ВАНАДИЙ: БИОЛОГИЧЕСКАЯ РОЛЬ, ТОКСИКОЛОГИЯ И ФАРМАКОЛОГИЧЕСКОЕ ПРИМЕНЕНИЕ

Н.М. Воробьева, Е.В. Федорова, Н.И. Баранова

Аннотация


В России химия и геохимия ванадия, элемента переменной валентности, стоящего в периодической таблице между титаном и хромом и имеющего широкий спектр степеней окисления, развиваются уже давно, но его фармакология сравнительно молода, и в современной русскоязычной научной литературе ванадий представлен в этом аспекте мало. В данной статье рассмотрены основные стороны геохимии ванадия, физико-химические методы его исследования, его биологическая роль и токсикология и обнаруженные на сегодняшний день фармакологические эффекты. Соединения ванадия приобретают все большее значение как возможные перспективные лекарственные средства.

Ключевые слова


ванадий, ванадил, ванадат, фармакология.

Полный текст:

PDF

Литература


Беляева Н.Ф., Городецкий В.К., Точилкин А.И., Голубев М.А. Ванадийсодержащие соединения – новый класс терапевтических средств для лечения сахарного диабета // Вопр. мед. хим. – 2000. – Т. 46. – С. 344–360.

Борисенко Л.Ф. Ванадий: минералогия, геохимия и типы эндогенных месторождений // М. : Недра, 1973. – 192 c.

Воробьева Н.М., Федорова Е.В. Фармакотоксикологическое исследование комплексов ванадия // II Международная научно-практическая конференция «Высокие технологии, фундаментальные и прикладные исследования в физиологии и медицине». – Санкт-Петербург, 2011 г.

Зайчик А.Ш., Чурилов Л.П. Патохимия (эндокринно-метаболические нарушения). – СПб. : ЭлБи-СПб, 2007. – 768 с.

Ноздрюхина Л.Р. Биологическая роль микроэлементов в организме животных и человека. – М. : Наука, 1977. – 184 с.

Перельман А.И. Геохимия. – M. : Высш. шк., 1989. – 528 с.

Ронов А.Б., Ярошевский А.А. Химическое строение земной коры // Геохимия. – 1967. – № 11. – С. 1285–1309.

Сауков А. А. Геохимия. – М. : Наука, 1975. – 477 c.

Чертко Н.К., Чертко Э.Н. Геохимия и экология химических элементов. – Минск : Изд. центр БГУ, 2008. – 137 с.

Юдович Я.Э., Кетрис М.П. Токсические элементы-примеси в ископаемых углях. – Екатеринбург : УрО РАН, 2005. – 655 с.

Abe Y., Nagata R., Hasunuma Y., Yokosawa H. Isolation, characterization and cDNA cloning of a one-lobed transferrin from the ascidian Halocynthia roretzi // Comp. Biochem. Physiol. B. – 2001. – Vol. 128. – P. 73–79.

Agency for Toxic Substances and Disease Registry (ATSDR). Toxicological profile for vanadium. (Draft for public comment). – Atlanta, 2009. – 240 p.

Andrezlov L., Gbelcov H., urakov Z. DNA damage induction and antiproliferative activity of vanadium (V) oxido monoperoxido complex containing two bidentate heteroligands // J. Trace Elements Med. Biol. – 2013. – Vol. 27. – P. 21–26.

Anisimov A.V., Lesnugin A.Z., Senyavin V.M., Fedorova E.V. Vanadium peroxo complexes with pyridine ligands as sulfide oxidation catalysts // Petroleum Chemistry. – 2002. – Vol. 42. – P. 121– 123.

Anisimov A.V., Fedorova E.V., Lesnugin A.Z., Senyavin V.M. et al. Vanadium peroxocomplexes as oxidation catalysts of sulfur organic compounds by hydrogen peroxide in bi-phase systems // Catalysis Today. – 2003. – Vol. 78. – № 1. – P. 319–325.

Anke M., Groppel B., Gruhn K., Kosla T. et al. Spurenelement symposium: new trace elements / Anke M., Baumann W., Braunlich H., Bruckner C. and Groppel B. (eds). – FriedrichSchiller-Universitat, Jena, 1986. – P. 1266–1275.

Aguirre G., Cerecetto H., Di Maio R. et al. Quinoxaline N,N’-dioxide derivatives and related compounds as growth inhibitors of Trypanosoma cruzi. Structure-activity relationships // Bioorg. Med. Chem. Lett. – 2004. – Vol. 14. – P. 3835– 3839.

