, Dickinson C, Yang L, et al. Identification of hydrazine in commercial preparations of carnosine and its influence on carnosine’s antioxidative properties. Anal Biochem 1998; 261: 79–86

    PubMed  CAS  Google Scholar 

  231. Kohen R, Yamamoto Y, Cundy KC, et al. Antioxidant activity of carnosine, homocarnosine, and anserine present in muscle and brain. Proc Natl Acad Sci USA 1988; 85: 3175–9

    PubMed  CAS  Google Scholar 

  232. Horning MS, Blakemore LJ, Trombley PQ. Endogenous mechanisms of neuroprotection: role of zinc, copper, and carnosine. Brain Res 2000; 852: 56–61

    PubMed  CAS  Google Scholar 

  233. Tayarani I, Chaudiere J, Lefauconnier JM, et al. Enzymatic protection against peroxidative damage in isolated brain capillaries. J Neurochem 1987; 48: 1399–402

    PubMed  CAS  Google Scholar 

  234. Agarwal R, Shukla GS. Potential role of cerebral glutathione in the maintenance of blood-brain barrier integrity in rat. Neurochem Res 1999; 24: 1507–14

    PubMed  CAS  Google Scholar 

  235. Sachdev P, Saharov T, Cathcart S. The preventive role of antioxidants (selegiline and vitamin E) in a rat model of tardive dyskinesia. Biol Psychiatry 1999; 46: 1672–81

    PubMed  CAS  Google Scholar 

  236. Halliwell B. Drug antioxidant effects: a basis for drug selection? Drugs 1991; 42: 569–605

    PubMed  CAS  Google Scholar 

  237. Floyd RA, Hensley K, Jaffery F, et al. Increased oxidative stress brought on by pro-inflammatory cytokines in neurodegenerative processes and the protective role of nitrone-based free radical traps. Life Sci 1999; 65: 1893–9

    PubMed  CAS  Google Scholar 

  238. Dorey G, Lockhart B, Lestage P, et al. Novel quinolinic derivatives as centrally active antioxidants. Bioorganic Med Chem Lett 2000; 10: 935–9

    CAS  Google Scholar 

  239. Martinez M, Hernandez AI, Martinez N. N-Acetylcysteine delays age-associated memory empairment in mice: role in synaptic mitochondria. Brain Res 2000; 855: 100–6

    PubMed  CAS  Google Scholar 

  240. Chabrier PE, Auguet M, Spinnewyn B, et al. BN 80933, a dual inhibitor of neuronal nitric oxide synthase and lipid peroxidation: a promising neuroprotective strategy. Proc Natl Acad Sci USA 1999; 96: 10557–8

    Google Scholar 

  241. Vajragupta O, Monthakantirat O, Wongkrajang Y, et al. Chroman amide 12P: inhibition of lipid peroxidation and protection against learning and memory impairment. Life Sci 2000; 67: 1725–34

    PubMed  CAS  Google Scholar 

  242. Callaway JK, Beart PM, Jarrott B, et al. Incorporation of sodium channel blocking and free radical scavenging into a single drug, AM-36, results inprofound inhibition of neuronal apoptosis. Br J Pharmacol 2001; 132: 1691–8

    PubMed  CAS  Google Scholar 

  243. Marshall JWB, Duffin KJ, Green R, et al. NXY-059, a free radical-trapping agent, substantially lessens the functional disability resulting from cerebral ischaemia in a primate species. Stroke 2001; 32: 190–8

    PubMed  CAS  Google Scholar 

  244. Mytilineou C, Leonardi EK, Radcliffe P, et al. Deprenyl and desmethyselgiline protect mescencephalic neurons from toxicity induced by glutathione depletion. J Pharmacol Exp Ther 1998; 284: 700–6

    PubMed  CAS  Google Scholar 

  245. Gassen M, Gross A, Youdim MB. Apomorphine, a dopamine receptor agonist with remarkable antioxidant and cytoprotective properties. Adv Neurol 1999; 80: 297–302

    PubMed  CAS  Google Scholar 

  246. Kitani K, Kanai S, Ivy GO, et al. Pharmacological modifications of endogenous antioxidant enzymes with special reference to the effects of deprenyl: a possible antioxidant strategy. Mech Ageing Dev 1999; 111: 211–21

    PubMed  CAS  Google Scholar 

  247. Dunnett SB, Bjorklund A. Prospects for new restorative and neuroprotective treatments in Parkinson’s disease. Nature 1999; 399: A32–9

    PubMed  CAS  Google Scholar 

  248. Dexter DT, Holley AE, Flitter WD, et al. Increased levels of lipid hydroperoxides in the parkinsonian substantia nigra: an HPLC and ESR study. Mov Disord 1994; 9: 92–7

    PubMed  CAS  Google Scholar 

  249. Yoritaka A, Hattori N, Uchida K, et al. Immunohistochemical detection of 4-hydroxynonenal protein adducts in Parkinson disease. Proc Natl Acad Sci USA 1996; 93: 2696–701

    PubMed  CAS  Google Scholar 

  250. Parkinson Study Group. Effects of tocopherol and deprenyl on the progression of disability in early Parkinson’s disease. N Engl J Med 1993; 328:176–83

    Google Scholar 

  251. Alam ZI, Jenner A, Daniel SE, et al. Oxidative DNA damage in the parkinsonian brain: an apparent selective increase in 8-hydroxyguanine levels in substantia nigra. J Neurochem 1997; 69: 1196–203

    PubMed  CAS  Google Scholar 

  252. Zhang J, Perry G, Smith MA, et al. Parkinson’s disease is associated with oxidative damage to cytoplasmic DNA and RNA in substantia nigra neurons. Am J Pathol 1999; 154: 1423–9

    PubMed  CAS  Google Scholar 

  253. Shimura-Miura H, Hattori N, Kang D, et al. Increased 8-oxo-dGTPase in the mitochondria of substantia nigral neurons in Parkinson’s disease. Ann Neurol 1999; 46: 920–4

