Skip to main content

Protective Effects of Adaptation to Hypoxia in Experimental Alzheimer’s Disease

  • Chapter
  • First Online:
Intermittent Hypoxia and Human Diseases

Abstract

Alzheimer’s disease (AD) is characterized by formation of amyloid plaques, intracellular neurofibrillary tangles, and cell death in the brain, resulting in progressive loss of memory and cognitive ability. Efficacy of drugs currently used for prevention and treatment of AD is limited by the fact that each drug influences only a single step of the pathogenesis in AD, and the drugs affect both damaged and normal cells. This is why major attention is now paid to nonpharmacological means that may enhance the adaptive capacity and mobilize the self-defense systems of the body. This chapter focuses on protective effects of adaptation to intermittent hypobaric hypoxia on the memory, brain neurons, and cerebral blood vessels in rats with experimental AD induced by intracerebral injections of beta-amyloid (Aβ) and mechanisms of these protective effects. Special attention is paid to intermittent hypobaric hypoxia’s ability to limit early stages in AD pathogenesis, such as nitrosative and oxidative stress in brain tissue. Presented data show that adaptation to hypoxia may be a promising approach to prevention and treatment of AD.

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

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 99.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 129.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 199.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Abbreviations

3-NT:

3-Nitrotyrosine

Ach:

Acetylcholine

AChE:

Acetylcholine esterase

AD:

Alzheimer’s disease

Aβ:

β-Amyloid

AMPA:

α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid

DNIC:

Dinitrosyl iron complex

HIF-1:

Hypoxia-inducible factor-1

HRE:

Hypoxic response element

HSP:

Heat shock protein

LCBF:

Local cerebral blood flow

IL:

Interleukin

LTP:

Long-term potentiation

MPTP:

Mitochondrial permeability transition pore

NAC:

N-Acetylcysteine

NMDA:

N-Methyl d-aspartate

L-NNA:

Nω-Nitro-L-arginine

NO:

Nitric oxide

NOS:

NO synthase

eNOS:

Endothelial NO synthase

iNOS:

Inducible NO synthase

nNOS:

Neuronal NO synthase

PKC:

Protein kinase C

ROS:

Reactive oxygen species

TBARS:

Thiobarbituric acid-reactive substances

TNFα:

Tumor necrosis factor α

VEGF:

Vascular endothelial growth factor

References

  1. Park SY. Potential therapeutic agents against Alzheimer’s disease from natural sources. Arch Pharm Res. 2010;33:1589–609.

    Article  PubMed  CAS  Google Scholar 

  2. Katzman R, Kawas C. Epidemiology of dementia and Alzheimer disease. In: Terry RD, Katzman R, Bick KL, editors. Alzheimer disease. New York: Raven; 1994. p. 105–22.

    Google Scholar 

  3. Wimo A, Winblad B, Aguero-Torres H, et al. The magnitude of dementia occurrence in the world. Alzheimer Dis Assoc Disord. 2003;17:63–7.

    Article  PubMed  Google Scholar 

  4. Cummings JL, Vinters HV, Cole GM, et al. Alzheimer’s disease: etiologies, pathophysiology, cognitive reserve, and treatment opportunities. Neurology. 1998;51:S2–17.

    Article  PubMed  CAS  Google Scholar 

  5. LaFerla FM. Pathways linking Abeta and tau pathologies. Biochem Soc Trans. 2010;38:993–5.

    Article  PubMed  CAS  Google Scholar 

  6. Green KN, Smith IF, Laferla FM. Role of calcium in the pathogenesis of Alzheimer’s disease and transgenic models. Subcell Biochem. 2007;45:507–21.

    Article  PubMed  CAS  Google Scholar 

  7. Small DH. Dysregulation of calcium homeostasis in Alzheimer’s disease. Neurochem Res. 2009;34:1824–9.

    Article  PubMed  CAS  Google Scholar 

  8. Ferreiro E, Oliveira CR, Pereira CM. The release of calcium from the endoplasmic reticulum induced by amyloid-beta and prion peptides activates the mitochondrial apoptotic pathway. Neurobiol Dis. 2008;30:331–42.

    Article  PubMed  CAS  Google Scholar 

  9. Mancuso M, Orsucci D, LoGerfo A, et al. Clinical features and pathogenesis of Alzheimer’s disease: involvement of mitochondria and mitochondrial DNA. Adv Exp Med Biol. 2010;685:34–44.

    Article  PubMed  CAS  Google Scholar 

  10. Murakami Y, Ohsawa I, Kasahara T, et al. Cytoprotective role of mitochondrial amyloid beta peptide-binding alcohol dehydrogenase against a cytotoxic aldehyde. Neurobiol Aging. 2009;30:325–9.

    Article  PubMed  CAS  Google Scholar 

  11. Singh S, Suman S, Chandra S, et al. Possible role of amyloid-beta, adenine nucleotide translocase and cyclophilin-D interaction in mitochondrial dysfunction of Alzheimer’s disease. Bioinformation. 2009;3:440–5.

    Article  PubMed  Google Scholar 

  12. Tillement L, Lecanu L, Papadopoulos V. Alzheimer’s disease: effects of β-amyloid on mitochondria. Mitochondrion. 2011;11:13–21.

    Article  PubMed  CAS  Google Scholar 

  13. Sergeant N, Wattez A, Galvan-Valencia M, et al. Association of ATP synthase alpha-chain with neurofibrillary degeneration in Alzheimer’s disease. Neuroscience. 2003;117:293–303.

    Article  PubMed  CAS  Google Scholar 

  14. Schmidt C, Lepsverdize E, Chi SL, et al. Amyloid precursor protein and amyloid beta-peptide bind to ATP synthase and regulate its activity at the surface of neural cells. Mol Psychiatry. 2008;13:953–69.

    Article  PubMed  CAS  Google Scholar 

  15. Hauptmann S, Scherping I, Dröse SU, et al. Mitochondrial dysfunction: an early event in Alzheimer pathology accumulates withy age in AD transgenic mice. Neurobiol Aging. 2009;30:1575–86.

    Article  CAS  Google Scholar 

  16. Rhein V, Baysang G, Rao S, et al. Amyloid-beta leads to impaired cellular respiration, energy production and mitochondrial electron chain complex activities in human neuroblastoma cells. Cell Mol Neurobiol. 2009;29:1063–71.

    Article  PubMed  CAS  Google Scholar 

  17. Aslan M, Özben T. Reactive oxygen and nitrogen species in Alzheimer’s disease. Curr Alzheimer Res. 2004;1:111–9.

    Article  PubMed  CAS  Google Scholar 

  18. Mecocci P, MacGarvey U, Beal MF. Oxidative damage to mitochondrial DNA is increased in Alzheimer’s disease. Ann Neurol. 1994;36:747–51.

