Skip to main content

MAO-B Inhibitors in Neurological Disorders with Special Reference to Selegiline

  • Chapter
Book cover Inhibitors of Monoamine Oxidase B

Part of the book series: Milestones in Drug Therapy ((MDT))

Abstract

It has been shown that monoamines like serotonin and noradrenaline are preferentially metabolized in man by MAO-A, while benzylamine, phenylethylamine, and phenylethanolamine are specific substrates for MAO-B. Other monoamines are metabolized by both forms, but regional differences exist [1]. In the intestine, MAO-A is the dominant form, while MAO-B is preferentially found in platelets and in the brain, especially in the glial cells [2] (see Chapters 2, 6, 9).

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 49.99
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 69.99
Price excludes VAT (USA)
  • Compact, lightweight 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

Preview

Unable to display preview. Download preview PDF.

Unable to display preview. Download preview PDF.

References

  1. Kabins D, Gershon S. Potential applications for monoamine oxidase inhibitors. Dementia 1990; 1: 323–348.

    Google Scholar 

  2. Riederer P, Youdim MBH, Rausch WD. On the mode of action of l-deprenyl in the human nervous system. J Neural Transm 1978; 43: 217–226.

    Google Scholar 

  3. Finberg JPM. Effects of selective inhibition of MAO types A and B on peripheral symphatetic function. In: Kamijo K, Usdin E, Nagatsu, editors. Monoamine oxidase: Basic and clinical frontiers. Princeton: Excerpta Medica 1982, 174–182.

    Google Scholar 

  4. Green AR, Mitchell BD, Tordoff A, Youdim MBH. Evidence for dopamine deamina-tion by both type A and B monoamine oxidase in rat brain in vivo and for the degree of inhibition of enzyme necessary for increased functional activity of dopamine and 5-hydroxytryptamine. Br J Pharmac 1977; 60: 343.

    Google Scholar 

  5. Demarest KT, Smith DJ, Azzaro AJ. The presence of type A form of monoamine oxidase within nigrostriatal dopamine containing neurons. J Pharmac Exp Ther 1980; 215:461–468.

    Google Scholar 

  6. Oreland, L, Arai Y, Stenström A. The effect of deprenyl (selegiline) on intra- and extraneuronal dopamine oxidation. Acta Neurol Scand 1983; 95 (Supplement): 81–95.

    Google Scholar 

  7. Konradi C, Kornhuber J, Froelich L, Fritze J, Heinsen H, Beckmann H, Schulz E, Riederer P. Demonstration of monoamine oxidase-A and B in the human brainstem by a histochemical technique. Neuroscience 1989; 33: 383–400.

    Google Scholar 

  8. Liccione J, Azzaro AJ. Different roles for type A and type B monoamine oxidase in regulating synaptic dopamine at D-l and D-2 receptors associated with adenosine-3’,5’-cyclic monophosphate (cyclic AMP) formation. Naunyn-Schmiedeberg’s Arch Pharmacol 1988; 337: 151–158.

    Google Scholar 

  9. Riederer P, Youdim MBH. Monomaine oxidase activity and monoamine metabolism in brains of Parkinsonian patients treated with l-deprenyl. J Neurochem 1986; 46: 1359–1365.

    Google Scholar 

  10. Youdim MBH, Finberg JPM. New directions in monomaine oxidase A and B selective inhibitors and substrates. Biochem Pharmacol 1990; 41: 155–162.

    Google Scholar 

  11. Ransom RW, Deschenes NL. Poly amines regulate glycine interaction with the N- methyl-D-aspartate receptor. Synapse 1990; 5: 294.

    Google Scholar 

  12. Sacaan AI, Johnson KM. Spermine enhances binding to the glycine site associated with the NMDA receptor complex. Mol Pharmacol 1989; 36: 758.

    Google Scholar 

  13. Ransom RW. Polyamine and ifenprodil interactions with the NMDA receptor’s glycine site. Eur J Pharmacol 1991; 208: 67–71.

    Google Scholar 

  14. Smith GS, Reid RA. The influence of the respiratory state on monoamine oxidase activity in the rate liver mitchondria. Biochem J 1978; 176: 1011–1014.

    Google Scholar 

  15. Traystman RJ, Gurtner GH, Koehler RC, Jones MD, Rogers MC. Central chemore-ceptor and oxygenase regulation of cerebral blood flow. J Cerebral Blood Flow Metab 1983; 3: 180–181.