Armstrong E. M., Collison D., Ertok N., Garner C.D. NMR studies on natural and synthetic Amavadin // Talanta. – 2000. – Vol. 53. – P. 75– 87.

Audin A., Sayal A., Sayin S., Erdem O. An investigation on the relationship between vanadium and antioxidative enzyme system in rats // Turkish J. Pharm. Sci. – 2005. – Vol. 2. – P. 17–24.

Azay J., Brs J., Krosniak M. et al. Vanadium pharmacokinetics and oral bioavailability upon single-dose administration of vanadyl sulfate to rats // Fundam. Clin. Pharmacol. – 2001. – Vol. 15. – P. 313–324.

Baran E. J. Vanadium detoxification: chemical and biochemical aspects // Chem. Biodiversity. – 2008. – Vol. 5. – P. 1475–1484. 89

Barceloux D.G. Vanadium // J. Toxicol. Clin. Toxicol. – 1999. – Vol. 37. – P. 265–278.

Barnett P.W., Hemrika H.L., Dekker A.O. et al. Isolation, characterization, and primary structure of the vanadium chloroperoxidase from the fungus Embellisia didymospor // J. Biol. Chem. – 1998. – Vol. 273. – P. 23381–23387.

Basso R. The crystal structure of palenzonaite, a new vanadate garnet from Val Graveglia (Northern Apennines, Italy) // N. Jb. Miner. Mh. – 1987. – P. 136–144.

Benitez J., Guggeri L., Tomaz I. et al. A novel vanadyl complex with a polypyridyl DNA intercalator as ligand: A potential anti-protozoa and anti-tumor agent // J. Inorg. Biochem. – 2009. – Vol. 103. – P. 1386–1394.

Benitez J., Becco L., Correia I. et al. Vanadium polypyridyl compounds as potential antiparasitic and antitumoral agent. New achievements // J. Inorg. Biochem. – 2011. – Vol. 10. – P. 303–312.

Benabe J.E., Echegoyen L.A., Pastrana B., Martinez-Maldonado M. Mechanism of inhibition of glycolysis by vanadate // J. Biol. Chem. – 1987. – Vol. 267. – P. 9555–9560.

Bergamashi G., Rosti V., Danava M. et al. Inhibitors of tyrosine phosphorylation induce apoptosis in human leukemic cell lines // Leukemia. – 1993. – Vol. 7. – P. 2012–2018.

Bernroitner M., Zamocky M., Furtmller P.G. et al. Occurrence, phylogeny, structure, and function of catalases and peroxidases in cyanobacteria // J. Exp. Bot. – 2009. – Vol. 60. – P. 423–440.

Bhanot S., McNeill J.H. Vanadyl sulfate lowers plasma insulin levels and blood pressure in spontaneously hypertensive rats // Hypertension. – 1994. – Vol. 24. – P. 625–632.

Bhanot S., McNeill J.H., Bryer-Ash M. Vanadyl sulfate prevents fructoseinduced hyperinsulinemia and hypertension in rats // Hypertension. – 1994. – Vol. 23. – P. 308–312.

Bhuiyan M.S., Shibuya M., Shioda N. et al. Cytoprotective effect of bis(1-oxy-2- pyridinethiolato)oxovanadium (IV) on myocardial ischemia / reperfusion injury elicits inhibition of Fas ligand and Bim expression and elevation of FLIP expression // Eur. J. Pharmacol. – 2007. – Vol. 571. – P. 180–188.

Bhuiyan M., Takada Y., Shioda N. et al. Cardioprotective effect of vanadyl sulfate on ischemia/reperfusion-induced injury in rat heart in vivo is mediated by activation of protein kinase B and induction of FLICE-inhibitory protein // Cardiovascular Therapeutics. – 2008. – Vol. 26. – P. 10–23.

Bhuiyan M., Fukunaga K. Cardioprotection by vanadium compounds targeting Akt-mediated signaling // J. Pharmacol. Sci. – 2009. – Vol. 110. – P. 1–13.

Bhuiyan M., Norifumi S., Masatoshi S. et al. Activation of endothelial nitric oxide synthase by a vanadium compound ameliorates pressure overload-induced cardiac injury in ovariectomized rats // Hypertension. – 2009. – Vol. 53. – P. 57–63.