    PubMed  CAS  Google Scholar 

  254. Alam ZI, Daniel SE, Lees AJ, et al. A generalised increase in protein carbonyls in the brain in Parkinson’s but not incidental Lewy body disease. J Neurochem 1997; 69: 1326–9

    PubMed  CAS  Google Scholar 

  255. Pennathur S, Jackson-Lewis V, Przedborski S, et al. Mass spectrometric quantification of 3-nitrotyrosine, ortho-tyrosine and o,o′-dityrosine in brain tissue of MPTP-treated mice, a model of oxidative stress in Parkinson’s disease. J Biol Chem 1999; 274: 34621–8

    PubMed  CAS  Google Scholar 

  256. Giasson BI, Duda JE, Murray IV, et al. Oxidative damage linked to neurodegeneration by selective alpha-synuclein nitration in synucleinopathy lesions. Science 2000; 290: 985–9

    PubMed  CAS  Google Scholar 

  257. Good PF, Hsu A, Werner P, et al. Protein nitration in Parkinson’s disease. J Neuropathol Exp Neurol 1998; 57: 338–42

    PubMed  CAS  Google Scholar 

  258. Gotz ME, Kunig G, Riederer P, et al. Oxidative stress: free radical production in neural degeneration. Pharmacol Ther 1994; 63: 37–122

    PubMed  CAS  Google Scholar 

  259. Sofic E, Paulus W, Jellinger K, et al. Selective increase of iron in substantia nigra zona compacta of parkinsonian brains. J Neurochem. 1991; 56: 978–82

    PubMed  CAS  Google Scholar 

  260. Dexter DT, Carayon A, Javoy-Agid F, et al. Alterations in the levels of iron, ferritin and other trace metals in Parkinson’s disease and other neurodegenerative diseases affecting the basal ganglia. Brain 1991; 114: 1953–75

    PubMed  Google Scholar 

  261. Faucheux BA, Nillesse N, Damier P, et al. Expression of lactoferrin receptors is increased in the mesencephalon of patients with Parkinson disease. Proc Natl Acad Sci USA 1995; 92: 9603–7

    PubMed  CAS  Google Scholar 

  262. Dexter DT, Wells FR, Lees AJ, et al. Increased nigral iron content and alterations in other metal ions occurring in brain in Parkinson’s disease. J Neurochem 1989; 52: 1830–6

    PubMed  CAS  Google Scholar 

  263. Marttila RJ, Lorentz H, Rinne UK. Oxygen toxicity protecting enzymes in Parkinson’s disease. Increase of superoxide dismutase-like activity in the substantia nigra and basal nucleus. J Neurol Sci 1988; 86: 321–31

    PubMed  CAS  Google Scholar 

  264. Sian J, Dexter DT, Lees AJ, et al. Alterations in glutathione levels inParkinson’s disease andotherneurodegenerative disorders affecting basal ganglia. Ann Neurol 1994; 36: 348–55

    PubMed  CAS  Google Scholar 

  265. Sian J, Dexter DT, Lees AJ, et al. Glutathione-related enzymes in brain in Parkinson’s disease. Ann Neurol 1994; 36: 356–61

    PubMed  CAS  Google Scholar 

  266. Krige D, Carroll MT, Cooper JM, et al. Platelet mitochondrial function in Parkinson’s disease. Ann Neurol 1992; 32: 782–8

    PubMed  CAS  Google Scholar 

  267. Shults CW, Haas RH, Passov D, et al. Coenzyme Q10 levels correlate with the activities of complexes I and II/III in mitochondria from parkinsonian and nonparkinsonian subjects. Ann Neurol 1997; 42: 261–4

    PubMed  CAS  Google Scholar 

  268. Castellani R, Smith MA, Richey PL, et al. Glycoxidation and oxidative stress in Parkinson disease and diffuse Lewy body disease. Brain Res 1996; 737: 195–200

    PubMed  CAS  Google Scholar 

  269. Hunot S, Brugg B, Ricard D, et al. Nuclear translocation of NF-kappaB is increased in dopaminergic neurons of patients with parkinson disease. Proc Natl Acad Sci USA 1997; 94: 7531–6

    PubMed  CAS  Google Scholar 

  270. McGeer PL, Itagaki S, Boyes BE, et al. Reactive microglia are positive for HLA-DR in the substantia nigra of Parkinson’s and Alzheimer’s disease brains. Neurology 1998; 38: 1285–91

    Google Scholar 

  271. Toffa S, Kunikowska GM, Zeng BY, et al. Glutathione depletion in rat brain does not cause nigrostriatal pathway degeneration. J Neural Transm 1997; 104: 67–75

    PubMed  CAS  Google Scholar 

  272. Seaton TA, Jenner P, Marsden CD. Mitochondrial respiratory enzyme function and superoxide dismutase activity following brain glutathione depletion in the rat. Biochem Pharmacol 1996; 52: 1657–63

    PubMed  CAS  Google Scholar 

  273. Tolwani RJ, Jakowec MW, Petzinger GM, et al. Experimental models of Parkinson’s disease: insights from many models. Lab Animal Sci 1999; 49: 363–71

    CAS  Google Scholar 

  274. Temlett JA, Landsberg JP, Watt F, et al. Increased iron in the substantia nigra compacta of the MPTP-lesioned hemiparkinsonian African green monkey: evidence from proton microprobe elemental microanalysis. J Neurochem 1994; 62: 134–46

    PubMed  CAS  Google Scholar 

  275. Kanda S, Bishop JF, Eglitis MA, et al. Enhanced vulnerability to oxidative stress by alpha-synuclein mutations and C-terminal truncation. Neuroscience 2000; 97: 279–84

    PubMed  CAS  Google Scholar 

  276. Alves da Costa C, Ancolio K, Cheeler F. Wild-type but not Parkinson’s disease-related Ala-53 → Thr mutant α-synuclein protects neuronal cells from apoptotic stimuli. J Biol Chem 2000; 275: 24065–9