    Article  PubMed  CAS  Google Scholar 

  19. Lovell MA, Ehmann WD, Butler SM, et al. Elevated thiobarbituric acid-reactive substances and antioxidant enzyme activity in the brain in Alzheimer’s disease. Neurology. 1995;45:1594–601.

    Article  PubMed  CAS  Google Scholar 

  20. 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.

    Article  PubMed  CAS  Google Scholar 

  21. Agostinho P, Cunha RA, Oliveira C. Neuroinflammation, oxidative stress and the pathogenesis of Alzheimer’s disease. Curr Pharm Des. 2010;16:2766–78.

    Article  PubMed  CAS  Google Scholar 

  22. Cagnin A, Brooks DJ, Kennedy AM, et al. In-vivo measurement of activated microglia in dementia. Lancet. 2001;358:461–7.

    Article  PubMed  CAS  Google Scholar 

  23. Di Bona D, Scapagnini G, Candore G, et al. Immune-inflammatory responses and oxidative stress in Alzheimer’s disease: therapeutic implications. Curr Pharm Des. 2010;16:684–91.

    Article  PubMed  Google Scholar 

  24. Brown GC. Nitric oxide and neuronal death. Nitric Oxide. 2010;23:153–65.

    Article  PubMed  CAS  Google Scholar 

  25. Ishii K, Muelhauser F, Liebl U, et al. Subacute NO generation induced by Alzheimer’s β-amyloid in the living brain: reversal by inhibition of the inducible NO synthase. FASEB J. 2000;14:1485–9.

    Article  PubMed  CAS  Google Scholar 

  26. Mori K, Togashi H, Ueno KI, et al. Aminoguanidine prevented the impairment of learning behavior and hippocampal long-term potentiation following transient cerebral ischemia. Behav Brain Res. 2001;120:159–68.

    Article  PubMed  CAS  Google Scholar 

  27. Hsu MJ, Sheu JR, Lin CH, et al. Mitochondrial mechanisms in amyloid beta peptide-induced cerebrovascular degeneration. Biochim Biophys Acta. 2010;1800:290–6.

    Article  PubMed  CAS  Google Scholar 

  28. Bertoni-Freddari C, Fattoretti P, Casoli T, et al. Neuronal apoptosis in Alzheimer’s disease: the role of age-related mitochondrial metabolic competence. Ann N Y Acad Sci. 2009;1171:18–24.

    Article  PubMed  CAS  Google Scholar 

  29. Kirsch M, Korth HG, Sustmann R, et al. The pathobiochemistry of nitrogen dioxide. Biol Chem. 2002;383:389–99.

    Article  PubMed  CAS  Google Scholar 

  30. Augusto O, Bonini MG, Amanso AM, et al. Nitrogen dioxide and carbonate radical anion: two emerging radicals in biology. Free Radic Biol Med. 2002;32:841–59.

    Article  PubMed  CAS  Google Scholar 

  31. Brennan ML, Wu W, Fu X, et al. A tale of two controversies: defining both the role of peroxidases in nitrotyrosine formation in vivo using eosinophil peroxidase and myeloperoxidase-deficient mice, and the nature of peroxidase-generated reactive nitrogen species. J Biol Chem. 2002;277:17415–27.

    Article  PubMed  CAS  Google Scholar 

  32. Souza JM, Daikhin E, Yudkoff M, et al. Factors determining the selectivity of protein tyrosine nitration. Arch Biochem Biophys. 1999;371:169–78.

    Article  PubMed  CAS  Google Scholar 

  33. Gow AJ, Duran D, Malcolm S, et al. Effects of peroxynitrite-induced protein modifications on tyrosine phosphorylation and degradation. FEBS Lett. 1996;385:63–5.

    Article  PubMed  CAS  Google Scholar 

  34. Castegna A, Thongboonkerd V, Klein JB, et al. Proteomic identification of nitrated proteins in Alzheimer’s disease brain. J Neurochem. 2003;85:1394–401.

    Article  PubMed  CAS  Google Scholar 

  35. Cenini G, Sultana R, Memo M, et al. Elevated levels of pro-apoptotic p53 and its oxidative modification by the lipid peroxidation product, HNE, in brain from subjects with amnestic mild cognitive impairment and Alzheimer’s disease. J Cell Mol Med. 2008;12:987–94.

    Article  PubMed  CAS  Google Scholar 

  36. Butterfield DA, Reed TT, Perluigi M, et al. Elevated levels of 3-nitrotyrosine in brain from subjects with amnestic mild cognitive impairment: implications for the role of nitration in the progression of Alzheimer’s disease. Brain Res. 2007;1148:243–8.

    Article  PubMed  CAS  Google Scholar 

  37. Ohtsuka K, Suzuki T. Roles of molecular chaperones in the nervous system. Brain Res Bull. 2000;53:141–6.

    Article  PubMed  CAS  Google Scholar 

  38. Kubes P, Kanwar S, Niu XF. Nitric oxide synthesis inhibition induces leukocyte adhesion via superoxide and mast cells. FASEB J. 1993;7:1293–9.

    PubMed  CAS  Google Scholar 

  39. Harry GJ, Sills R, Schlosser MJ, et al. Neurodegeneration and glia response in rat hippocampus following nitro-L-arginine methyl ester (LNAME). Neurotox Res. 2001;3:307–19.

    Article  PubMed  CAS  Google Scholar 

  40. Takuma K, Phuagphong P, Lee E, et al. Anti-apoptotic effect of cGMP in cultured astrocytes: inhibition by cGMP-dependent protein kinase of mitochondrial permeable transition pore. J Biol Chem. 2001;276:48093–9.

    PubMed  CAS  Google Scholar 

  41. Bonthius DJ, Luong T, Bonthius NE, et al. Nitric oxide utilizes NF-kappaB to signal its neuroprotective effect against alcohol toxicity. Neuropharmacology. 2009;56:716–31.

    Article  PubMed  CAS  Google Scholar 

  42. Na HJ, Chung HT, Ha KS, et al. Detection and measurement for the modification and inactivation of caspase by nitrosative stress in vitro and in vivo. Methods Enzymol. 2008;441:317–27.

    Article  PubMed  CAS  Google Scholar 

  43. Gentile MT, Vecchione C, Maffei A, et al. Mechanisms of soluble beta-amyloid impairment of endothelial function. J Biol Chem. 2004;279:48135–42.

    Article  PubMed  CAS  Google Scholar 

  44. Kalaria RN, Hedera P. Differential degeneration of the cerebral microvasculature in Alzheimer’s disease. Neuroreport. 1995;6:477–80.

    Article  PubMed  CAS  Google Scholar 

  45. Mueller XM, Tevaearai H, Chaubert P, et al. Mechanism of action of transmyocardial revascularization. Schweiz Med Wochenschr. 1999;129:1889–92 [In French].