Bishayee A., Oinam S., Basu M., Chatterjee M. Vanadium chemoprevention of 7,12-dimethybenz()anthracene-induced rat mammary carcinogenesis: probable involvement of representative hepatic phase I and II xenobiotic metabolizing enzymes // Breast Cancer Res. Treat. – 2000. – Vol. 63. – P. 133–145.

Black W.A.P., Mitchell R.L. Trace elements in the common brown algae and in seawater // J. Mar. Biol. Assoc. UK. – 1952. – Vol. 30. – P. 575–584.

Bollen M., Miralpeix M., Ventura B. et al. Oral administration of vanadate to streptozotocin-diabetic rats restores the glucose-induced activation of liver glycogen syntase // Biochem J. – 1990. – Vol. 267. – P. 269–271.

Bosch F., Hatzoglou M., Park E.A., Hanson B.W. Vanadate inhibits expression of the gene phosphoenolpyruvate carboxykinase (GTP) in rat hepatoma cells // J Cell Biochem. – 1990. – Vol. 23. – P. 13677–13682.

Boulassel B., Sadeg N., Roussel O. et al. Fatal poisoning by vanadium // Forensic Sci. Internat. – 2011. – Vol. 206. – P. e79–e81.

Brichard S.M., Bailey C.J., Henquin J.C. Marked improvement of glucose homeostasis in diabetic ob/ob mice given oral vanadate // Diabetes. – 1990. – Vol. 11. – P. 1326–1332.

Brichard S., Desbuquois B., Girard J. Vanadate treatment of diabetic rats reverses the impaired expression of genes involved in hepatic glucose metabolism: effects on glucolitic and glukoneogenetic enzymes, and on glucose transporter GLUT2 // Mol. Cell. Endocrinol. – 1993. – Vol. 91. – P. 91–97.

Brichard S.M., Ongemba L.N., Girard J., Henquin J.C. Tissue-specific correction of lipogenic enzyme gene expression in diabetic rats given vanadate // Diabetologia. – 1994. – Vol. 37. – P. 1065–1072.

Brooks R.R., Rumsby M.G. The biogeochemistry of trace element uptake by some New Zealand bivalves // Limnol. Oceanogr. – 1965. – Vol. 10. – P. 521–527.

Butler A. Vanadium bromoperoxidase // Bioinorganic Catalysis / Ed. by J. Reedijk. – New York : Marcel Dekker, 1992. – P. 425–445.

Butler A., Carter-Franklin J.N. The role of vanadium bromoperoxidase in the biosynthesis of halogenated marine natural products // Nat. Prod. Rep. – 2004. – Vol. 21. – P. 180–188.

Butler A., Sandy M. Mechanistic considerations of halogenating enzymes // Nature. – 2009. – Vol. 460. – P. 848–854.

Byrne A.R., Costa L. Vanadium in foods and human body fluids and tissues // Sci. Total Environ. – 1978. – Vol. 10. – P. 17–30.

Cantley L.C., Josephson L., Warner R. et al. Vanadate is a potent (Na,K)-ATPase inhibitor found in ATP derived from muscle // J Biol Chem. – 1977. – Vol. 252. – P. 7421–7423.

Cerecetto H., Gonzlez M. Anti-T. cruzi agents: our experience in the evaluation of more than five hundred compounds // Mini Rev. Med. Chem. – 2008. – Vol. 8. – P. 1355–1383.

Chakraborty A., Ghosh R., Roy K. et al. Vanadium: a modifier of drug metabolizing enzyme patterns and its critical role in cellular proliferation in transplantable murine lymphoma // Oncology. – 1995. – Vol. 52. – P. 310–314.

Chakraborty A., Selvaraj S. Differential modulation of xenobiotic metabolizing enzymes by vanadium during diethylnitrosamine-induced hepatocarcinogenesis in Sprangue-Dawley rats // Neoplasma. – 2000. – Vol. 47. – P. 81–89.

Chasteen N.D. The biochemistry of vanadium // Struct and Bonding. – 1983. – Vol. 53. – P. 104–138.

Cheta D., Orasanu G., Nicolaie T. et al. The influence of sodium metavanadate on the process of diabetogenesis in BB rats // J. Cell. Mol. Med. – 2003. – Vol. 7. – P. 447–454.

Cho H., Thorvaldsen J.L., Chu Q. et al. Akt1/ PKBalpha is required for normal growth but dispensable for maintenance of glucose homeostasis in mice // J Biol Chem. – 2001. – Vol. 276. – P. 38349–38352.

