350 руб
Журнал «Вопросы биологической, медицинской и фармацевтической химии» №3 за 2015 г.
Статья в номере:
Микроэлементы: биологическая роль и значение для медицинской практики. Сообщение 3. Марганец
Авторы:
М.Г. Скальная - д.м.н., профессор, кафедра профилактической медицины, Оренбургский государственный университет; АНО «Центр биотической медицины» (Москва). E-mail: skalnaya@yandex.ru А.В. Скальный - д.м.н., профессор, генеральный директор Института микроэлементов ЮНЕСКО (Лион, Франция); Институт биоэлементологии, Оренбургский государственный университет; лаборатория биотехнологии и прикладной биоэлементологии, Ярославский государственный университет им. П.Г. Демидова. E-mail: teu.moscow@gmail.com
Аннотация:
Статья продолжает серию обзоров о биологической роли эссенциальных микроэлементов. Данное сообщение посвящено марганцу. Рассмотрены накопленные к настоящему времени сведения о его участии в физиологических процессах, метаболизме, путях и объемах поступления в организм, связи с различными заболеваниями, применении в медицине.
Страницы: 14-25
Список источников

 

  1. Santamaria A.B. Manganese exposure, essentiality and toxicity/ Indian J. Med. Res. 2008. V. 128. P. 484-500.
  2. EPA. Health effects support document for manganese/ Health and ecological criteria division. Washington. DC. 2003. P. 673.
  3. Brown D.R. Prions and manganese: a maddening beast // Metallomics. 2011. V. 3. P. 229-238.
  4. Matsuoka A. et al. Progesterone increases manganese superoxide dismutase expression via a cAMP-dependent signaling mediated by noncanonical Wnt5a pathway in human endometrial stromal cells // J. Clin. Endocrinol. Metab. 2010. V. 95. № 11. P. 291-299.
  5. Sugino N. et al. Superoxide dismutase expression in the human corpus luteum during the menstrual cycle and in early pregnancy // Mol. Hum. Reprod. 2000. V. 6. № 1. P. 19-25.
  6. Sugino N. et al. Hormonal regulation of copper-zinc superoxide dismutase and manganese superoxide dismutase messenger ribonucleic acid in the rat corpus luteum: induction by prolactin and placental lactogens // Biol. Reprod. 1998. V. 59. № 3. P. 599-605.
  7. Cheng J., Fu J.L. and Zhou Z.C. The inhibitory effects of manganese on steroidogenesis in rat primary Leydig cells by disrupting steroidogenic acute regulatory (StAR) protein expression // Toxicology. 2003. V. 187. № 2-3. P. 139-148.
  8. Dhar S.K. et al. Manganese superoxide dismutase is a p53-regulated gene that switches cancers between early and advanced stages // Cancer Res. 2011. V. 71. № 21. P. 6684-6695.
  9. Holley A.K., Dhar S.K., Xu Y. and St Clair D.K. Manganese superoxide dismutase: beyond life and death // Amino Acids. 2012. V. 42. № 1. P. 139-158.
  10. Kalea A.Z., Harris P.D. and Klimis-Zacas D.J.Dietary manganese suppresses alpha1 adrenergic receptor-mediated vascular contraction // J. Nutr. Biochem. 2005. V. 16. № 1. P. 44-49.
  11. Nordhøy W. et al. Manganese ions as intracellular contrast agents: proton relaxation and calcium interactions in rat myocardium // N.M.R. Biomed. 2003. V. 16. № 2. P. 82-95.
  12. Tuschl K. et al. Syndrome of hepatic cirrhosis, dystonia, polycythemia, and hypermanganesemia caused by mutations in SLC30A10, a manganese transporter in man // Am. J. Hum. Genet. 2012. V. 90. № 3. P. 457-466.
  13. Tan J. et al. Regulation of intracellular manganese homeostasis by Kufor-Rakeb syndrome-associated ATP13A2 protein // J. Biol. Chem. 2011. V. 286. № 34. P. 29654-29662.