    Google Scholar 

  16. Kozniewska E, Weiler L, Höper J, Harrison DK, Kessler M. Cerebrocortical microcirculation in different stages of hypoxic hypoxia. J Cerebral Blood Flow Metab 1987; 7: 464–470.

    Google Scholar 

  17. Höper J, Kozniewska E. Attentuation of hypoxic response in cerebral microcirculation following deprenyl. Int J Microcirc. 1992.

    Google Scholar 

  18. Globus MYT, Ginsberg MD, Dietrich WE, Busto R, Scheinberg P. Substantia nigra lesions protects against ischemic damage in the striatum. Neurosci Lett 1987; 80: 251–256.

    Google Scholar 

  19. Kumagae Y, Matsui Y, Iwata N. Participation of type A monoamine oxidase in the activated deamination of brain monoamines shortly after perfusion in rats. Jpn J Pharmacol 1990; 54: 407–413.

    Google Scholar 

  20. Damsa G, Boisvert DP, Mudrick LA, Wenkstein D, Fibiger HC. Effects of transient forebrain ischemia and pargyline on extracellular concentrations of dopamine, serotonin, and their metabolites in the rat striatum as determined by in vivo microdialysis. J Neurochem 1990; 54: 801–808.

    Google Scholar 

  21. Tatyanenko LV, Raikhman LM, Gorkin VZ. Type B monoamine oxidase and function of Ca2+, Mg2+-dependent adenosine tri-phosphate in preparations from sarcoplastic reticulum vesicles. Byulleten Eksperimental noi Biologii Meditsiny 1977; 83: 283–284.

    Google Scholar 

  22. Robinson DS, Nies A, Davis JN, Bunney WE, Dévies JM, Colbuin RW. Ageing, monoamines, and monoamine levels. Lancet 1972; i: 290–291.

    Google Scholar 

  23. Sparks DL, Woeltz van M, Markesberry WR. Alterations of brain monoamine oxidase activity in ageing, Alzheimer’s disease and Pick’s disease. Arch Neurol 1991; 48: 718–721.

    Google Scholar 

  24. Fowler CJ, Wiberg A, Oreland L, Marcusson J, Winblad B. The effect of age on the activity and molecular properties of human brain monoamine oxidase. J Neural Transm 1990; 49: 1–20.

    Google Scholar 

  25. Jossan SS, Gilberg PG, Karlsson I, Gottfries CG, Oreland L. Visualisation of brain monoamine oxidase B (MAO-B) in dementia of the Alzheimer’s type of means of large cryosection autoradiography; a pilot study. J Neural Transm 1990; (Supplement 32): 61–65.

    Google Scholar 

  26. Oreland L, Fowler CJ, Carlsson A, Magnusson T. Monoamine oxidase A- and B activity in the rat brain after hemitransection. Life Sci 1990; 26: 139–146.

    Google Scholar 

  27. Cohen G. Oxygen radicals and Parkinson’s disease. Upjohns Symposium/Oxygen Radicals April 1987; 130–135.

    Google Scholar 

  28. Perry TL, Godin DV, Hansen S. Parkinson’s disease: A disorder due to nigral glutathione deficiency? Neurosci Lett 1982; 33: 305–310.

    Google Scholar 

  29. Riederer P, Sofic E, Rausch W-D, Schmidt B, Reynolds GP, Jellinger K. Transition metals, ferritin, glutathione, and ascorbic acid in parkinsonian brains. J Neurochem 1989; 52: 515–520.

    Google Scholar 

  30. Burns RS. Subclinical damage of the nigrostriatal dopamine system by MPTP as a model of preclinical Parkinson’s disease: a review. Acta Neurol Scand 1991; 136 (Supplement 84): 29–36.

    Google Scholar 

  31. Heikkila RE, Terleckyj I, Sieber BA. Monoamine oxidase and the bioactivation of MPTP and related neurotoxins: relevance to DATATOP. J Neural Transm 1990; 32: 217–227.

    Google Scholar 

  32. West ED, Dally PJ. Effect of iproniazid in depressive syndromes. Br Med J 1959; 1: 1491–1494.

    Google Scholar 

  33. Sanchez-Ramos, JR. Banisterine and Parkinson’s disease. Clin Neuropharmacol 1991; 14: 391–402.

    Google Scholar 

  34. Squires R. Multiple forms of monoamine oxidase in intact mitochondria as characterized by selective inhibitors and thermal stability. Adv Biochem Psychopharmacol 1972; 5: 335–370.