    PubMed  CAS  Google Scholar 

  46. Johnson KA, Jones K, Holman BL, et al. Preclinical prediction of Alzheimer’s disease using SPECT. Neurology. 1998;50:1563–71.

    Article  PubMed  CAS  Google Scholar 

  47. Meier-Ruge W, Bertoni-Freddari C. The significance of glucose turnover in the brain in the pathogenic mechanisms of Alzheimer’s disease. Rev Neurosci. 1996;7:1–19.

    Article  PubMed  CAS  Google Scholar 

  48. Ferreira IL, Resende R, Ferreiro E, et al. Multiple defects in energy metabolism in Alzheimer’s disease. Curr Drug Targets. 2010;11:1193–206.

    Article  PubMed  CAS  Google Scholar 

  49. de la Torre JC, Stefano GB. Evidence that Alzheimer’s disease is a microvascular disorder: the role of constitutive nitric oxide. Brain Res Rev. 2000;34:119–36.

    Article  PubMed  Google Scholar 

  50. Duara R, Barker WW, Chang J, et al. Viability of neocortical function shown in behavioral activation state PET studies in Alzheimer disease. J Cereb Blood Flow Metab. 1992;12:927–34.

    Article  PubMed  CAS  Google Scholar 

  51. Massoud F, Gauthier S. Update on the pharmacological treatment of Alzheimer’s disease. Curr Neuropharmacol. 2010;8:69–80.

    Article  PubMed  CAS  Google Scholar 

  52. Tzourio C, Anderson C, Chapman N, et al. Effects of blood pressure lowering with perindopril and indapamide therapy on dementia and cognitive decline in patients with cerebrovascular disease. Arch Intern Med. 2003;163:1069–75.

    Article  PubMed  CAS  Google Scholar 

  53. Forette F, Seux ML, Staessen JA, et al. Systolic hypertension in Europe investigators. The prevention of dementia with antihypertensive treatment: new evidence from the systolic hypertension in Europe (Syst-Eur) study. Arch Intern Med. 2002;162:2046–52.

    Article  PubMed  Google Scholar 

  54. Deschaintre Y, Richard F, Leys D, et al. Treatment of vascular risk factors is associated with slower decline in Alzheimer disease. Neurology. 2009;73:674–80.

    Article  PubMed  Google Scholar 

  55. Francis PT, Palmer AM, Snape M, et al. The cholinergic hypothesis of Alzheimer’s disease: a review of progress. J Neurol Neurosurg Psychiatry. 1999;66:137–47.

    Article  PubMed  CAS  Google Scholar 

  56. Lipton SA. Pathologically-activated therapeutics for neuroprotection: mechanism of NMDA receptor block by memantine and S-nitrosylation. Curr Drug Targets. 2007;8:621–32.

    Article  PubMed  CAS  Google Scholar 

  57. Ferris S, Ihl R, Robert P, et al. Treatment effects of memantine on language in moderate to severe Alzheimer’s disease patients. Alzheimers Demen. 2009;5:369–74.

    Article  CAS  Google Scholar 

  58. Mecocci P, Bladstrom A, Stender K. Effects of memantine on cognition in patients with moderate to severe Alzheimer’s disease: post-hoc analyses of ADAScog and SIB total and single-item scores from six randomized, double-blind, placebo-controlled studies. Int J Geriatr Psychiatry. 2009;24:532–8.

    Article  PubMed  Google Scholar 

  59. McShane R, Areosa Sastre A, Minakaran N. Memantine for dementia. Cochrane Database Syst Rev. 2009;2:CD003154. doi:10.1002/14651858.CD003154.pub5.

    Google Scholar 

  60. Doody RS, Stevens JC, Beck C, et al. Practice parameter: management of dementia (an evidence-based review). report of the quality standards subcommittee of the American Academy of Neurology. Neurology. 2001;56:1154–66.

    Article  PubMed  CAS  Google Scholar 

  61. Lyketsos CG, Colenda CC, Beck C, et al. Position statement of the American Association for Geriatric Psychiatry regarding principles of care for patients with dementia resulting from Alzheimer disease. Am J Geriatr Psychiatry. 2006;14:561–72.

    Article  PubMed  Google Scholar 

  62. Kaduszkiewicz H, Zimmermann T, Beck-Bornholdt HP, et al. Cholinesterase inhibitors for patients with Alzheimer’s disease: systematic review of randomised clinical trials. BMJ. 2005;331:321–7.

    Article  PubMed  CAS  Google Scholar 

  63. Raina P, Santaguida P, Ismaila A, et al. Effectiveness of cholinesterase inhibitors and memantine for treating dementia: evidence review for a clinical practice guideline. Ann Intern Med. 2008;148:379–97.

    PubMed  Google Scholar 

  64. Lleo A, Greenberg SM, Growdon JH. Current pharmacotherapy for Alzheimer’s disease. Annu Rev Med. 2006;57:513–33.

    Article  PubMed  CAS  Google Scholar 

  65. McDonnell J, Redekop WK, van der Roer N, et al. The cost of treatment of Alzheimer’s disease in the Netherlands: a regression-based simulation model. Pharmacoeconomics. 2001;19:379–90.

    Article  PubMed  CAS  Google Scholar 

  66. Berghmans RL. Anti-Alzheimer drugs: ethical aspects of research and practice. Tijdschr Gerontol Geriatr. 2000;31:100–6 [In Dutch].

    PubMed  CAS  Google Scholar 

  67. Clippingdale AB, Wade JD, Barrow CJ. The amyloid-beta peptide and its role in Alzheimer’s disease. J Pept Sci. 2001;7:227–9.

    Article  PubMed  CAS  Google Scholar 

  68. Kontush A. Amyloid-beta: an antioxidant that becomes a pro-oxidant and critically contributes to Alzheimer’s disease. Free Radic Biol Med. 2001;31:1120–31.

    Article  PubMed  CAS  Google Scholar 

  69. Kakimura J, Kitamura Y, Takata K, et al. Microglial activation and amyloid-beta clearance induced by exogenous heat-shock proteins. FASEB J. 2002;16:601–3.

    PubMed  CAS  Google Scholar 

  70. Dodd PR. Excited to death: different ways to lose your neurons. Biogerontology. 2002;3:51–6.

    Article  PubMed  CAS  Google Scholar 

  71. Penner J, Rupsingh R, Smith M, et al. Increased glutamate in the hippocampus after galantamine treatment for Alzheimer disease. Prog Neuropsychopharmacol Biol Psychiatry. 2010;34:104–10.

    Article  PubMed  CAS  Google Scholar 

  72. Wild-Bode C, Yamazaki T, Capell A, et al. Intracellular generation and accumulation of amyloid beta-peptide terminating at amino acid 42. J Biol Chem. 1997;272:16085–8.