  14. García-Rodríguez N. et al. Impaired manganese metabolism causes mitotic misregulation // J. Biol. Chem. 2012. V. 287. № 22. P. 18717-18729.
  15. Rodríguez-Rodríguez E., Bermejo L.M., López-Sobaler A.M., Ortega R.M. An inadequate intake of manganese may favour insulin resistance in girls // Nutr. Hosp. 2011. V. 26. № 5. P. 965-970.
  16. Tuschl K. et al. Syndrome of hepatic cirrhosis, dystonia, polycythemia, and hypermanganesemia caused by mutations in SLC30A10, a manganese transporter in man // Am. J. Hum. Genet. 2012. V. 90. № 3. P. 457-466.
  17. DeWitt M.R., Chen P. and Aschner M. Manganese efflux in Parkinsonism: insights from newly characterized SLC30A10 mutation // Biochem. Biophys. Res. Commun. 2013. V. 432. № 1. P. 1-4.
  18. Guilarte T.R.Manganese and Parkinson\'s disease: a critical review and new findings // Cien. Saude Colet. 2011. V. 16- № 11. P. 4549-4566.
  19. Ilies M. et al. 2-aminoimidazole amino acids as inhibitors of the binuclear manganese metalloenzyme human arginase I // J. Med. Chem. 2010. V. 53. № 10. P. 4266-4276.
  20. Lee B.H. et al. Argininemia presenting with progressive spastic diplegia // Pediatr. Neurol. 2011. V. 44. № 3. P. 218-220.
  21. Gomes M.E. et al. Neonatal cholestasis: an uncommon presentation of hyperargininemia // J. Inherit. Metab. Dis. 2011. Suppl. 3. P. 503-506.
  22. Yin Z. et al.Ferroportin is a manganese-responsive protein that decreases manganese cytotoxicity and accumulation // J. Neurochem. 2010. V. 112. № 5. P. 1190-1198.
  23. Madejczyk M.S. and Ballatori N. The iron transporter ferroportin can also function as a manganese exporter // Biochim. Biophys. Acta. 2012. V. 1818. № 3. P. 651-657.
  24. Bai S.P. et al. Manganese source affects manganese transport and gene expression of divalent metal transporter 1 in the small intestine of broilers // Br. J.Nutr. 2012. V. 108. № 2. P. 267-276.
  25. He W., Hu Z.The role of the Golgi-resident SPCA Ca²⁺/Mn²⁺ pump in ionic homeostasis and neural function // Neurochem. Res. 2012. V. 37. № 3. P. 455-468.
  26. Garrick M.D. et al. DMT1: a mammalian transporter for multiple metals // Biometals. 2003. V. 16. № 1. P. 41-54.
  27. Zheng G., Chen J., Zheng W. Relative contribution of CTR1 and DMT1 in copper transport by the blood-CSF barrier: implication in manganese-induced neurotoxicity // Toxicol. Appl. Pharmacol. 2012. V. 260. № 3. P. 285-293.
  28. Finley J.W., Davis C.D. Manganese deficiency and toxicity: are high or low dietary amounts of manganese cause for concern - // Biofactors. 1999. V. 10. № 1. P. 15-24.
  29. Mukhopadhyay S., Linstedt A.D. Identification of a gain-of-function mutation in a Golgi P-type ATPase that enhances Mn2+ efflux and protects against toxicity // Proc. Natl. Acad. Sci. USA. 2011. V. 108. № 2. P. 858-863.
  30. Tuschl K., Mills P.B., Clayton P.T. Manganese and the brain/ Int. Rev. Neurobiol. 2013. V. 10. P. 277-312.
  31. Kimura M., Ujihara M., Yokoi K. Tissue manganese levels and liver pyruvate carboxylase activity in magnesium-deficient rats // Biol. Trace Elem. Res. 1996. V. 52. № 2. P. 171-179.