    Google Scholar 

  35. Johnston JP. Some observations upon a new inhibitor of monoamine oxidase in brain tissue. Biochem Pharmacol 1968; 17: 335–370.

    Google Scholar 

  36. Knoll J, Magyar K. Some puzzling pharmacological effects of monoamine oxidase inhibitors. Adv Biochem Psychopharmacol 1972; 5: 393–408.

    Google Scholar 

  37. Knoll J, Ecseri Z, Kelemen K, Nievel J, Knoll B. Phenylisopropylmethylpropinylamine (E-250), a new spectrum psychic energizer. Arch Int Pharmacodyn 1965; 155: 154–164.

    Google Scholar 

  38. Mann J, Gershon S. l-Deprenyl: A selective monoamine oxidase-B inhibitor in endogenous depression. Life Sci 1980; 26: 877–882.

    Google Scholar 

  39. Mann J, Frances A, Kaplan RD, Kocsic J, Peselow ED, Gershon S. The relative efficacy of l-deprenyl, a selective monoamine oxidase type B inhibitor, in endogenous and nonendogenous depression. J Clin Psychopharmacol 1982; 2: 54–57.

    Google Scholar 

  40. Quitkin FM, Leibowitz MR, Stewart W, McGrath PJ, Harrison W, Rabkin JG. l-Deprenyl in atypical depressives. Arch Gen Psychiarty 1983; 142: 508–511.

    Google Scholar 

  41. Wender PH. Minimal brain dysfunction: An overview. In: Lipton MA, DiMascio A, Killam KF, editors. Psychopharmacol: A generation of progress. New York: Raven Press, 1978: 1429–1435.

    Google Scholar 

  42. Mann J, Aarons SF, Wilner PJ. A controlled study of the antidepressant efficacy and side effects of deprenyl: A selective monoamine oxidase inhibitor. Arch Gen Psychiatry 1989; 46: 45–50.

    Google Scholar 

  43. Ichikawa J, Meltzer H. The effect of clozapine and haloperidol on the basal dopamine release and metabolism in the rat striatum and nucleus accumbens studied by in vivo microdialysis. Eur J Pharmacol 1990; 176: 371–374.

    Google Scholar 

  44. Cadet JL, Lohr JB. Possible involvement of free radicals in neuroleptic-induced movement disorders. In: Vitamin E: Biochemistry and health implications. Ann NY Acad Sci 1989; 570: 176–185.

    Google Scholar 

  45. Lai S, Merbtiz LP, Grip JG. Modification of function in headinjured patients with sinemet. Brain Inj 1988; 2: 225–233.

    Google Scholar 

  46. Gaultieri T, Chandler M, Coons T, Brown LT. Amantadine: A new clinical profile for traumatic brain injury. Clin Neuropharmacol 1989; 12: 258–270.

    Google Scholar 

  47. Hardy J, Allsop D. Amyloid deposition as the central event in the etiology of Alzheimer’s disease. TiPS 1991; 12: 383–388.

    Google Scholar 

  48. Tariot PN, Sunderland T, Cohen RM, Newhouse PA, Mueller EA, Murphy DL. Tranylcypromine compared with l-deprenyl in Alzheimer’s disease. J Clin Psychopharmacol 1988; 8: 23–27.

    Google Scholar 

  49. Tariot PH, Cohen RM, Sunderland T, Newhouse PA, Yount D, Mellow A. l-Deprenyl in Alzheimer’s disease. Arch Gen Psychiat 1987; 44: 427–433.

    Google Scholar 

  50. Sunderland T, Tariot PH, Cohen RM, Newhouse PA, Mellow A, Mueller EA. Dose-dependent effects of deprenyl on CSF monoamine metabolites in patients with Alzheimer’s disease. Psychopharmacol 1987; 91: 293–296.

    Google Scholar 

  51. Agnoli A, Martucci N, Fabbrini G, Buckley AE, Fioravanti M. Monomaine oxidase and dementia: Treatment with an inhibitor of MAO-B activity. Dementia 1990; 1: 109–114.