    Article  PubMed  CAS  Google Scholar 

  73. Larson EB. Prospects for delaying the rising tide of worldwide, late-life dementias. Int Psychogeriatr. 2010;22:1196–202.

    Article  PubMed  Google Scholar 

  74. Ahlemeyer B, Krieglstein J. Pharmacological studies supporting the therapeutic use of Ginkgo biloba extract for Alzheimer’s disease. Pharmacopsychiatry. 2003;36 Suppl 1:S8–14.

    PubMed  CAS  Google Scholar 

  75. Shi C, Liu J, Wu F, et al. Ginkgo biloba extract in Alzheimer’s disease: from action mechanisms to medical practice. Int J Mol Sci. 2010;11:107–23.

    Article  PubMed  CAS  Google Scholar 

  76. Chauhan NB, Sandoval J. Amelioration of early cognitive deficits by aged garlic extract in Alzheimer’s transgenic mice. Phytother Res. 2007;21:629–40.

    Article  PubMed  CAS  Google Scholar 

  77. Rezai-Zadeh K, Shytle D, Sun N, et al. Green tea epigallocatechin-3-gallate (EGCG) modulates amyloid precursor protein cleavage and reduces cerebral amyloidosis in Alzheimer transgenic mice. J Neurosci. 2005;25:8807–14.

    Article  PubMed  CAS  Google Scholar 

  78. Lee JW, Lee YK, Ban JO, et al. Green tea (-)-epigallocatechin-3-gallate inhibits beta-amyloid-induced cognitive dysfunction through modification of secretase activity via inhibition of ERK and NF-kappaB pathways in mice. J Nutr. 2009;139:1987–93.

    Article  PubMed  CAS  Google Scholar 

  79. Giunta B, Hou H, Zhu Y, et al. Fish oil enhances anti-­amyloidogenic properties of green tea EGCG in Tg2576 mice. Neurosci Lett. 2010;471:134–8.

    Article  PubMed  CAS  Google Scholar 

  80. Kalmijn S, van Boxtel MP, Ocke M, et al. Dietary intake of fatty acids and fish in relation to cognitive performance at middle age. Neurology. 2004;62:275–80.

    Article  PubMed  CAS  Google Scholar 

  81. Morris MC, Evans DA, Bienias JL, et al. Consumption of fish and n − 3 fatty acids and risk of incident Alzheimer disease. Arch Neurol. 2003;60:940–6.

    Article  PubMed  Google Scholar 

  82. Malyshev IYu, Manukhina EB. Stress, adaptation and nitric oxide. In: Pandolf BB, Takeda N, Singal PK, editors. Adaptation biology and medicine. New Delhi: Narosa Publ House; 1999. p. 375–92.

    Google Scholar 

  83. Meerson FZ. Essentials of adaptive medicine: protective effects of adaptation. Moscow: Hypoxia Medical Ltd.; 1994.

    Google Scholar 

  84. Esposito E, Cuzzocrea S. New therapeutic strategy for Parkinson’s and Alzheimer’s disease. Curr Med Chem. 2010;17:2764–74.

    Article  PubMed  CAS  Google Scholar 

  85. Ball LJ, Birge SJ. Prevention of brain aging and dementia. Clin Geriatr Med. 2002;18:485–503.

    Article  PubMed  Google Scholar 

  86. Heininger K. A unifying hypothesis of Alzheimer’s disease. IV. Causation and sequence of events. Rev Neurosci. 2000;11:213–328.

    PubMed  Google Scholar 

  87. Halagappa VK, Guo Z, Pearson M, et al. Intermittent fasting and caloric restriction ameliorate age-related behavioral deficits in the triple-transgenic mouse model of Alzheimer’s disease. Neurobiol Dis. 2007;26:212–20.

    Article  PubMed  CAS  Google Scholar 

  88. Qin W, Chachich M, Lane M, et al. Calorie restriction attenuates Alzheimer’s disease type brain amyloidosis in Squirrel monkeys (Saimiri sciureus). J Alzheimers Dis. 2006;10:417–22.

    PubMed  CAS  Google Scholar 

  89. Mattson MP. Neuroprotective signaling and the aging brain: take away my food and let me run. Brain Res. 2000;886:47–53.

    Article  PubMed  CAS  Google Scholar 

  90. Hendrie HC, Ogunniyi A, Hall KS, et al. Incidence of dementia and Alzheimer disease in 2 communities – Yoruba residing in Ibadan, Nigeria, and African Americans residing in Indianapolis, Indiana. JAMA. 2001;285:739–74.

    Article  PubMed  CAS  Google Scholar 

  91. Luchsinger JA, Tang MX, Shea S, et al. Caloric intake and the risk of Alzheimer disease. Arch Neurol. 2002;59:1258–63.

    Article  PubMed  Google Scholar 

  92. Féart C, Samieri C, Rondeau V, et al. Adherence to a Mediterranean diet, cognitive decline, and risk of dementia. JAMA. 2009;302:638–48.

    Article  PubMed  Google Scholar 

  93. Scarmeas N, Luchsinger JA, Schupf N, et al. Physical activity, diet, and risk of Alzheimer disease. JAMA. 2009;302:627–37.

    Article  PubMed  CAS  Google Scholar 

  94. Moceri VM, Kukull WA, Emanuel I, et al. Early-life risk factors and the development of Alzheimer’s disease. Neurology. 2000;554:415–20.

    Article  Google Scholar 

  95. Willis SL, Tennstedt SL, Marsiske M, et al. Long-term effects of cognitive training on everyday functional outcomes in older adults. JAMA. 2006;296:2805–14.

    Article  PubMed  CAS  Google Scholar 

  96. Buschert V, Bokde AL, Hampel H. Cognitive intervention in Alzheimer disease. Nat Rev Neurol. 2010;6:508–17.

    PubMed  CAS  Google Scholar 

  97. Addae JI, Youssef FF, Stone TW. Neuroprotective role of learning in dementia: a biological explanation. J Alzheimers Dis. 2003;5:91–104.

    PubMed  CAS  Google Scholar 

  98. Mahendra N, Arkin S. Effects of four years of exercise, language, and social interventions on Alzheimer discourse. J Commun Disord. 2003;36:395–422.

    Article  PubMed  Google Scholar 

  99. Teri L, Gibbons LE, McCurry SM, et al. Exercise plus behavioral management in patients with Alzheimer disease: a randomized controlled trial. JAMA. 2003;290:2015–22.

    Article  PubMed  CAS  Google Scholar 

  100. Laurin D, Verreault R, Lindsay J, et al. Physical activity and risk of cognitive impairment and dementia in elderly persons. Arch Neurol. 2001;58:498–504.

    Article  PubMed  CAS  Google Scholar 

  101. Lautenschlager NT, Cox KL, Flicker L, et al. Effect of physical activity on cognitive function in older adults at risk for Alzheimer disease: a randomized trial. JAMA. 2008;300:1027–37.