    Google Scholar 

  277. Ostrerova-Golts N, Petrucelli L, Hardy J, et al. The A53T α-synuclein mutation increases iron-dependent aggregation and toxicity. J Neurosci 2000; 20: 6048–54

    PubMed  CAS  Google Scholar 

  278. Jenner P, Dexter DT, Sian J, et al. Oxidative stress as a cause of nigral cell death in Parkinson’s disease and incidental Lewy body disease. The Royal Kings and Queens PD Study group. Ann Neurol 1992; 32 Suppl.: S82–7

    PubMed  CAS  Google Scholar 

  279. Marshall KA, Daniel SE, Cairns N, et al. Upregulation of the anti-apoptotic protein Bcl-2 may be an early event in neurodegeneration: studies on Parkinson’s and incidental Lewy body disease. Biochem Biophys Res Commun 1997; 240: 84–7

    PubMed  CAS  Google Scholar 

  280. Dexter DT, Sian J, Rose S, et al. Indices of oxidative stress and mitochondrial function in individuals with incidental Lewy body disease. Ann Neurol 1994; 35: 38–44

    PubMed  CAS  Google Scholar 

  281. Dickson DW, Lin W, Liu WK, et al. Multiple system atrophy: a sporadic synucleinopathy. Brain Pathol 1999; 9: 721–32

    PubMed  CAS  Google Scholar 

  282. Perez M, Valpuesta JM, de Garcini EM, et al. Ferritin is associated with the aberrant tau filaments present in progressive supranuclear palsy. Am J Pathol 1998; 152: 1531–9

    PubMed  CAS  Google Scholar 

  283. Lyras L, Zheng BY, McKenzie G, et al. Chronic high dose L-DOPA alone or in combination with the COMT inhibitor entacapone does not decrease oxidative damage or impair the function of the nigro-striatal pathway in normal cynomologous monkeys. J Neural Transm. In press

  284. Selkoe DJ. Alzheimer’s disease: genes, proteins and therapy. Physiol Rev 2001; 81: 741–66

    PubMed  CAS  Google Scholar 

  285. Mattson MP. Free radical-mediated disruption of cellular ion homeostasis, mitochondrial dysfunction and neuronal degeneration in sporadic and inherited Alzheimer’s disease. In: Poli G, Cadenas E, Packer L, editors. Free radicals in brain pathophysiology. New York: Marcel Dekker, 2000: 323–57

    Google Scholar 

  286. Emilien G, Beyreuther K, Masters CL, et al. Prospects for pharmacological intervention in Alzheimer disease. Arch Neurology 2000; 57: 454–9

    CAS  Google Scholar 

  287. Yatin SM, Varadarajan S, Link CD, et al. In vitro and in vivo oxidative stress associated with Alzheimer’s amyloid β-peptide (1–42). Neurobiol Aging 1999; 20: 325–30

    PubMed  CAS  Google Scholar 

  288. Iversen LL, Mortishire-Smith RJ, Pollack SJ, et al. The toxicity in vitro of β-amyloid protein. Biochem J 1995; 311: 1–16

    PubMed  CAS  Google Scholar 

  289. Barkats M, Millecamps S, Abrioux P, et al. Overexpression of glutathione peroxidase increases the resistance of neuronal cells to Aβ-mediated neurotoxicity. J Neurochem 2000; 75: 1439–46

    Google Scholar 

  290. Iadecola C, Zhang F, Niwa K, et al. SOD I rescues cerebral endothelial dysfunction in mice overexpressing amyloid precursor protein. Nature Neurosci 1999; 2: 157–61

    PubMed  CAS  Google Scholar 

  291. Dikalov SI, Vitek MP, Maples KR, et al. Amyloid β-peptides do not form peptide-derived free radicals spontaneously but can enhance metal-catalyzed oxidation of hydroxylamines to nitroxides. J Biol Chem 1999; 274: 9392–9

    PubMed  CAS  Google Scholar 

  292. Practico D, Lee VMY, Trozanowski JQ, et al. Increased F2-isoprostanes in Alzheimer’s disease: evidence for enhanced lipid peroxidation in vivo. FASEB J 1998; 12: 1777–83

    Google Scholar 

  293. Aksenov M, Aksenova M, Butterfield DA, et al. Oxidative modification of creatine kinase BB in Alzheimer’s disease brain. J Neurochem 2000; 74: 2520–7

    PubMed  CAS  Google Scholar 

  294. Montine TJ, Markesberg WR, Zachert W, et al. The magnitude of brain lipid peroxidation correlates with the extent of degeneration but not with density of neurotic plaques or neurofibrillary tangles or with APOE genotype in Alzheimer’s disease patients. Am J Pathol 1999; 155: 863–8

    PubMed  CAS  Google Scholar 

  295. Lyras L, Cairns NJ, Jenner A, et al. An assessment of oxidative damage to proteins, lipids, and DNA in brain from patients with Alzheimer’s disease. J Neurochem 1997; 68: 2061–9

    PubMed  CAS  Google Scholar 

  296. Good PF, Werner P, Hsu A, et al. Evidence of neuronal oxidative damage in Alzheimer’s disease. Am J Pathol 1996; 149: 21–8

    PubMed  CAS  Google Scholar 

  297. Gabbita SP, Lovell MA, Markesbery WR. Increased nuclear DNA oxidation in the brain in Alzheimer’s disease. J Neurochem 1998; 71: 2034–40

    PubMed  CAS  Google Scholar 

  298. Montine TJ, Beal MF, Cudkowicz ME, et al. Increased CSF F2-isoprostane concentration in probable Alzheimer’s disease. Neurology. 1999; 52: 562–5

    PubMed  CAS  Google Scholar 

  299. Aksenova MV, Aksenov MY, Payne RM, et al. Oxidation of cytosolic proteins and expression of creatine kinase BB in frontal lobe in different neurodegenerative disorders. Dementia Geriatr Cogn Dis 1999; 10: 158–65