    Google Scholar 

  52. Campi N, Todeschini GP, Scarella L. Selegiline versus l-acetylcarnitine in the treatment of Alzheimer-type dementia. Clin Therap 1990; 12 (4): 306–314.

    Google Scholar 

  53. Martignoni E, Bono G, Blandini F, Sinforiani E, Merlo P, Nappi G. Monoamines and related metabolite levels in the cerebrospinal fluid of patients with dementia of Alzheimer-type. Influence of treatment with l-deprenyl. J Neural Transm (PDSect) 1991; 3: 15–25.

    Google Scholar 

  54. Monteverde A, Gnemmi P, Rossi F, Monteverde A, Finali GC. Selegiline in the treatment of mild or moderate Alzheimer-type dementia. Clin Therap 1990; 12: 315–322.

    Google Scholar 

  55. Piccinin GL, Finali G, Piccirilli M. Neuropsychological effects of l-deprenyl in Alzheimer’s type dementia. Clin Neuropharmacol 1990; 12 (2): 147–163.

    Google Scholar 

  56. Schneider LS, Pollock VE, Zemansky MF, Gleason RP, Palmer R, Sloane RB. A pilot study of low-dose l-deprenyl in Alzheimer’s disease. J Geriatric Psych Neurol 1991; 4: 143–148.

    Google Scholar 

  57. Knoll J. The striatal dopamine dependency of life span in male rats. Longevity study with (-)-deprenyl. Mech Ageing Dev 1988; 46: 237–262.

    Google Scholar 

  58. Milgram NW, Racine RJ, Nellis P, Mendonca A, Ivy GO. Effect of selective monoamine oxidase inhibitors on the morphine-induced hypothermia in retrained rats. Gen Pharmac 1987; 18: 185–188.

    Google Scholar 

  59. Birkmayer W, Knoll J, Riederer P, Youdin MBH, Hars V. Increased life expectancy resulting from addition of l-deprenyl to Madopar treatment in Parkinson’s disease: a long term study. J Neural Transm 1985; 64: 113–127.

    Google Scholar 

  60. Birkmayer W, Riederer P, Youdim MBH, Linauer W. The potentiation of the anti-akinetic effect of L-dopa treatment by the inhibitor of MAO-B, deprenyl. J Neural Transm 1975; 36: 303–326.

    Google Scholar 

  61. Riederer P, Youdim MBH, Rausch WD, Birkmayer W, Sellinger K. On the mode of action of l-deprenyl in human central nervous system. J Neural Transm 1978; 43: 217–226.

    Google Scholar 

  62. Stern GM, Elsworth JD, Sandler M, Lees AS, Kohout LS, Shaw KM. Deprenyl in Parkinson’s disease. Lancet 1977; i: 791–796.

    Google Scholar 

  63. Birkmayer W, Riederer P, Ambrozi L. Implications of combined treatment with Madopar and l-deprenyl in Parkinson’s disease. Lancet 1977; i: 439–444.

    Google Scholar 

  64. Broderson P, Philbert A, Gulliksen G, Stigard A. The effect of l-deprenyl on the on-off phenomena in Parkinson’s disease. Acta Neurol Scand 1985; 71: 494.

    Google Scholar 

  65. Csanda E, Antal J, Antony M, Csanady A. Experience with l-deprenyl in parkinsonism. J Neural Transm 1978; 43: 263–269.

    Google Scholar 

  66. Rinne UK, Siirtola T, Sonninen V. l-Deprenyl treatment of on-off-phenomena in Parkinson’s disease. J Neural Transm 1978; 43: 253–262.

    Google Scholar 

  67. Presthus J, Hajba A. Deprenyl combined with levodopa and a decarboxylase inhibitor in the treatment of Parkinson’s disease. Acta Neurol Scand 1983; 95 (Supplement 68): 127–133.

    Google Scholar 

  68. Birkmayer W, Knoll J, Riederer P, Youdim MBH. Deprenyl leads to prolongation of L-dopa efficacy in Parkinson’s disease. Mod Probl Pharmacopsych 1983; 19: 170–176.

    Google Scholar 

  69. Rinne UK. New Strategies in the treatment of early Parkinson’s disease. Acta Neurol Scand 1991; 136 (Supplement 84): 95–98.

    Google Scholar 

  70. Fornadi F, Ulm G. Early combination with deprenyl: a retrospective study. Adv Neurol 1990; 53: 437–440.

    Google Scholar 

  71. Ulm G, Fornadi F. R-(-)-deprenyl in the treatment of end-of-dose akinesia. J Neural Transm 1987; 25 (Supplement): 163–172.