    Article  PubMed  CAS  Google Scholar 

  102. Malouf R, Birks J. Donepezil for vascular cognitive impairment. Cochrane Database Syst Rev. 2004;1:CD004395.

    PubMed  Google Scholar 

  103. Swaab DF, Dubelaar EJ, Scherder EJ, et al. Therapeutic strategies for Alzheimer disease: focus on neuronal reactivation of metabolically impaired neurons. Alzheimer Dis Assoc Disord. 2003;17:S114–22.

    Article  PubMed  Google Scholar 

  104. Song Y, Dowling GA, Wallhagen MI, et al. Sleep in older adults with Alzheimer’s disease. J Neurosci Nurs. 2010;42:190–8.

    Article  PubMed  Google Scholar 

  105. Zhu LL, Wang X, Wang X-M, et al. Effects of intermittent hypoxia on neurogenesis in the adult and parkinsonian rat. In: Xi L, Serebrovskaya TV, editors. Intermittent hypoxia: from molecular mechanisms to clinical applications. New York: Nova Science Publ., Inc.; 2009. p. 329–43.

    Google Scholar 

  106. Zhu XH, Yan HC, Zhang J, et al. Intermittent hypoxia promotes hippocampal neurogenesis and produces antidepressant-like effects in adult rats. J Neurosci. 2010;30:12653–63.

    Article  PubMed  CAS  Google Scholar 

  107. Meerson FZ, Pinelis VG, Koshelev VB, et al. Adaptation to periodic hypoxia restricts subdural hemorrhages in audiogenic epileptiform convulsions in rats. Biull Eksp Biol Med. 1993;116:572–4 [In Russian].

    PubMed  CAS  Google Scholar 

  108. Meerson FZ. Adaptation, stress and prophylaxis. Berlin: Springer; 1984.

    Book  Google Scholar 

  109. Jung ME, Simpkins JW, Wilson AM, et al. Intermittent hypoxia conditioning prevents behavioral deficit and brain oxidative stress in ethanol-withdrawn rats. J Appl Physiol. 2008;105:510–7.

    Article  PubMed  Google Scholar 

  110. Bernaudin M, Nedelec AS, Divoux D, et al. Normobaric hypoxia induces tolerance to focal permanent cerebral ischemia in association with an increased expression of hypoxia-inducible factor-1 and its target genes, erythropoietin and VEGF, in the adult mouse brain. J Cereb Blood Flow Metab. 2002;22:393–403.

    Article  PubMed  CAS  Google Scholar 

  111. Prass K, Scharff A, Ruscher K, et al. Hypoxia-induced stroke tolerance in the mouse is mediated by erythropoietin. Stroke. 2003;34:1981–6.

    Article  PubMed  CAS  Google Scholar 

  112. Sharp FR, Ran R, Lu A, et al. Hypoxic preconditioning protects against ischemic brain injury. NeuroRx. 2004;1:26–35.

    Article  PubMed  Google Scholar 

  113. Lin AMY. Hypoxic preconditioning protects against oxidative injury in the central nervous system. In: Xi L, Serebrovskaya TV, editors. Intermittent hypoxia: from molecular mechanisms to clinical applications. New York: Nova Science Publ., Inc.; 2009. p. 313–27.

    Google Scholar 

  114. Gulyaeva NV, Stepanichev MV, Onifriev MV, et al. Interval hypoxia training prevents oxidative stress in striatum and locomotor disturbances in a rat model of Parkinsonism. In: Fisher A, Hanln I, Yoshida M, editors. Progress in Alzheimer’s and Parkinson’s diseases. New York: Plenum Press; 1998. p. 717–23.

    Chapter  Google Scholar 

  115. Kolesnikova EE, Serebrovskaya TV. Parkinson’s disease and intermittent hypoxia training. In: Xi L, Serebrovskaya TV, editors. Intermittent hypoxia: from molecular mechanisms to clinical applications. New York: Nova Science Publ., Inc.; 2009. p. 549–60.

    Google Scholar 

  116. Gutsaeva DR, Carraway MS, Suliman HB, et al. Transient hypoxia stimulates mitochondrial biogenesis in brain subcortex by a neuronal nitric oxide synthase-dependent mechanism. J Neurosci. 2008;28:2015–24.

    Article  PubMed  CAS  Google Scholar 

  117. Lukyanova LD. Novel approach to the understanding of molecular mechanisms of adaptation to hypoxia. In: Hargens A, Takeda N, Singal PK, editors. Adaptation biology and medicine. New Delhi: Narosa Publ.; 2005. p. 1–19.

    Google Scholar 

  118. Sazontova TG, Arkhipenko YuV. Membranoprotective effects of adaptation in the heart and skeletal muscles. In: Hargens A, Takeda N, Signal PK, editors. Adaptation biology and medicine. New Delhi: Narosa Publ.; 2005. p. 112–23.

    Google Scholar 

  119. Arkhipenko IuV, Sazontova TG, Rozhitskaia II, et al. Effects of adaptation to periodic hypoxia on Ca2+ pump of the cardiac sarcoplasmic reticulum and its resistance to endogenous damaging factors. Kardiologiia. 1992;32:57–61 [In Russian].

    PubMed  Google Scholar 

  120. Manukhina EB, Downey HF, Mallet RT. Role of nitric oxide in cardiovascular adaptation to intermittent hypoxia. Exp Biol Med. 2006;231:343–65.

    CAS  Google Scholar 

  121. Xu KY, Kuppusamy SP, Wang JQ, et al. Nitric oxide protects cardiac sarcolemmal membrane enzyme function and ion active transport against ischemia-induced inactivation. J Biol Chem. 2003;278:41798–803.

    Article  PubMed  CAS  Google Scholar 

  122. Correia SC, Moreira PI. Hypoxia-inducible factor 1: a new hope to counteract neurodegeneration? J Neurochem. 2010;112:1–12.

    Article  PubMed  CAS  Google Scholar 

  123. Lukyanova LD, Dudchenko AV, Germanova EL, et al. Mitochondrial signaling in formation of body resistance to hypoxia. In: Xi L, Serebrovskaya TV, editors. Intermittent hypoxia: from molecular mechanisms to clinical applications. New York: Nova Science Publ., Inc.; 2009. p. 391–417.

    Google Scholar 

  124. Heidbreder M, Frolich F, Johren O, et al. Hypoxia rapidly activates HIF-3α mRNA expression. FASEB J. 2003;10:1096–115.

    Google Scholar 

  125. Sazontova TG, Arkhipenko YuV. Intermittent hypoxia in resistance of cardiac membrane structures: role of reactive oxygen species and redox signaling. In: Xi L, Serebrovskaya TV, editors. Intermittent hypoxia: from molecular mechanisms to clinical applications. New York: Nova Science Publ., Inc.; 2009. p. 113–50.