Clancy R. M., Leszczynska-Piziak J., Abramson S. B. J. Nitric oxide reacts with intracellular glutathione and activates the hexose monophosphate shunt in human neutrophils: evidence for S-nitrosoglutathione as a bioactive intermediary // Clin. Invest. – 1992. – Vol. 90. – P. 1116– 1121.

Colin M., Madoulet C., Baccard N. et al. Study of sodium orthovanadate as a reverser of multidrug resistance on lymphoblastic leukemic CEM/VLB100 cells // Anticancer Res. – 1994. – Vol. 14. – P. 2383–2387.

Conconi M. T., De Carlo E., Vigolo S. et al. Effects of some vanadyl coordination compounds on the in vitro insulin release from rat pancreatic islets // Horm. Metab. Res. – 2003. – Vol. 35. – P. 402–406.

Cortijo J., Villagrasa Vol., Marti-Cabrera M. et al. The spasmogenic effects of vanadate in human isolated bronchus // Br. J. Pharmacol. – 1997. – Vol. 121. – P. 1339–1349.

Cortizo A.M., Bruzzonne L., Molinueve S., Echevery S.B. A possible role of oxidative stress in the vanadium-induced cytotoxicity and the MC3T3E1 osteoblast and UMR106 osteosarcoma cell lines // Toxicology. – 2000. – Vol. 147. – P. 89– 99.

Chang Z., Sitachitta N., Rossi J.V. et al. Biosynthetic pathway and gene cluster analysis of curacin A, an antitubulin natural product from the tropical marine cyanobacterium Lyngbya majuscula // J. Nat. Prod. – 2004. –Vol. 67. – P. 1356– 1367.

Charles R., Cornman E., Zovinka P. et al. Structural and EPR studies of vanadium complexes of deprotonated amide ligands: effects on the 51V hyperfine coupling constant // Inorg. Сhem. – 1995. – Vol. 34. – P. 4213–4219.

Chasteen N.D. The Biochemistry of vanadium // Struct. Bonding. – 1983. – Vol. 53. – P. 105– 137.

Chong I.W., Shi M.M., Love J.A. et al. Regulation of mRNA expression in a rat model of vanadium-induced pulmonary inflammation // Inflammation. – 2000. – Vol. 24. – P. 505–517.

Coulombe R.A.J., Briskin D.P., Keller R.J. et al. Vanadate-dependent oxidation of pyridine nucleotides in rat liver microsomal membranes // Biochim. Biophys. Acta. – 1987. – Vol. 255. – P. 267–273.

Courville P., Chaloupka R., Cellie M.F. Recent progress in structure-function analyses of NRAMP proton-dependent metal-ion transporters // Biochem. Cell Biol. – 2006. – Vol. 84. – P. 960– 978.

Crans D.C., Simone C.M., Saha A.K., Glew R.H. Vanadate monomers and dimers both inhibit the human prostatic acid-phosphatase // Biochem. Biophys. Res. Commun. – 1989. – Vol. 1675. – P. 246–250.

Crans D.C., Willging E.M., Butler S.R. Vanadate tetramer as the inhibiting species in enzymereactions in vitro and in vivo // J. Amer. Chem. Soc. – 1990. – Vol. 112. – P. 427–432.

Crockford S.J. Evolutionary roots of iodine and thyroid hormones in cell–cell signaling // Integr. Comp. Biol. – 2009. – Vol. 49. – P. 155–166.

Cupo M.A., Donaldson W.E. Chromium and vanadium effects on glucose metabolism and lipid synthesis in the chick // Poult. Sci. – 1987. – Vol. 66. – P. 120–126.

Curran G.L., Azarnoff D.L., Bohlinger R.E. Effect of cholesterol synthesis inhibition in normocholesteremic young men // J Clin. Invest. – 1959. – Vol. 38. – P. 1251–1261.

Сui W., Cui H.M., Peng Xi. et al. Dietary vanadium induces decrease in antioxidant enzyme activities and oxidative stress in the spleens of broilers // Med. Chem. – 2012. – Vol. 2. – № 2. – P. 1–5.

Dai S., Thompson K.H., Vera E., McNeill J.H. Toxicity studies on one-year treatment of non-diabetic and streptozotocin-diabetic rats with vanadyl sulphate // Pharmacol. Toxicol. – 1994. – Vol. 75. – P. 265–273.