  32. Baly D.L., Keen C.L., Hurley L.S. Pyruvate carboxylase and phosphoenolpyruvate carboxykinase activity in developing rats: effect of manganese deficiency // J. Nutr. 1985. V. 115. № 7. P. 872-879.
  33. Cizewski V.C., Yang M., Hall M.D. Manganese Transport and Trafficking: Lessons Learned from Saccharomyces cerevisiae // Eukaryot. Cell. 2005. V. 4. № 7. P. 1159-1165.
  34. Blairanne W. et al. Altered Manganese Homeostasis and Manganese Toxicity in a Huntington\'s Disease Striatal Cell Model Are Not Explained by Defects in the Iron Transport SystemB // Toxicol. Sci. 2010. V. 117. № 1. P. 169-179.
  35. Fujishiro H.Roles of ZIP8, ZIP14, and DMT1 in transport of cadmium and manganese in mouse kidney proximal tubule cells // Metallomics. 2012. V. 4. № 7. P. 700-708.
  36. Kim H.Y. et al. Effects of manganese exposure on dopamine and prolactin production in rat // Neuroreport. 2009. V. 20. № 1. P. 69-73.
  37. Soldin O.P., Aschner M. Effects of manganese om thyroid hormone homeostasis // Neurotoxicology. 2007. V. 28. № 5. P. 951-956.
  38. Fitsanakis V.A., Au C., Erikson K.M., Aschner M. The effects of manganese on glutamate, dopamine and gamma-aminobutyric acid regulation // Neurochem. Int. 2006. V. 48. № 6-7. P. 426-433.
  39. Di J.-W., Bi S.P. UV-Vis spectroscopic study of aluminum-manganese cooperative effect on the melanin formation from DOPA oxidation // Guang Pu Xue Yu Guang Pu Fen Xi. 2005.V. 25. № 1.P. 83-85.
  40. Hamberg M., Su C., Oliw E. Manganese lipoxygenase. Discovery of a bis-allylic hydroperoxide as product and intermediate in a lipoxygenase reaction // J. Biol. Chem. 1998. V. 273. № 21. P. 13080-13088.
  41. Perry C.A. et al. Perthes\' disease and blood manganese levels // Arch. Dis. Child. 2000. V. 82. № 5. P. 428.
  42. Uramatsu M. et al. Different effects of sulfur amino acids on prolidase and prolinase activity in normal and prolidase-deficient human erythrocytes // Clin. Chim. Acta. 2007. V. 375. № 1-2. P. 129-135.
  43. Besio R. et al. A Mn(II)-Mn(II) center in human prolidase // Biochim. Biophys. Acta. 2013. V. 1834. № 1. P. 197-204.
  44. Besio R. et al. Kinetic and Structural Evidences on Human Prolidase Pathological Mutants Suggest Strategies for Enzyme Functional Rescue // PLoS One. 2013. V. 8. № 3. P. 58792.
  45. Furukawa K., Okajima T. Galactosyltransferase I is a gene responsible for progeroid variant of Ehlers-Danlos syndrome: molecular cloning and identification of mutations // Biochim. Biophys. Acta. 2002. V. 1573. № 3. P. 377-381.
  46. Just N., Cudalbu C., Lei H., Gruetter R. Effect of manganese chloride on the neurochemical profile of the rat hypothalamus // J. Cereb. Blood Flow Metab. 2011. V. 31. № 12.P. 2324-2333.
  47. Larrèque M. et al. Prolidase and manganese deficiency. Apropos of a case: diagnosis and treatment // Аnn. Dermatol. Venereol. 1982.V. 109. № 8. P. 667-678.
  48. Авцын А.П., Жаворонков А.А., Риш М.А., Строчкова Л.С. Микроэлементозы человека: этиология, классификация, органопатология. М.: Медицина. 1991. 496 с.