    Google Scholar 

  126. Soucek T, Cumming R, Dargusch R, et al. The regulation of glucose metabolism by HIF-1 mediates a neuroprotective response to amyloid beta peptide. Neuron. 2003;39:43–56.

    Article  PubMed  CAS  Google Scholar 

  127. Wakutani Y, Urakami K, Shimomura T, et al. Heat shock protein 70 mRNA levels in mononuclear blood cells from patients with dementia of the Alzheimer type. Dementia. 1995;6:301–5.

    PubMed  CAS  Google Scholar 

  128. Behl C, Schubert D. Heat shock partially protects rat pheochromocytoma PC12 cells from amyloid beta peptide toxicity. Neurosci Lett. 1993;154:1–4.

    Article  PubMed  CAS  Google Scholar 

  129. Yao S, Peng M, Zhu X, et al. Heat shock protein 72 protects hippocampal neurons from apoptosis induced by chronic psychological stress. Int J Neurosci. 2007;117:1551–64.

    Article  PubMed  CAS  Google Scholar 

  130. Brown IR. Heat shock proteins and protection of the nervous system. Ann N Y Acad Sci. 2007;1113:147–58.

    Article  PubMed  CAS  Google Scholar 

  131. Malyshev IY, Wiegant FA, Mashina SY, et al. Possible use of adaptation to hypoxia in Alzheimer’s disease: a hypothesis. Med Sci Monit. 2005;11:HY31–8.

    PubMed  CAS  Google Scholar 

  132. Malyshev IYu, Manukhina EB. Stress proteins in Alzheimer’s disease. Vestn Ross Akad Med Nauk. 2005;7:40–6 [In Russian].

    PubMed  Google Scholar 

  133. Malyshev IY, Zenina TA, Golubeva LY, et al. NO-dependent mechanisms of adaptation to hypoxia. Nitric Oxide. 1999;3:105–13.

    Article  PubMed  CAS  Google Scholar 

  134. Goryacheva AV, Kruglov SV, Pshennikova MG, et al. Adaptation to intermittent hypoxia restricts nitric oxide overproduction and prevents beta-amyloid toxicity in rat brain. Nitric Oxide. 2010;23:289–99.

    Article  PubMed  CAS  Google Scholar 

  135. Belykh AG, Gukasov VM, Chukaev SA. State of the free-radical oxidation system in normobaric hypoxia. Fiziol Zhurnal. 1992;38:73–6 [In Russian].

    CAS  Google Scholar 

  136. Manukhina EB, Pshennikova MG, Goryacheva AV, et al. Role of nitric oxide in prevention of cognitive disorders in neurodegenerative brain injuries in rats. Bull Exp Biol Med. 2008;146:391–5.

    Article  PubMed  CAS  Google Scholar 

  137. Manukhina EB, Goryacheva AV, Barskov IV, et al. Prevention of neurodegenerative damage to the brain in rats in experimental Alzheimer’s disease by adaptation to hypoxia. Neurosci Behav Physiol. 2010;40:737–43.

    Article  PubMed  CAS  Google Scholar 

  138. Asha Devi S, Subramanyam MV, Vani R, et al. Adaptations of the antioxidant system in erythrocytes of trained adult rats: impact of intermittent hypobaric-hypoxia at two altitudes. Comp Biochem Physiol C Toxicol Pharmacol. 2005;140:59–67.

    Article  PubMed  CAS  Google Scholar 

  139. El’chaninova SA, Koreniak NA, Smagina IV, et al. Intermittent hypoxia in the treatment of dyscirculatory encephalopathy. Zh Nevrol Psikhiatr Im SS Korsakova. 2002;102:29–32 [In Russian].

    Google Scholar 

  140. Cardinali DP, Brusco LI, Liberczuk C, et al. The use of melatonin in Alzheimer’s disease. Neuro Endocrinol Lett. 2002;1(suppl):20–3.

    Google Scholar 

  141. Sánchez-Barceló EJ, Mediavilla MD, Tan DX, et al. Clinical uses of melatonin: evaluation of human trials. Curr Med Chem. 2010;17:2070–95.

    Article  PubMed  Google Scholar 

  142. 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 N Y Acad Sci. 1999;890:471–85.

    Article  PubMed  CAS  Google Scholar 

  143. Pappolla MA, Sos M, Omar RA, et al. The heat shock/oxidative stress connection. Relevance to Alzheimer disease. Mol Chem Neuropathol. 1996;28:21–34.

    Article  PubMed  CAS  Google Scholar 

  144. Wang X. The antiapoptotic activity of melatonin in neurodegenerative diseases. CNS Neurosci Ther. 2009;15:345–57.

    Article  PubMed  CAS  Google Scholar 

  145. Kaur C, Srinivasan KN, Singh J, et al. Plasma melatonin, pinealocyte morphology, and surface receptors/antigen expression on macrophages/microglia in the pineal gland following a high-altitude exposure. J Neurosci Res. 2002;67:533–43.

    Article  PubMed  CAS  Google Scholar 

  146. LaManna JC, Chavez JC, Pichiule P. Structural and functional adaptation to hypoxia in the rat brain. J Exp Biol. 2004;207:3163–9.

    Article  PubMed  CAS  Google Scholar 

  147. Turek Z, Hoofd L, Rakusan K. Myocardial capillaries and tissue oxygenation. Can J Cardiol. 1986;2:98–103.

    PubMed  CAS  Google Scholar 

  148. Wagner PD. Skeletal muscle angiogenesis. A possible role for hypoxia. Adv Exp Med Biol. 2001;502:21–38.

    PubMed  CAS  Google Scholar 

  149. Lauro KL, LaManna JC. Adequacy of cerebral vascular remodeling following three weeks of hypobaric hypoxia. Examined by an integrated composite analytical model. Adv Exp Med Biol. 1997;411:369–76.

    Article  PubMed  CAS  Google Scholar 

  150. Harik N, Harik SI, Kuo N-T, et al. Time course and reversibility of the hypoxia-induced alterations in cerebral vascularity and cerebral capillary glucose transporter density. Brain Res. 1996;737:335–8.

    Article  PubMed  CAS  Google Scholar 

  151. Harik SI, Hritz MA, LaManna JC. Hypoxia-induced brain angiogenesis in the adult rat. J Physiol (Lond). 1995;485:525–30.

    CAS  Google Scholar 

  152. Boero JA, Ascher J, Arregui A, et al. Increased brain capillaries in chronic hypoxia. J Appl Physiol. 1999;86:1211–9.

    PubMed  CAS  Google Scholar 

  153. Bell RD, Zlokovic BV. Neurovascular mechanisms and blood-brain barrier disorder in Alzheimer’s disease. Acta Neuropathol. 2009;118:103–13.