Dart R.C. (ed.) Medical Toxicology. – 3rd edn. – Baltimore, MD : Williams and Wilkins, 2004.

Dharwan S., Singh S., Aggarwal B.B. Induction of endothelial cell surface adhesion molecules by tumor necrosis factor is blocked by protein tyrosine phosphatase inhibitors: role of nuclear transcription factor NF-B // Eur Immunol. – 1997. – Vol. 27. – P. 2172–2179.

Dikanov S.A., Liboiron B.D., Thompson K.H. et al. In vivo electron spin-echo envelope modulation (ESEEM) spectroscopy: First observation of vanadyl coordination to phosphate in bone // J. Am. Chem. Soc. – 1999. – Vol. 121. – P. 11004– 11005.

Dingley A.L., Kustin K., Macara I.G., McLeod G.C. Accumulation of vandium by tunicate blood cells occurs via a specific anion transport system // Biochim. Biophys. Acta. – 1981. – Vol. 649. – P. 493–502.

Djordjevitz C., Wampler G.L. Antitumor activity of peroxoheteroligant vanadates (V) in relation to biochemistry of vanadium // J. Inorg. Biochem. – 1985. – Vol. 25. – P. 51–55.

Djordjevic С., Vuletic N., Renslo M.L. et al. Peroxo heteroligant vanadates (V): synthesis, spectra-structure relationships and stability toward decomposition // Mol. Cell. Biochem. – 1995. – Vol. 153. – P. 25–29.

Domingo J.L., Gomez M., Llobet J.M., Corbella J. Chelating agents in the treatment of acute vanadyl sulphate intoxication in mice // Toxicology. – 1990. – Vol. 62. – P. 203–211.

Duffus J.H. Carcinogenity classification of vanadium pentoxide and inorganic vanadium compounds, the NTP study of carcinogenity of inhaled vanadium pentoxide, and vanadium 91 chemistry // Regul. Toxicol. Pharmacol. – 2007. – Vol. 47. – P. 110–114.

Eady R.R. Vanadium nitrogenases of azotobacter // Metal Ions Biol. Syst. – 1995. – Vol. 31. – P. 363–405.

El-Naggar M.M., El-Waseef A.M., El-Halafawy K.M., El-Sayed I.H. Antitumor activities of vanadium (IV), manganese (IV), iron (III), cobalt (II) and copper (II) complexes of 2-methylaminopyridine // Cancer Lett. – 1998. – Vol. 133. – P. 71–76.

English L., Macara J.G., Cantley L.C. Vanadium stimulates (Na,K)ATPase in friend еrythroleukemia cells and blocks erythropoiesis // J. Cell. Biol. – 1997. – Vol. 983. – P. 1299–12302.

Evangelou A., Karkabounas S., Kalpouzos G. et al. Comparison of the therapeutic effects of two vanadium complexes administered at low doses on benzoapyrene-induced malignant tumors in rats // Cancer Letters. – 1997. – Vol. 119. – P. 221–225.

Evangelou A. Vanadium in cancer treatment // Crit. Rev. Oncol. Hematol. – 2002. – Vol. 42. – P. 249–265.

Fantus I. G., Kadota S., Deragon G. et al. Pervanadate peroxide(s) of vanadate mimics insulin action in rat adipocytes via activation of the insulin receptor tyrosine kinase // Biochemistry. – 1989. – Vol. 28. – P. 8864–8871.

Faure R., Vincent M., Dufour M. et al. Arrest of the G2/M transition of the cell cycle by protein tyrosine phosphatase inhibition: studies on a neuronal and a glial cell line // J. Cell. Biochem. – 1995. – P. 389–401.

Faulkner D.J. Marine natural products // Nat. Prod. Rep. – 2002. – Vol. 19. – P. 1–66.

Faulkner-Hudson T.G. Vanadium: Toxicology and Biological Significance. – Amsterdam : Elsevier, 1964. – 135 p.

Fedorova E.V., Rybakov V.B., Senyavi V.M. et al. Synthesis and crystal structure of sodium (2,2-bipyridyl)oxodiperoxovanadate(V) octahydrate // Russ. J. Coord. Chem. – 2002. – Vol. 28. – P. 483–486.

Ferrer E., Salinas M.V., Correa M.J. et al. Synthesis, characterization, antitumoral and osteogenic activities of quercetin vanadyl(IV) complexes // J. Biol. Inorg. Chem. – 2006. – Vol. 11. – P. 791–801.