    Google Scholar 

  72. Liebermann AN, Gopinathan G, Neophytides A, Foo SH. Deprenyl in the treatment of Parkinson’s disease. NY State J Med 1984; 84: 13–16.

    Google Scholar 

  73. Eisler T, Teräväinen H, Nelson R, Krebs H, Weise V. Deprenyl in Parkinson’s disease. Neurology 1981; 31: 19–23.

    Google Scholar 

  74. Frankel JP, Kempster PA, Stibe CM, Eatough VMH, Nathanson M, Lees AJ. A double-blind, controlled study of high-dose l-deprenyl in the treatment of Parkinson’s disease. Clin Neuropharmacol 1989; 12: 448–451.

    Google Scholar 

  75. Goldstein L. The “on-off” pheomena in Parkinson’s disease — treatment and theoretical considerations. The Mount Sinai J Med 1980; 47: 80–84.

    Google Scholar 

  76. Golbe LI, Liebermann AN, Muenter MD. Ahlskog JE, Gopinathan E. Deprenyl in the treatment of symptom fluctuations in advanced Parkinson’s disease. Clin Neuropharmacol 1988; 11: 45–55.

    Google Scholar 

  77. Duvoisin RC, Golbe LI, Liebermann AN. Double-blind, randomized parallel placebo-controlled clinical trial of 10 mg l-deprenyl per day for 6 weeks as adjunctive therapy with Sinemet in Parkinson patients who are achieving less optimal response to Sinemet therapy. Study report. Data on file.

    Google Scholar 

  78. Heinonen E, Rinne UK, Tuominen J. Selegiline in the treatment of daily fluctuations in disability of parkinsonian patients with long-term levodopa treatment. Acta Neurol Scand 1989; 126 (Supplement 90): 113–118.

    Google Scholar 

  79. Lander CM, Lees A, Stern G. Oscillations in performance in levodopa-treated parkinsonians: Treatment with bromocrip-tine and l-deprenyl. Clin Exp Neurol 1979; 16: 197–203.

    Google Scholar 

  80. Lees AJ, Kohout LJ, Shaw KM, Stern GM, Elsworth JD. Deprenyl in Parkinson’s disease. Lancet 1977; ii: 791–795.

    Google Scholar 

  81. Przuntek H, Kuhn W. The effect of R-(-)-deprenyl in de novo Parkinson patients on combination therapy with levodopa and decarboxylase inhibitor. J Neural Transm 1987; 25 (Supplement): 97–104.

    Google Scholar 

  82. Rascol O, Montastruc JL, Senard JM. Two weeks of treatment with deprenyl (selegiline) does not prolong L-dopa effect in parkinsonian patients: a double-blind cross-over placebo-controlled trial. Neurology 1988; 38: 1387–1391.

    Google Scholar 

  83. Schachter M, Marsden CD, Parkes JD, Jenner P, Testa B. Deprenyl in the management of response fluctuations in patients with Parkinson’s disease on levodopa. J Neurol Neurosurg Psychiatry 1980; 40: 1016–1021.

    Google Scholar 

  84. Sivertson B, Dupont E, Mikkeslson B, Mogensen P, Rasmussen C. Selegiline and levodopa in early or moderately advanced Parkinson’s disease: a double-blind controlled short- and long-term study. Acta Neurol Scand 1989; 126: 147–152.

    Google Scholar 

  85. Teychenne PF, Parker S. Double-blind, cross-over placebo controlled trial of selegiline in Parkinson’s disease — an interim analysis. Acta Neurol Scand 1989; 126: 119–125.

    Google Scholar 

  86. Liebermann A, Fazzini E. Experience with selegiline and levodopa in advanced Parkinson’s disease. Acta Neurol Scand 1991; 136 (Supplement 84): 66–69.

    Google Scholar 

  87. Cohen G, Spina MB. Deprenyl suppresses the oxidant stress associated with increased dopamine turnover. Ann Neurol 1989; 26: 689–690.

    Google Scholar 

  88. Csanda E, Tarczy M, Takats A, Mogyoros I, Köves A. (—)-Deprenyl in the treatment of Parkinson’s disease. J Neural Transm 1983; 19: 283–290.