    Article  PubMed  CAS  Google Scholar 

  154. Brown WR, Moody DM, Thore CR, et al. Microvascular changes in the white mater in dementia. J Neurol Sci. 2009;283:28–31.

    Article  PubMed  Google Scholar 

  155. Farkas E, De Jong GI, Apró E, et al. Similar ultrastructural breakdown of cerebrocortical capillaries in Alzheimer’s disease, Parkinson’s disease, and experimental hypertension. Ann N Y Acad Sci. 2000;903:72–82.

    Article  PubMed  CAS  Google Scholar 

  156. Hanyu H, Sato T, Hirao K, et al. The progression of cognitive deterioration and region al cerebral blood flow patterns in Alzheimer’s disease: a longitudinal SPECT study. J Neurol Sci. 2010;290:96–101.

    Article  PubMed  Google Scholar 

  157. Fischer VW, Siddiqi A, Yusufaly Y. Altered angioarchitecture in selected areas of brains with Alzheimer’s disease. Acta Neuropathol. 1990;79:672–9.

    Article  PubMed  CAS  Google Scholar 

  158. Chávez JC, Agani F, Pichiule P, et al. Expression of hypoxic inducible factor 1alpha in the brain of rats during chronic hypoxia. J Appl Physiol. 2000;89:1937–42.

    PubMed  Google Scholar 

  159. Pichiule P, LaManna JC. Angiopoietin-2 and rat brain capillary remodeling during adaptation and deadaptation to prolonged mild hypoxia. J Appl Physiol. 2002;93:1131–9.

    PubMed  CAS  Google Scholar 

  160. Black JE, Polinsky M, Greenough WT. Progressive failure of cerebral angiogenesis supporting neural plasticity in aging rats. Neurobiol Aging. 1989;10:353–8.

    Article  PubMed  CAS  Google Scholar 

  161. Rivard A, Fabre JE, Silver M, et al. Age-dependent impairment of angiogenesis. Circulation. 1999;99:111–20.

    Article  PubMed  CAS  Google Scholar 

  162. Brown WR, Thore CR. Review: cerebral microvascular pathology in aging and neurodegeneration. Neuropathol Appl Neurobiol. 2011;37:56–74.

    Article  PubMed  CAS  Google Scholar 

  163. Rivard A, Berthou-Soulie L, Principe N, et al. Age-dependent defect in vascular endothelial growth factor expression is associated with reduced hypoxia-inducible factor 1 activity. J Biol Chem. 2000;275:29643–7.

    Article  PubMed  CAS  Google Scholar 

  164. Rapino C, Bianchi G, Di Giulio GC, et al. HIF-1alpha cytoplasmic accumulation is associated with cell death in old rat cerebral cortex exposed to intermittent hypoxia. Aging Cell. 2005;4:177–85.

    Article  PubMed  CAS  Google Scholar 

  165. Manukhina EB, Sokolova IA, Rodionov IM. Changes in density of the vascular network on brain surface in rats in experiments hypertension and adaptation to high altitude. Kardiologiya. 1982;10:1982 [In Russian].

    Google Scholar 

  166. Prewitt RL, Cardoso SS, Wood WB. Prevention of arteriolar rarefaction in the spontaneously hypertensive rat by exposure to simulated high altitude. J Hypertens. 1986;4:735–40.

    Article  PubMed  CAS  Google Scholar 

  167. LaManna JC, Vendel LM, Farrell RM. Brain adaptation to chronic hypobaric hypoxia in rats. J Appl Physiol. 1992;72:2238–43.

    PubMed  CAS  Google Scholar 

  168. Calabrese V, Boyd-Kimball D, Scapagnini G, et al. Nitric oxide and cellular stress response in brain aging and neurodegenerative disorders: the role of vitagenes. In Vivo. 2004;18:245–67.

    PubMed  CAS  Google Scholar 

  169. Takahashi K, Hara E, Suzuki H, et al. Expression of heme oxygenase isozyme mRNAs in the human brains and induction of heme oxygenase-1 by nitric oxide donors. J Neurochem. 1996;67:482–9.

    Article  PubMed  CAS  Google Scholar 

  170. Kurauchi Y, Hisatsune A, Isohama Y, et al. Nitric oxide-cyclic GMP signaling pathway limits inflammatory degeneration of midbrain dopaminergic neurons: cell type-specific regulation of heme oxygenase-1 expression. Neuroscience. 2009;158:856–66.

    Article  PubMed  CAS  Google Scholar 

  171. McCarty MF. Vascular nitric oxide may lessen Alzheimer’s risk. Med Hypotheses. 1998;51:465–76.

    Article  PubMed  CAS  Google Scholar 

  172. Cellermajer DS. Endothelial dysfunction: does it matter? Is it reversible? J Am Coll Cardiol. 1997;30:325–53.

    Article  Google Scholar 

  173. Pedrazachaverri J, Tapia E, Medinacampos ON, et al. Garlic prevents hypertension induced by chronic inhibition of nitric oxide synthesis. Life Sci. 1998;62:PL71–7.

    Article  CAS  Google Scholar 

  174. Manukhina EB, Malyshev IYu. Role of nitric oxide in protective effects of adaptation. In: Moravec J, Takeda N, Singal PK, editors. Adaptation biology and medicine. New Delhi: Narosa Publishing House; 2002. p. 312–27.

    Google Scholar 

  175. Lange-Asschenfeldt C, Kojda G. Alzheimer’s disease, cerebrovascular dysfunction and the benefits of exercise: from vessels to neurons. Exp Gerontol. 2008;43:499–504.

    Article  PubMed  CAS  Google Scholar 

  176. Chung KKK, David KK. Emerging roles of nitric oxide in neurodegeneration. Nitric Oxide. 2010;22:290–5.

    Article  PubMed  CAS  Google Scholar 

  177. Ghosh C, Lahiri DK. Increased vulnerability of neuronal cell lines to sodium nitroprusside-mediated toxicity is caused by the decreased level of nitric oxide metabolites. J Mol Neurosci. 1999;13:77–92.

    Article  PubMed  CAS  Google Scholar 

  178. Wilcock DM, Lewis MR, Van Nostrand WE, et al. Progression of amyloid pathology to Alzheimer’s disease pathology in an amyloid precursor protein transgenic mouse model by removal of nitric oxide synthase 2. J Neurosci. 2008;28:1537–45.

    Article  PubMed  CAS  Google Scholar 

  179. Tarkowski E, Ringqvist A, Blennow K, et al. Intrathecal release of nitric oxide in Alzheimer’s disease and vascular dementia. Dement Geriatr Cogn Disord. 2000;11:322–6.

    Article  PubMed  CAS  Google Scholar 

  180. Pshennikova MG, Popkova EV, Khomenko IP, et al. Resistance to neurodegenerative brain damage in August and Wistar rats. Bull Exp Biol Med. 2005;139:540–2.