    CAS  Google Scholar 

  300. Reich EE, Markesberg WE, Roberts II LJ, et al. Brain regional quantification of F-ring and D-/E-ring isoprostanes and neuroprostanes in Alzheimer’s disease. Am J Pathol 2001; 158: 293–7

    PubMed  CAS  Google Scholar 

  301. Prabico D, Clark CM, Lee VMY, et al. Increased 8,12-iso-iPF2d-VI in Alzheimer’s disease: correlation of a noninvasive index of lipid peroxidation with disease severity. Ann Neurol 2000; 48: 809–12

    Google Scholar 

  302. Russell RL, Siedlak SL, Raina AK, et al. Increased neuronal glucose-6-phosphate dehydrogenase and sulfhydryl levels indicate reductive compensation to oxidative stress in Alzheimer disease. Arch Biochem Biophys 1999; 370: 236–9

    PubMed  CAS  Google Scholar 

  303. Smith MA, Richey PL, Kutty RK, et al. Ultrastructural localization of heme oxygenase-1 to the neurofibrillary pathology of Alzheimer disease. Mol Chem Neuropathol 1995; 24: 227–30

    PubMed  CAS  Google Scholar 

  304. Su JH, Deng G, Cotman CW. Neuronal DNA damage precedes tangle formation and is associated with up-regulation of nitrotyrosine in Alzheimer’s disease brain. Brain Res 1997; 774: 193–9

    PubMed  CAS  Google Scholar 

  305. Hensley K, Maidt ML, Yu Z, et al. Electrochemical analysis of protein nitrotyrosine and dityrosine in the Alzheimer brain indicates region-specific accumulation. J Neurosci 1998; 18: 8126–32

    PubMed  CAS  Google Scholar 

  306. Favit A, Grimaldi M, Alkon DL. Prevention of β-amyloid neurotoxicity by blockade of the ubiquitin-proteasome proteolytic pathway. J Neurochem 2000; 75: 1258–63

    PubMed  CAS  Google Scholar 

  307. Ii K, Ito H, Tanaka K, et al. Immunocytochemical co-localization of the proteasome in ubiquitinated structures in neurodegenerative diseases and the elderly. J Neuropathol Exp Neurol 1997; 56: 125–31

    PubMed  CAS  Google Scholar 

  308. Gregori L, Hainfeld JF, Simon MN, et al. Binding of amyloid beta protein to the 20 S proteasome. J Biol Chem 1997; 272: 58–62

    PubMed  CAS  Google Scholar 

  309. Keller JN, Hanni KB, Markesbery WR. Impaired proteasome function in Alzheimer’s disease. J Neurochem 2000; 75: 436–9

    PubMed  CAS  Google Scholar 

  310. Lee MH, Hyun DH, Jenner P, et al. Effect of proteasome inhibition on cellular oxidative damage and antioxidant defences. J Neurochem 2001; 78: 32–41

    PubMed  CAS  Google Scholar 

  311. Friguet B, Szweda LI. Inhibition of the multicatalytic proteinase (proteasome) by 4-hydroxy-2-nonenal cross-linked protein. FEBS Lett 1997; 405: 21–5

    PubMed  CAS  Google Scholar 

  312. Shringarpure R, Grune T, Sitte N, et al. 4-Hydroxynonenalmodified amyloid-β peptide inhibits the proteasome: possible importance in Alzheimer’s disease. Cell Mol Life Sci 2000; 57: 1802–9

    PubMed  CAS  Google Scholar 

  313. Pappolla MA, Chyan YJ, Sos M, et al. Role of transgenic models for the study of oxidative neurotoxicity in Alzheimer’s disease. In: Poli G, Cadenas E, Packer L, editors. Free radicals in brain pathophysiology. New York (NY): Marcel Dekker, 2000: 359–81

    Google Scholar 

  314. Lyras L, Perry RH, Perry EK, et al. Oxidative damage to proteins, lipids, and DNA in cortical brain regions from patients with dementia with Lewy bodies. J Neurochem 1998; 71: 302–12

    PubMed  CAS  Google Scholar 

  315. Greeve I, Hermans-Borgmeyer I, Brellinger C, et al. The human DIMINUTO/DWARFI homology Seladin — 1 confers resistance to Alzheimer’s disease — associated neurodegeneration and oxidative stress. J Neurosci 2000; 20: 7345–52

    PubMed  CAS  Google Scholar 

  316. Frenkel D, Katz O, Solomon B. Immunization against Alzheimer’s β-amyloid plaques via EFRH phage administration. Proc Natl Acad Sci USA 2000; 97: 11455–9

    PubMed  CAS  Google Scholar 

  317. Sayre LM, Perry G, Harris PLR, et al. In situ oxidative catalysis by neurofibrillary tangles and senile plaques in Alzheimer’s disease: a central role for bound transition metals. J Neurochem 2000; 74: 270–9

    PubMed  CAS  Google Scholar 

  318. Huang X, Cuajungo, MP, Atwood CS, et al. Cu (II) potentiation of Alzheimer Aβ neurotoxicity. J Biol Chem 1999; 274: 37111–6

    PubMed  CAS  Google Scholar 

  319. Bush AI, Huang X, Fairlie DP. The possible origin of free radicals from amyloid β peptides in Alzheimer’s disease. Neurobiol Aging 1999; 20: 335–7

    Google Scholar 

  320. Cherny RA, Legg JT, McLean CA, et al. Aqueous dissolution of Alzheimer’s disease Abeta amyloid deposits by biometal depletion. J Biol Chem 1999; 274: 23223–8

    PubMed  CAS  Google Scholar 

  321. Cuajungco MP, Goldstein LE, Nunomura A, et al. Evidence that the beta-amyloid plaques of Alzheimer’s disease represent the redox-silencing and entombment of A beta by zinc. J Biol Chem 2000; 275: 19439–42