    Google Scholar 

  89. Tetrud JW, Langston JW. The effect of deprenyl (selegiline) on the natural history of Parkinson’s disease. Science 1989; 245: 519–522.

    Google Scholar 

  90. Myllylä VV, Sotaniemi KA, Vuorinen JA, Heinonen E. Selegiline as a primary treatment of Parkinson’s disease. Acta Neurol Scand 1991; 136 (Supplement 84): 70–72.

    Google Scholar 

  91. The Parkinson Study Group. Effect of deprenyl on the progression of disability in early Parkinson’s disease. N Engl J Med 1989a; 321: 1364–1371.

    Google Scholar 

  92. Allain H, Coungnard J, Neukirch HC, the FSMT members. Selegiline in de novo parkinsonian patients: the French selegiline multicenter trial (FSMT). Acta Neurol Scand 1991; 136 (Supplement 84): 73–84.

    Google Scholar 

  93. Langston JW. Selegiline as neuroprotective therapy in Parkinson’s disease: concepts and controversies. Neurology 1990; 40: 61–69.

    Google Scholar 

  94. Rinne JO. Nigral degeneration in Parkinson’s disease in relation to clinical features. Acta Neurol Scand 1991; 136 (Supplement 84): 87–90.

    Google Scholar 

  95. Tetrud JW, Langston JW. Comparison of patients with Parkinson’s disease treated with selegiline (deprenyl) and levodopa compared to treatment with levodopa alone: a prospective, double-blind study. 5th Int Con Movement Disorders 1990; Washington D.C.; Abstract 179.

    Google Scholar 

  96. Marley E, Blackwell B. Interactions of monoamine oxidase inhibitors, amines and foodstuffs. Adv Pharmacol Chemother 1970; 8: 186–239.

    Google Scholar 

  97. Elsworth JD, Glover V, Reynolds GP. Deprenyl administration in man: A selective monoamine oxidase inhibitor without the cheese effect. Psychopharmacol 1978; 57: 33–38.

    Google Scholar 

  98. Heinonen EH, Lammintausta R. A review of the pharmacology of selegiline. Acta Neurol Scand 1991; 84 (Supplement 136): 44–59.

    Google Scholar 

  99. Pickar D, Cohen RM, Jimerson DC, Murphy LD, Tyramine infusions and selective monoamine oxidase inhibitor treatment. Psychopharmacol 1981; 74: 4–7.

    Google Scholar 

  100. Prasad A, Glover V, Goodwin BL. Enhanced pressor sensitivity to oral tyramine challenge following high doses of selegiline treatment. Psychopharmacol 1988; 95: 540–543.

    Google Scholar 

  101. Sunderland T, Mueller EA, Cohen RM, Jimerson DC, Picker D, Murphy DL. Tyramine pressor sensitivity changes during deprenyl treatment. Psychopharmacol 1985; 86: 432–437.

    Google Scholar 

  102. Schulz R, Antonin K-H, Hoffman E, Jedrychowsky M, Nillson E, Schick C. Tyramine kinetics and pressor sensitivity during monoamine oxidase inhibition by selegiline. Clin Pharmacol Therap 1989; 46: 528–536.

    Google Scholar 

  103. Stern GM, Lees A J, Sandler M. Recent observations on the clinical pharmacology of deprenyl. J Neural Transm 1978; 43: 245–251.

    Google Scholar 

  104. Mann J, Aarons SF, Frances AJ, Browns RD. Studies of selective and reversible monoamine oxidase inhibitors. J Clin Psychiatry 1984; 45: 62–66.

    Google Scholar 

  105. Waters CH. Side effects of selegiline (Elderpryl). Abstract 938p. Neurology 1990; 40 (Supplement): 370.

    Google Scholar 

  106. Golbe LI. Long term efficacy and safety of deprenyl (selegiline) in advanced Parkinson’s disease. Neurology 1989; 39: 1109–1111.

    Google Scholar 

  107. Zsilla G, Földi P, Held G, Szekely Am, Knoll J. The effect of repeated doses of (—)-deprenyl on the dynamics of monoaminergic transmission. Comparison with clorgyline. Pol J Pharmacol Pharm 1986; 38: 57–67.

    Google Scholar 

  108. Knoll J. R-(-)-deprenyl facilitates the activity of the nigrostriatal dopaminergic neuron. J Neural Transm 1987; 25 (Supplement): 45–66.