    Article  PubMed  CAS  Google Scholar 

  181. Pshennikova MG, Popkova EV, Pokidyshev DA, et al. The effects of adaptation to hypoxia on the resistance to neurodegenerative disorders in the brain of rats of different genetic strains. Vestn Ross Akad Med Nauk. 2007;2:50–5 [In Russian].

    PubMed  Google Scholar 

  182. Manukhina EB, Goryacheva AV, Barskov IV, et al. Nitric oxide prevents cognitive disorders and neurodegeneration in rats with experimental Alzheimer’s disease. FASEB J. 2010;24:568.4.

    Google Scholar 

  183. Mikoyan VD, Kubrina LN, Manukhina EB, et al. Differential stimulation of NO synthesis by heat shock in rats of genetically different populations. Bull Exp Biol Med. 1996;6:634–7 [In Russian].

    Google Scholar 

  184. Niwa K, Kazama K, Younkin L, et al. Cerebrovascular autoregulation is profoundly impaired in mice overexpressing amyloid precursor protein. Am J Physiol. 2002;283:H315–23.

    CAS  Google Scholar 

  185. Bartenstein P, Minoshima S, Hirsch C, et al. Quantitative assessment of cerebral blood flow in patients with Alzheimer’s disease by SPECT. J Nucl Med. 1997;38:1095–101.

    PubMed  CAS  Google Scholar 

  186. Zhang F, Eckman C, Younkin S, et al. Increased susceptibility to ischemic brain damage in transgenic mice overexpressing the amyloid precursor protein. J Neurosci. 1997;17:7655–61.

    PubMed  CAS  Google Scholar 

  187. Mashina SY, Aleksandrin VV, Goryacheva AV, et al. Adaptation to hypoxia prevents disturbances in cerebral blood flow during neurodegenerative process. Bull Exp Biol Med. 2006;142:169–72.

    Article  PubMed  CAS  Google Scholar 

  188. Dede DS, Yavuz B, Yavuz BB, et al. Assessment of endothelial function in Alzheimer’s disease: is Alzheimer’s disease a vascular disease? J Am Geriatr Soc. 2007;55:1613–7.

    Article  PubMed  Google Scholar 

  189. Manukhina EB, Vanin AF, Malyshev IYu, et al. Intermittent hypoxia-induced cardio- and vasoprotection: role of NO stores. In: Xi L, Serebrovskaya TV, editors. Intermittent hypoxia: from molecular mechanisms to clinical applications. New York: Nova Science Publ., Inc.; 2009. p. 113–46.

    Google Scholar 

  190. Vlasova MA, Vanin AF, Muller B, et al. Detection and description of various stores of nitric oxide store in vascular wall. Bull Exp Biol Med. 2003;136:226–30.

    Article  PubMed  CAS  Google Scholar 

  191. Manukhina EB, Malyshev IYu, Smirin BV, et al. Production and storage of nitric oxide in adaptation to hypoxia. Nitric Oxide. 1999;3:393–401.

    Article  PubMed  CAS  Google Scholar 

  192. Vlasova MA, Smirin BV, Pokidyshev DA, et al. Mechanism of adaptation of the vascular system to chronic changes in nitric oxide level in the organism. Bull Exp Biol Med. 2006;142:670–4.

    Article  PubMed  CAS  Google Scholar 

  193. Wallace MN, Geddes JG, Farquhar DA, et al. Nitric oxide synthase in reactive astrocytes adjacent to β-amyloid plaques. Exp Neurol. 1997;144:266–72.

    Article  PubMed  CAS  Google Scholar 

  194. de la Monte SM, Lu BX, Sohn YK, et al. Aberrant expression of nitric oxide synthase III in Alzheimer’s disease: relevance to cerebral vasculopathy and neurodegeneration. Neurobiol Aging. 2000;21:309–19.

    Article  PubMed  Google Scholar 

  195. Lüth HJ, Holzer M, Gärtner U, et al. Expression of endothelial and inducible NOS-isoforms is increased in Alzheimer’s disease, in APP23 transgenic mice and after experimental brain lesion in rat: evidence for an induction by amyloid pathology. Brain Res. 2001;913:57–67.

    Article  PubMed  Google Scholar 

  196. Gabbott PL, Bacon SJ. Localisation of NADPH diaphorase activity and NOS immunoreactivity in astroglia in normal adult rat brain. Brain Res. 1996;714:135–44.

    Article  PubMed  CAS  Google Scholar 

  197. Förstermann U, Kleinert H. Nitric oxide synthase expression and expressional control of the three isoforms. Naunyn Schmiedebergs Arch Pharmacol. 1995;352:351–64.

    Article  PubMed  Google Scholar 

  198. Aguilera P, Chánez-Cárdenas ME, Floriano-Sánchez E, et al. Time-related changes in constitutive and inducible nitric oxide synthases in the rat striatum in a model of Huntington’s disease. Neurotoxicology. 2007;28:1200–7.

    Article  PubMed  CAS  Google Scholar 

  199. Fernández-Vizarra P, Fernández AP, Castro-Blanco S, et al. Expression of nitric oxide system in clinically evaluated cases of Alzheimer’s disease. Neurobiol Dis. 2004;15:287–305.

    Article  PubMed  CAS  Google Scholar 

  200. Lüth HJ, Münch G, Arendt T. Aberrant expression of NOS isoforms in Alzheimer’s disease is structurally related to nitrotyrosine formation. Brain Res. 2002;953:135–43.

    Article  PubMed  Google Scholar 

  201. Smith MA, Richey Harris PL, Sayre LM, et al. Widespread peroxynitrite-mediated damage in Alzheimer’s disease. J Neurosci. 1997;17:2653–7.

    PubMed  CAS  Google Scholar 

Download references

Acknowledgments

This work was supported by the Russian Foundation for Basic Research (grant 07–04–00650).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Eugenia B. Manukhina .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2012 Springer-Verlag London

About this chapter

Cite this chapter

Manukhina, E.B., Goryacheva, A.V., Pshennikova, M.G., Malyshev, I.Y., Mallet, R.T., Downey, H.F. (2012). Protective Effects of Adaptation to Hypoxia in Experimental Alzheimer’s Disease. In: Xi, L., Serebrovskaya, T. (eds) Intermittent Hypoxia and Human Diseases. Springer, London. https://doi.org/10.1007/978-1-4471-2906-6_13

Download citation

  • DOI: https://doi.org/10.1007/978-1-4471-2906-6_13

  • Published:

  • Publisher Name: Springer, London

  • Print ISBN: 978-1-4471-2905-9

  • Online ISBN: 978-1-4471-2906-6

  • eBook Packages: MedicineMedicine (R0)

Publish with us

Policies and ethics