  49. Патент № 2230491 (РФ). Способ выявления группы риска заболеваний молочной железы у женщин / В.А. Семикопенко, А.В. Скальный, В.А.Демидов. 2004.
  50. Оберлис Д., Харланд Б., Скальный А.Биологическая роль макро- и микроэлементов у человека и животных. СПб.: Наука. 2008. 544 с.
  51. Cai Q., Shu X.O., Wen W., Cheng J.R., Dai Q., Gao Y.T., Zheng W. Genetic polymorphism in the manganese superoxide dismutase gene, antioxidant intake, and breast cancer risk: results from the Shanghai Breast Cancer Study // Breast Cancer Res. 2004. V. 6. № 6. R647-55.
  52. Liu G., Sun G., Wang Y., Wang D., Hu W., Zhang J. Association between manganese superoxide dismutase gene polymorphism and breast cancer risk: a meta-analysis of 17,842 subjects // Mol. Med. Rep. 2012. V.6. № 4. Р. 797-804.
  53. Hart P.C., Mao M., de Abreu A.L., Ansenberger-Fricano K., Ekoue D.N., Ganini D., Kajdacsy-Balla A., Diamond A.M., Minshall R.D., Consolaro M.E., Santos J.H., Bonini M.G. MnSOD upregulation sustains the Warburg effect via mitochondrial ROS and AMPK-dependent signalling in cancer // Nat. Commun. 2015. № 5.
  54. Matyal R., Sakamuri S., Huang T., Owais K., Parikh S., Khabbaz K., Wang A., Sellke F., Mahmood F. Oxidative stress and nerve function after cardiopulmonary bypass in patients with diabetes // Ann. Thorac. Surg. 2014. V.98. № 5. P.1635-1643.
  55. Unfer T.C., Figueiredo C.G., Zanchi M.M., Maurer L.H., Kemerich D.M., Duarte M.M., Konopka C.K., Emanuelli T. Estrogen plus progestin increase superoxide dismutase and total antioxidant capacity in postmenopausal women // Climacteric. 2014. V.12. Р. 1-10.
  56. Kattan Z1, Minig V, Leroy P, Dauça M, Becuwe P. Role of manganese superoxide dismutase on growth and invasive properties of human estrogen-independent breast cancer cells // Breast. Cancer. Res. Treat. 2008. V. 108. № 2. Р. 203-215.
  57. Dong-Won Shin, Eun-Ji Kim, Se-Won Lim, Young-Chul Shin, Kang-Seob Oh, Eun-Jin Kim.Association of Hair Manganese Level with Symptoms in Attention-Deficit/Hyperactivity Disorder // Psychiatry Investig. 2015. V. 12. № 1. Р. 66-72.
  58. Kim Y., Lobdell D.T., Wright C.W., Gocheva V.V., Hudgens E., Bowler R.M. Blood Metal Concentrations of Manganese, Lead, and Cadmium in Relation to Serum Ferritin Levels in Ohio Residents // Biol Trace Elem Res. 2015. 13 р.
  59. Byung-Kook Lee, Yangho Kim. Sex-specific Profiles of Blood Metal Levels Associated with Metal-Iron Interactions // Saf Health Work. 2014. V. 5. № 3. P. 113-117.
  60. Lee B.K., Kim Y. Effects of menopause on blood manganese levels in women: analysis of 2008-2009 Korean National Health and Nutrition Examination Survey data // Neurotoxicology. 2012. V. 33. № 3. Р. 401-405.
  61. Friedman B.J., Freeland-Graves J.H., Bales C.W., Behmardi F., Shorey-Kutschke R.L., Willis R.A., Crosby J.B., Trickett P.C., Houston S.D. Manganese balance and clinical observations in young men fed a manganese-deficient diet // J. Nutr. 1987. V. 117. № 1. Р. 133-143.