    Google Scholar 

  109. Knoll J. Deprenyl (selegiline): The history of its development and pharmacological action. Acta Neurol Scand 1983; 95: 57–80.

    Google Scholar 

  110. Zsilla G, Knoll J. The action of (-)-deprenyl on monoamine oxidase turnover rate in rat brain. In: Costa E, Racagni G, editors. Typical and atypical antidepressants: molecular mechanisms. New York: Raven Press, 1982; 211–217.

    Google Scholar 

  111. Fuxe K, Grobecker H, Jonsson J. Effect of beta-phenylethylamine on central and peripheral monoamine containing neurons. Eur J Pharmacol 1967; 2: 203–207.

    Google Scholar 

  112. Antelmann SM, Edwards DJ, Lin M. Phenylethylamine: evidence for a direct, post- synatpic dopamine-receptor stimulating action. Brain Res 1977; 127: 317–322.

    Google Scholar 

  113. Heikkila RE, Cabbat FS, Manzoni L, Duvoisin RC. Potentiation by deprenil of L-dopa induced circling in nigral lesioned rats. Pharmacol Biochem Behav 1981; 15: 75–79.

    Google Scholar 

  114. Cedarbaum JM, Silvestri M, Clark M, Hart A, Kutt H. l-Deprenyl, levodopa pharmacokinetics, and response fluctuations in Parkinson’s disease. Clin Neuropharmacol 1990; 13: 29–35.

    Google Scholar 

  115. Halliwell B. Oxidants and central nervous system: some fundamental questions. Is oxidant damage relevant to Parkinson’s disease, Alzheimer’s disease, traumatic injury or stroke? Acta Neurol Scand 1989; 80 (Supplement 126): 23–34.

    Google Scholar 

  116. Youdim MBH, Ben-Shachar D, Riederer P. Iron in brain function and dysfunction with emphasis on Parkinson’s disease. Eur Neurol 1991; 31 (Supplement 1): 34–40.

    Google Scholar 

  117. Olanow CW. Oxidation reactions in Parkinson’s disease. Neurology 1990; 40 (Supplement 3): 32–37.

    Google Scholar 

  118. Spina MB, Cohen G. Dopamine turnover and glutathione oxidation: implications for Parkinson’s disease. Proc Natl Acad Sci USA 1989; 86: 1398–1400.

    Google Scholar 

  119. Kish S J, Monto C, Hornykiewicz O. Glutathione peroxidation activity in Parkinson’s disease brain. Neurosci Lett 1985; 58: 343–346.

    Google Scholar 

  120. Ambani LM, van de Woert MH, Murphy S. Brain peroxidase and catalase in Parkinson’s disease. Arch Neurol 1975; 32: 114–118.

    Google Scholar 

  121. Sofic E, Paulus W, Jellinger K, Riederer P, Youdim MBH. Selective increase of iron in substantia nigra zona compacta of parkinsonian brains. J Neurochem 1991; 56: 978–982.

    Google Scholar 

  122. Dexter DT, Carter CJ, Wells FR, Javoy-Agid F, Agid Y, Lees A. Basal Lipid peroxidation in the substantia nigra in increased in Parkinson’s disease. J Neurochem 1989; 52: 381–389.

    Google Scholar 

  123. Szökö E, Bathory G, Tekes K, Magyar K. Effect of l-deprenyl on lipid peroxidation in rat brain homogenate. Eur J Pharmacol 1990; 183: 1549.

    Google Scholar 

  124. Carillo M-C, Kanai S, Nokubo M, Kitani K. (—)-Deprenyl induced activities of both superoxide dismutase and catalase but not of glutathione peroxidase in the striatum of young male rats. Life Sci 1991; 48: 517–521.

    Google Scholar 

  125. Rabey JM, Hefti F. Neuromelanin synthesis in rat and human substantia nigra. J Neural Transm (PDSec) 1990; 2: 1–14.

    Google Scholar 

  126. Davies GC, Williams AC, Markey SP, Ebert MH, Caine ED, Reichert CM. Chronic parkinsonism secondary to intravenous injection of meperidine-analogues. Psychiatry Res 1979; 1: 249–254.

    Google Scholar 

  127. Langston JW, Ballard P, Tetrud JW. Irwin I. Chronic parkinsonism in humans due to a product of meperidine-analog synthesis. Science 1983; 219: 249–254.