    PubMed  CAS  Google Scholar 

  322. Smith MA, Wehr K, Harris PLR, et al. Abnormal localization of iron regulatory protein in Alzheimer’s disease. Brain Res 1998; 788: 232–6

    PubMed  CAS  Google Scholar 

  323. Rottkamp CA, Raina AK, Zhu X, et al. Redox-active iron mediates amyloid-β toxicity. Free Rad Biol Med 2001 30: 447–50

    PubMed  CAS  Google Scholar 

  324. Cherny RA, Atwood CS, Xilinas ME, et al. Treatment with a copper-zinc chelator markedly and rapidly inhibits beta-amyloid accumulation in Alzheimer’s disease transgenic mice. Neuron 2001; 30: 665–76

    PubMed  CAS  Google Scholar 

  325. Bence NF, Sampat RM, Kopito RR. Impairment of the ubiquitin-proteasome system by protein aggregation. Science 2001; 292: 1552–5

    PubMed  CAS  Google Scholar 

  326. Dukan S, Farewell A, Ballesteros M, et al. Protein oxidation in response to increased transcriptional or translational errors. Proc Natl Acad Sci USA 2000; 97: 5746–9

    PubMed  CAS  Google Scholar 

  327. McGeer EG, McGeer PL. The importance of inflammatory mechanisms in Alzheimer’s disease. Exp Gerontol 1998; 33: 371–8

    PubMed  CAS  Google Scholar 

  328. Della Bianca V, Dusi S, Bianchini E, et al. β-Amyloid activates the O2 forming NADPH oxidase in microglia monocytes and neutrophils. J Biol Chem 1999; 274: 15493–9

    PubMed  CAS  Google Scholar 

  329. Hull M, Fiebich BL, Schumann G, et al. Anti-inflammatory substances: a new therapeutic option in Alzheimer’s disease. Drug Discov Today 1999; 4: 275–82

    PubMed  CAS  Google Scholar 

  330. Prasad KN, Hovland AR, La Rosa FG, et al. Prostaglandins as putative neurotoxins in Alzheimer’s disease. Proc Soc Exp Biol Med 1998; 219: 120–5

    PubMed  CAS  Google Scholar 

  331. Lim GP, Yang F, Chu T, et al. Ibuprofen suppresses plaque pathology and inflammation in a mouse model for Alzheimer’s disease. J Neurosci 2000; 20: 5709–14

    PubMed  CAS  Google Scholar 

  332. Du Yan S, Zhu H, Fu J, et al. Amyloid-beta peptide-receptor for advanced glycation endproduct interaction elicits neuronal expression of macrophage-colony stimulating factor: a proinflammatory pathway in Alzheimer disease. Proc Natl Acad Sci USA 1997; 94: 5296–301

    PubMed  CAS  Google Scholar 

  333. Sasaki N, Fukatsu R, Tsuzuki K, et al. Advanced glycation end products in Alzheimer’s disease and other neurodegenerative diseases. Am J Pathol 1998; 153: 1149–55

    PubMed  CAS  Google Scholar 

  334. Giulian D, Vaca K. Inflammatory glia mediate delayed neuronal damage after ischaemia in the central nervous system. Stroke 1993; 24: I84–90

    PubMed  CAS  Google Scholar 

  335. Vatassery GT, Bauer T, Dysken M. High doses of vitamin E in the treatment of disorders of the central nervous system in the aged. Am J Clin Nutr 1999; 70: 793–801

    PubMed  CAS  Google Scholar 

  336. Morris MC, Beckett LA, Scherr PA, et al. Vitamin E and vitamin C supplement use and risk of incident Alzheimer disease. Alz Dis Assoc Disord 1998; 12: 121–6

    CAS  Google Scholar 

  337. Masaki KH, Losonczy KG, Izmirlian G, et al. Association of vitamin E and C supplement use with cognitive function and dementia in elderly men. Neurology 2000; 54: 1265–72

    PubMed  CAS  Google Scholar 

  338. Murray CA, Lynch MA. Dietary supplementation with vitamin E reverses the age-related deficit in long term potentiation in dentate gyrus. J Biol Chem 1998; 273: 12161–8

    PubMed  CAS  Google Scholar 

  339. Logroscino G, Marder K, Cote L, et al. Dietary lipids and antioxidants in Parkinson’s disease: a population-based, case-control study. Ann Neurol 1996; 39: 89–94

    PubMed  CAS  Google Scholar 

  340. Radak S, Kaneko T, Tahara S, et al. Regular exercise improves cognitive function and decreases oxidative damage in rat brain. Neurochem Int 2001; 38: 17–23

    PubMed  CAS  Google Scholar 

  341. Bickford PC, Gould T, Briederick L, et al. Antioxidant-rich diets improve cerebellar physiology and motor learning in aged rats. Brain Res 2000; 866: 211–7

    PubMed  CAS  Google Scholar 

  342. Alexi T, Borlongan CV, Faull RLM, et al. Neuroprotective strategies for basal ganglia degeneration: Parkinson’s and Huntington’s diseases. Prog Neurobiol 2000; 60: 409–70

    PubMed  CAS  Google Scholar 

  343. Alam ZI, Halliwell B, Jenner P. No evidence for increased oxidative damage to lipids, proteins, or DNA in Huntington’s disease. J Neurochem 2000; 75: 840–6

    PubMed  CAS  Google Scholar 

  344. Deckel AW, Volmer P, Weiner R, et al. Dietary arginine alters time of symptom onset in Huntington’s disease transgenic mice. Brain Res 2000; 875: 187–95

    PubMed  CAS  Google Scholar 

  345. Morrison BM, Morrison JH. Amyotrophic lateral sclerosis associated with mutations in superoxide dismutase: a putative mechanism of degeneration. Brain Res 1999; 29: 121–35

    CAS  Google Scholar 

  346. Gurney ME, Liu R, Althaus JS, et al. Mutant CuZn superoxide dismutase in motor neuron disease. J Inherit Metab Dis 21: 587–97