  62. Takeda A. Manganese action in brain function // Brain Res. Brain. Res. Rev. 2003. V. 41. № 1. Р. 79-87.
  63. Carl G.F., Blackwell L.K., Barnett F.C., Thompson L.A., Rissinger C.J., Olin K.L., Critchfield J.W., Keen C.L., Gallagher B.B. Manganese and epilepsy: brain glutamine synthetase and liver arginase activities in genetically epilepsy prone and chronically seizured rats. Epilepsia // 1993. V. 34. № 3. Р. 441-446.
  64. Porcheron G., Garénaux A., Proulx J., Sabri M., Dozois C.M. Iron, copper, zinc, and manganese transport and regulation in pathogenic Enterobacteria: correlations between strains, site of infection and the relative importance of the different metal transport systems for virulence // Front Cell Infect Microbiol. 2013. № 3. Р. 90.
  65. Lisher J.P., Giedroc D.P. Manganese acquisition and homeostasis at the host-pathogen interface // Front Cell Infect Microbiol. 2013. № 3. Р. 91.
  66. Kemmerer A.R., Elvehjem C.A., Hart E.B. Studies on the relation of manganese to the nutrition of the mouse // J. Biol. Chem. 1931. V. 92. № 3. P. 623-630.
  67. Underwood E.G.Trace elements in human and animal nutrition. 4rd ed. Acad. Press. New York. 1977. 402 p.
  68. Doisey E.A. Jr. In-. Hemphill D.D. (ed) Trace Substances m Environmental Health // VI. UniversityofMissouri. Columbia. Missouri. 1972. P. 193-199.
  69. Скальный А.В. Микроэлементы // Лабораторная диагностика инфекционных болезней. Справочник / под ред. В.И. Покровского, М.Г. Твороговой, Г.А. Шипулина. М.: БИНОМ. 2013. С. 447-467.
  70. Маневич А.З., Островский А.Ю., Островская М.А. Лабораторная диагностика для всех. М. 2012. 732 с.
  71. Афтанас Л.И. и др. Элементный статус населения России. Ч. 2. Элементный статус населения Центрального федерального округа / под ред. А.В. Скального, М.Ф. Киселева.СПб.: Медкнига «ЭЛБИ-СПб». 2011. 432 с.
  72. Скальная М.Г., Скальный А.В., Демидов В.А., Грабеклис А.Р., Лобанова Ю.Н.Установление границ физиологического (нормального) содержания некоторых химических элементов в волосах жителей г. Москвы с применением центильных шкал // Вестник С.-Петербургской ГМА им. И.И.Мечникова. 2004. № 4. С. 82-88.
  73. Скальный А.В. Эколого-физиологическое обоснование эффективности использовании макро - и микроэлементов при нарушениях гомеостаза у обследуемых из различных климатогеографических регионов: Дис. ... докт. мед. наук. М. 2000. 352 с.
  74. Just N., Gruetter R. Detection of neuronal activity and metabolism in a model of dehydration-induced anorexia in rats at 14.1 T using manganese-enhanced MRI and 1H MRS // NMR Biomed. 2011. V. 24. № 10. Р. 1326-1336.
  75. Hankir M.K., Parkinson J.R., Minnion J.S., Addison M.L., Bloom S.R., Bell J.D. Peptide YY 3-36 and pancreatic polypeptide differentially regulate hypothalamic neuronal ac­tivity in mice in vivo as measured by manganese-enhanced magnetic resonance imaging // J. Neuroendocrinol. 2011. V. 23. № 4. Р. 371-380.
  76. Wieland H.A., Willim K., Doods H.N. Divalent cations in­fluencing neuropeptide Y receptor subtype binding in rat hip­pocampus and cortex membranes as well as in recombinant cells // Regul. Pept. 1998. V. 25. № 75-76. Р. 263-269.
  77. Irvine R.F.Manganese-stimulated phosphatidylinositol headgroup exchange in rat liver microsomes // Biochem. Biophys. Acta. 1998. V. 1393. №. 2-3. P. 292-298.