    Google Scholar 

  128. Mallard PA, Tetrud JW, Langston JW, Permanent human parkinsonism due to MPTP. Neurology 1985; 35: 949–980.

    Google Scholar 

  129. Langston JW, Forno LS, Rebert CS, Irwin I. Selective nigral toxicity after systemic administration of MPTP in the squirrel monkey. Brain Res 1984; 292: 390–394.

    Google Scholar 

  130. Fuller RW, Hemrick-Luecke SK, Perry KW. Deprenyl antagonises acute lethality of MPTP in mice. J Pharmacol Exp Therap 1988; 247: 531–535.

    Google Scholar 

  131. Markey SP, Johannson JN, Chiueh CC, Burns RS, Herkenham MA. Intraneuronal generation of a pyridinium metabolite may cause drug-induced parkinsonism. Nature 1984;311:464–467.

    Google Scholar 

  132. Javitch JA, D’Amato RJ, Strittmatter SM, Snyder SH. Parkinsonism-inducing neurotoxin, N-methyl-4-phenyM,2,3,6-tetrahydropyridine: Uptake of the metabolite N- methyl-4-phenylpyridine by dopamine neurons explains selective toxicity. Proc Natl Acad Sci USA 1985; 82: 2173–2177.

    Google Scholar 

  133. Mizuno Y, Saitoh T, Sone N. Inhibition of mitochondrial NADH-ubiquinone oxido- reductase activity by l-methyl-4-phenyl-pyridinium ion. Biochem Biophys Res Commun 1989; 163: 1450–1455.

    Google Scholar 

  134. Mytiliuneou C, Cohen G. Deprenyl protects dopamine neurons from neurotoxic effect of l-methyl-4-phenylpyridinium ion. J Neurochem 1985; 45: 1951–1953.

    Google Scholar 

  135. Bertocci B, Gill G, Da Prada M. Prevention of the DSP-4-induced noradrenergic neurotoxicity by irreversible, not by reversible MAO-B inhibitors. Pharmacol Res Commun 1988; 20 (Supplement 4): 131–132.

    Google Scholar 

  136. Finnegan KT, Skrati JJ, Irwin I, DeLanney LE, Langston JW. Protection against DSP-4 induced neurotoxicity by deprenyl is not related to its inhibition of MAO-B. Eur J Pharmacol Í990; 184: 119–126.

    Google Scholar 

  137. Knoll J. The possible mechanism of action of (—)-deprenyl in Parkinson’s disease. J Neural Transm 1978: 43: 177–198.

    Google Scholar 

  138. Mena MA, Pardo B, Casarejos MJ, Fahn S, Garcia de Yebenes J. Neurotoxicity of levodopa on catecholamine-rich neurons. Movement Disorders 1992; 7: 23–31.

    Google Scholar 

  139. Tatton WG, Greenwood CE. Rescue of dying neurons: a new action for deprenyl in MPTP parkinsonism. J Neurosci Res 1991; 30: 666–672.

    Google Scholar 

  140. Csanda E, Tarczy M. Selegiline in the early and late phase of Parkinson’s disease. J Neural Transm 1987; 25 (Supplement): 105–113.

    Google Scholar 

  141. Elizan TS, Yahr MD, Moros DA, Mendoza MR, Pang S, Bodian CA. Selegiline use to prevent progression of Parkinson’s disease. Arch Neurol 1989; 46: 1275–1279.

    Google Scholar 

  142. Teräväinen H. Selegiline in Parkinson’s disease. Acta Neurol Scand 1990; 81: 333–336.

    Google Scholar 

Download references

Authors

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 1993 Springer Basel AG

About this chapter

Cite this chapter

Wessel, K. (1993). MAO-B Inhibitors in Neurological Disorders with Special Reference to Selegiline. In: Szelenyi, I. (eds) Inhibitors of Monoamine Oxidase B. Milestones in Drug Therapy. Birkhäuser, Basel. https://doi.org/10.1007/978-3-0348-6348-3_13

Download citation

  • DOI: https://doi.org/10.1007/978-3-0348-6348-3_13

  • Publisher Name: Birkhäuser, Basel

  • Print ISBN: 978-3-0348-6349-0

  • Online ISBN: 978-3-0348-6348-3

  • eBook Packages: Springer Book Archive

Publish with us

Policies and ethics