  347. Gahtan E, Auerbach JM, Groner Y, et al. Reversible impairment of long-term potentiation in transgenic Cu/Zn-SOD mice. Eur J Neurosci 1998; 10: 538–44

    PubMed  CAS  Google Scholar 

  348. Cleveland DW. From Charcot to SOD1: mechanisms of selective motor neuron death in ALS. Neuron 1999; 24: 515–20

    PubMed  CAS  Google Scholar 

  349. Cookson MR, Shaw PJ. Oxidative stress and motor neurone disease. Brain Pathol 1999; 9: 165–86

    PubMed  CAS  Google Scholar 

  350. Estévez AG, Crow JP, Sampson JB, et al. Induction of nitric oxide-dependent apoptosis in motor neurons by zinc-deficient superoxide dismutase. Science 1999; 286: 2498–500

    PubMed  Google Scholar 

  351. Gabbianelli R, Ferri A, Rotilio G, et al. Aberrant copper chemistry as a major mediator of oxidative stress in a human cellular model of amyotrophic lateral sclerosis. J Neurochem 1999; 73: 1175–80

    PubMed  CAS  Google Scholar 

  352. Won SE, Jung HK. Release of copper ions from the familial amyotrophic lateral sclerosis-associated Cu,Zn-superoxide dismutase mutants. Mol Cell 1999; 9: 110–4

    Google Scholar 

  353. Ookawara T, Kawamura N, Kitagawa Y, et al. Site-specific and random fragmentation of Cu,Zn-superoxide dismutase by glycation reaction: implication of reactive oxygen species. J Biol Chem 1992; 267: 18505–10

    PubMed  CAS  Google Scholar 

  354. Ogawa Y, Kosaka H, Nakanishi T, et al. Stability of mutant superoxide dismutase-1 associated with familial amyotrophic lateral sclerosis determines the manner of copper release and induction of thioredoxin in erythrocytes. Biochem Biophys Res Commun 1997; 241: 251–7

    PubMed  CAS  Google Scholar 

  355. Wiedau-Pazos M, Goto JJ, Rabizadeh S, et al. Altered reactivity of superoxide dismutase in familial amyotrophic lateral sclerosis. Science 1996; 271: 515–8

    PubMed  CAS  Google Scholar 

  356. Café C, Testa MP, Sheldons PJ, et al. Loss of oxidation-reduction specificity in amyotrophic lateral sclerosis-associated copper-zinc superoxide dismutase mutants. J Biol Chem 1998; 273: 1–6

    Google Scholar 

  357. Williamson TL, Cleveland DW. Slowing of axonal transport is a very early event in the toxicity of ALS-linked SOD1 mutants to motor neurons. Nature Neurosci 1999; 2: 50–6

    PubMed  CAS  Google Scholar 

  358. Ferrante RJ, Browne SE, Shinobu LA, et al. Evidence of increased oxidative damage in both sporadic and familial amyotrophic lateral sclerosis. J Neurochem 1997; 69: 2064–74

    PubMed  CAS  Google Scholar 

  359. Tohgi H, Abe T, Yamazaki K, et al. Remarkable increase in cerebrospinal fluid 3-Nitrotyrosine in patients with sporadic amyotrophic lateral sclerosis. Ann Neurol 1999; 46: 129–31

    PubMed  CAS  Google Scholar 

  360. Bogdanov M, Brown Jr RH, Matson W, et al. Increased oxidative damage to DNA in ALS patients. Free Radic Biol Med 2000; 29: 652–8

    PubMed  CAS  Google Scholar 

  361. Andreassen OA, Dedeoglu A, Klivenyi P, et al. N-acetyl-L-cysteine improves survival and preserves motor performance in an animal model of familial amyotrophic lateral sclerosis. Neuroreport 2000; 11: 2491–3

    PubMed  CAS  Google Scholar 

  362. Andreassen OA, Dedeoglu A, Friedlich A, et al. Effect of an inhibitor of poly (ADP-ribose) polymerase, desmethylselegiline, trientine and lipoic acid in transgenic ALS mice. Exp Neurol 2001; 168: 419–24

    PubMed  CAS  Google Scholar 

  363. Dugan LL, Lovelt EG, Quick KL, et al. Fullerene-based antioxidants and neurodegenerative disorders. Parkinsonism Relat Dis 2001;7:243–6

    Google Scholar 

  364. Hottinger AF, Fine EG, Gurney ME, et al. The copper chelator d-penicillamine delays onset of disease and extends survival in a transgenic mouse model of familial ALS. Eur J Neurosci 1997; 9: 1548–51

    PubMed  CAS  Google Scholar 

  365. Poduslo JF, Whelan SL, Curren GL, et al. Therapeutic benefit of polyamine-modified catalase as a scavenger of hydrogen peroxide in familial amyotrophic lateral sclerosis transgenics. Ann Neurol 2000; 48: 943–7

    PubMed  CAS  Google Scholar 

  366. Reinholz MM, Merkle CM, Poduslo JF. Therapeutic benefits of putrescine-modified catalase in a transgenic mouse model of familial amyotrophic lateral sclerosis. Exp Neurol 1999; 159: 204–16

    PubMed  CAS  Google Scholar 

  367. Kennel P, Revah F, Bohme GA, et al. Riluzole prolongs survival and delays muscle strength deterioration in mice with progressive motor neuropathy (pmn). J Neurol Sci 2000; 180: 55–61

    PubMed  CAS  Google Scholar 

  368. Miller RG. Examining the evidence about treatment in ALS/ MND. Amyotroph Lateral Scler Other Motor Neuron Disord 2001; 2: 3–7

    PubMed  CAS  Google Scholar 

  369. Desnuelle C, Dib M, Garrel C, et al. A double-blind placebo-controlled randomized trial of α-tocopherol (vitamin E) in the treatment of ALS. Amyotroph Lateral Scler Other Motor Neuron Disor 2001; 2: 9–18

    CAS  Google Scholar 

  370. Vyth A, Timmer JG, Bossvyt PM, et al. Survival in patients with ALS treated with an array of antioxidants. J Neurol Sci 1996; 139 Suppl.: 99–103

    PubMed  CAS  Google Scholar 

  371. Lange DJ, Murphy PL, Diamond B, et al. Selegiline is ineffective in a collaborative double-blind placebo-controlled trial for treatment of ALS. Arch Neurol 1998; 55: 93–6

    PubMed  CAS  Google Scholar 

  372. Upton-Rice MN, Cudkowicz ME, Mathew RK, et al. Administration of nitric oxide synthase inhibitors does not alter disease course of amyotrophic lateral sclerosis SOD1 mutant transgenic mice. Ann Neurol 1999; 45: 413–4

    PubMed  CAS  Google Scholar 

  373. Pedersen WA, Mattson MP. No benefit of dietary restriction on disease onset or progression in amytrophic lateral sclerosis Cu/Zn-superoxide dismutase mutant mice. Brain Res 1999; 833: 117–20

    PubMed  CAS  Google Scholar 

  374. Kostic V, Jackson-Lewis V, de Bilbao F, et al. Bcl-2: prolonging life in a transgenic mouse model of familial amyotrophic lateral sclerosis. Science 1997; 277: 559–62

    PubMed  CAS  Google Scholar 

  375. Okado-Matsumoto A, Myint T, Fujii J, et al. Gain in functions of mutant Cu,Zn-superoxide dismutases as a causative factor in familial amyotrophic lateral sclerosis: less reactive oxidant formation but high spontaneous aggregation and precipitation. Free Radic Res 2000; 33: 65–73

    PubMed  CAS  Google Scholar 

  376. Durham HD, Roy J, Dong L, et al. Aggregation of mutant Cu/Zn superoxide dismutase proteins in a culture model of ALS. J Neuropathol Exp Neurol 1997; 56: 523–30

    PubMed  CAS  Google Scholar 

  377. Stieber A, Gonatas JO, Gonatas NK. Aggregation of ubiquitin and a mutant ALS-linked SOD 1 protein correlate with disease progression and fragmentation of the Golgi apparatus. J Neurol Sci 2000; 173: 53–62

    PubMed  CAS  Google Scholar 

  378. Kakizuka A. Protein precipitation: a common etiology in neurodegenerative disorders? Trends Genet 1998; 14: 396–402

    PubMed  CAS  Google Scholar 

  379. Marshall KA, Reiter RJ, Poeggeler B, et al. Evaluation of the antioxidant activity of melatonin in vitro.Free Radic Biol Med 1996; 21: 307–15

    PubMed  CAS  Google Scholar 

  380. Reiter RJ, Cabrera J, Sainz RM, et al. Melatonin as a pharmacological agent against neuronal loss in experimental models of Huntington’s disease, Alzheimer’s disease and parkinsonism. Ann NY Acad Sci 1999; 890: 471–85

    PubMed  CAS  Google Scholar 

  381. Anderson ME, Underwood M, Bridges RJ, et al. Glutathione metabolism at the blood-cerebrospinal fluid barrier. FASEB J 1989; 3: 2527–31

    PubMed  CAS  Google Scholar 

  382. Anderson ME, Meister A. Marked increase of cysteine levels in many regions of the brain after administration of 2-oxothiazolidine-4-carboxylate. FASEB J 1989; 3: 1632–6

    PubMed  CAS  Google Scholar 

  383. Packer L, Tritschler HJ, Wessel K. Neuroprotection by the metabolic antioxidant alpha-lipoic acid. Free Radic Biol Med 1997; 22: 359–78

    PubMed  CAS  Google Scholar 

  384. Barbiroli B, Medori R, Tritschler HJ, et al. Lipoic (thioctic) acid increases brain energy availability and skeletal muscle performance as shown by in vivo 31P-MRS in a patient with mitochondrial cytopathy. J Neurol 1995; 242: 472–7

    PubMed  CAS  Google Scholar 

  385. Seaton TA, Jenner P, Marsden CD. The isomers of thioctic acid alter C-deoxyglucose incorporation in rat basal ganglia. Biochem Pharmacol 1996; 51: 983–6

    PubMed  CAS  Google Scholar 

  386. DANA Consortium. A randomized, double-blind, placebo controlled trial of deprenyl and thioctic acid in HIV-associated cognitive impairment. Neurology 1998; 50: 645–51

    Google Scholar 

  387. Chao CC, Lee EH. Neuroprotective mechanism of glial cell line-derived neurotrophic factor on dopamine neurons: role of antioxidation. Neuropharmacology 1999; 38: 913–6

    PubMed  CAS  Google Scholar 

  388. Hou JG, Cohen G, Mytilineou C. Basic fibroblast growth factor stimulation of glial cells protects dopamine neurons from 6-hydroxydopamine toxicity: involvement of the glutathione system. J Neurochem 1997; 69: 76–83

    PubMed  CAS  Google Scholar 

  389. Suzuki YJ, Tsuchiya M, Safadi A, et al. Antioxidant properties of nitecapone. Free Radic Biol Med 1992; 13: 517–25

    PubMed  CAS  Google Scholar 

  390. Sramek JJ, Cutler NR. Recent developments in the drug treatment of Alzheimer’s disease. Drugs Aging 1999; 14: 359–73

    PubMed  CAS  Google Scholar 

  391. Rustin P, von Kleist-Retzow JC, Chantrel-Groussard K, et al. Effect of idebenone on cardiomyopathy in Friedreich’s ataxia: a preliminary study. Lancet 1999; 354: 477–9

    PubMed  CAS  Google Scholar 

  392. Gutzmann H, Hadler D. Sustained efficacy and safety of idebenone in the treatment of Alzheimer’s disease: update on a 2-year double-blind multicentre study. J Neural Transm 1998; Suppl 54: 301–10

    Google Scholar 

  393. Adkins JC, Noble S. Idebenone: a review of its use in mild to moderate Alzheimer’s disease. CNS Drugs 1998; 9: 403–19

    CAS  Google Scholar 

  394. Shults CW, Haas RH, Beal MF. A possible role of coenzyme Q10 in the etiology and treatment of Parkinson’s disease. Biofactors 1999; 9: 267–72

    PubMed  CAS  Google Scholar 

  395. Carney JM, Starke-Reed PE, Oliver CN, et al. Reversal of age-related increase in brain protein oxidation, decrease in enzyme activity, and loss in temporal and spatial memory by chronic administration of the spin-trapping compound N-tert-butyl-alpha-phenylnitrone. Proc Natl Acad Sci USA 1991; 88: 3633–6

    PubMed  CAS  Google Scholar 

  396. Floyd RA. Antioxidants, oxidative stress, and degenerative neurological disorders. Proc Soc Exp Biol Med 1999; 222: 236–45

    PubMed  CAS  Google Scholar 

  397. Kuroda S, Tsuchidate R, Smith ML, et al. Neuroprotective effects of a novel nitrone, NXY-059, after transient focal cerebral ischemia in the rat. J Cereb Blood Flow Metab 1999; 19: 778–87

    PubMed  CAS  Google Scholar 

  398. Kotake Y, Sang H, Miyajima T, et al. Inhibition of NF-kappaB, iNOS mRNA, COX2 mRNA, and COX catalytic activity by phenyl-N-tert-butylnitrone (PBN). Biochim Biophys Acta 1998; 1448: 77–84

    PubMed  CAS  Google Scholar 

  399. Chamulitrat W, Parker CE, Tomer KB, et al. Phenyl N-tert-butyl nitrone forms nitric oxide as a result of its Fe(III)-catalyzed hydrolysis or hydroxyl radical adduct formation. Free Radic Res 1995: 23: 1–14

    PubMed  CAS  Google Scholar 

  400. Janzen EG, Poyer JL, Schaefer CF, et al. Biological spin trapping. II. Toxicity of nitrone spin traps: dose-ranging in the rat. J Biochem Biophys Methods 1995; 30: 239–47

    PubMed  CAS  Google Scholar 

  401. Schewe T. Molecular actions of ebselen: an anti-inflammatory antioxidant. Gen Pharmacol 1995; 26: 1153–69

    PubMed  CAS  Google Scholar 

  402. Ogawa A, Yoshimoto T, Kikuchi H, et al. Ebselen in acute middle cerebral artery occlusion: a placebo-controlled, doubleblind clinical trial. Cerebrovasc Dis 1999; 9: 112–8

    PubMed  CAS  Google Scholar 

  403. Saito I, Asano T, Sano K, et al. Neuroprotective effect of an antioxidant, ebselen, in patients with delayed neurological deficits after aneurismal subarachnoid hemorrhage. Neurosurgery 1998; 42: 269–77

    PubMed  CAS  Google Scholar 

  404. Gladilin S, Bidmon Hj, Divanach A, et al. Ebselen lowers plasma interleukin-6 levels and glial heme oxygenase-1 expression after focal photothrombotic brain ischaemia. Arch Biochem Biophys 2000; 380: 237–42

    PubMed  CAS  Google Scholar 

  405. Glick H, Willke R, Polsky D, et al. Economic analysis of tirilazad mesylate for aneurismal subarachnoid hemorrhage: economic evaluation of a phase III clinical trial in Europe and Australia. Int J Technol Assess Health Care 1998; 14: 145–60

    PubMed  CAS  Google Scholar 

  406. Tirilazad International Steering Committee. Tirilazad mesylate in acute ischemic stroke: a systematic review. Stroke 2000; 32: 2257–65

    Google Scholar 

  407. Hall ED, Andrus PK, Smith SL, et al. Neuroprotective efficacy of microvascularly-localized versus brain-penetrating antioxidants. Acta Neurochirurgica Suppl 1996; 66: 107–13

    CAS  Google Scholar 

  408. Schmid-Elsaesser R, Hungerhuber E, Zausinger S, et al. Neuroprotective effects of the novel brain-penetrating antioxidant U-101033E and the spin-trapping agent alpha-phenyl-N-tertbutyl nitrone (PBN). Exp Brain Res 2000; 130: 60–6

    PubMed  CAS  Google Scholar 

  409. Oshiro Y, Sakurai Y, Tanaka T, et al. Novel cerebroprotective agents with central nervous system stimulating activity. 2. Synthesis and pharmacology of the 1-(acylamino)-7-hydroxyindan derivatives. J Med Chem 1991; 34: 2014–23

    PubMed  CAS  Google Scholar 

  410. Nakai M, Qin ZH, Wang Y, et al. Free radical scavenger OPC-14117 attentuates quinolinic acid-induced NF-kappaB activation and apoptosis in rat striatum. Brain Res 1999; 64: 59–68

    CAS  Google Scholar 

  411. Safety and tolerability of the free-radical scavenger OPC-14117 in Huntington’s disease. The Huntington Study Group. Neurology 1998; 50: 1366–73

  412. Safety and tolerability of the antioxidant OPC-14117 in HIV-associated cognitive impairment. The Dana Consortium on the Therapy of HIV Dementia and Related Cognitive Disorders. Neurology 1997; 49: 142–6

  413. Blanchet PJ, Konitsiotis S, Hyland K, et al. Chronic exposure to MPTP as a primate model of progressive parkinsonism: a pilot study with a free radical scavenger. Exp Neurol 1998; 153: 214–22

    PubMed  CAS  Google Scholar 

  414. Abe K, Morita S, Kikuchi T, et al. Protective effect of a novel free radical scavenger, OPC-14117, on Wobbler mouse motor neuron disease. J Neurosci Res 1997; 48: 63–70

    PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Barry Halliwell.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Halliwell, B. Role of Free Radicals in the Neurodegenerative Diseases. Drugs & Aging 18, 685–716 (2001). https://doi.org/10.2165/00002512-200118090-00004

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2165/00002512-200118090-00004

Keywords

Navigation