Folbergova J., Lisa V., Mares V. Na+-K+ATPase activity in cultured C6 glioma cells // Neurochem. Res. – 1989. – Vol. 14. – P. 392–439.

Food and Nutrition Board. National Research Council: Recommended Dietary Allowances. 10th ed. –Washington, DC : National Academy Press, 1989.

Frank P., Hedman B., Hodgson K.O. Sulfur allocation and vanadium-sulfate interactions in whole blood cells from the tunicate Ascidia ceratodes, investigated using X-ray absorption spectroscopy // Inorg. Chem. – 1999. – Vol. 38. – P. 260–270.

Gail R., Willsky L.-H., Chi M. et al. Anti-diabetic effects of a series of vanadium dipicolinate complexes in rats with streptozotocin-induced diabetes // Coordination Chem. Rev. – 2011. – Vol. 255. – P. 2258–2269.

Gil J., Miralpeix M., Carreras J., Bartrons R. Insulin-like effects of vanadate on glucokinase activity and fructose 2,6-bisphosphate levels in the liver of diabetic rats // J. Biol. Chem. – 1988. – Vol. 263. – P. 1868–1871.

Germinario R. J., Colby-Germinario S.P., Posner B. I., Nahm K. Different forms of vanadate on sugar transport in insulin target and nontarget cells // J. Biomed. Biotechnol. – 2002. – Vol. 2. – № 1. – P. 22–30.

Gioacchino M., Sabbioni E., Di Giampaolo L. et al. In vitro effects of vanadate on human immune functions // Ann. Clin. Lab. Sci. – 2002. – Vol. 32. – P. 148–154.

Golden M.H., Golden B.E. Trace elements, potential importance in human nutrition with particular reference to zinc and vanadium // Br. Med. Bull. – 1981. – Vol. 37. – P. 31–36.

Gresser M.J., Tracey A.S., Chasteen N.D. Vanadates as phosphate analogs in biochemistry // Vanadium in Biological Systems. – Dordrecht, Boston, London : Kluwer Academic Publishers, 1990. – P. 63–79.

Gummow B. Vanadium Mining and Cattle Health. Sentinel Studies, Epidemiological and Veterinary Public Health Issues. PhD Thesis. – Utrecht : University of Utrecht, 2005.

Gummow B. Vanadium: environmental pollution and health effects // Encyclopedia of Environmental Health. – Elsevier, 2011. – P. 628–636.

Hackbarth I., Schmitz W., Scholz H. et al. Positive inotropism of vanadate in cat papillary muscle // Nature. – 1978. – Vol. 275. – P. 67–68.

Hagenmeyer A., Wierichs R., Bader H. Vanadate inhibition of Ca2+-ATPase of sarcoplasmic reticulum from pig heart // Basic Res. Cardiol. – 1980. – Vol. 75. – P. 452–454.

Hajjar J.J., Fucci J.C., Rowe W.A., Tomicic T.K. Effect off vanadate on amino acid transport in rat jejunum // Proc. Soc. Exp. Biol. Med. – 1987. – Vol. 184. – P. 403–409.

Hales B.J., Langosch D.J., Case E.E. Isolation and characterization of a second nitrogenase Fe-protein from Azotobacter vinelandii // J. Biol. Chem. – 1986. – Vol. 261. – P. 15301–15306.

Hanada M., Feng J., Hemmings B.A. Structure, regulation and function of PKB/AKT – a major therapeutic target // Biochim Biophys Acta. – 2004. – Vol. 1697. – P. 3–16.

Hanauske U., Hanauske A.R., Marshal M.H. et al. Biphasic effects of vanadium salts on in vitro tumor colony growth // Int. J. Cell. Cloning. – 1987. – Vol. 5. – P. 170–178.

Hirano S., Suzuki K.T. Exposure metabolism and toxicity for rare earths and related compounds // Environ. Health. Perspect. – 1996. – Vol. 104. – P. 85–95.

Henze M. Untersuchungen uber das Blut der Ascidien. I. Mittielung. Die vanadium-verbinding der Blutkorchen // Hoppe-Seyer‘s Z. Physiol. Chem. – 1911. – Vol. 72. – P. 494–501.

Honn K.V., Tang D.G. Adhesion molecules and cancer cell interaction with endothelium and subendothelial matrix // Cancer Metastasis Rev. – 1992. – Vol. 11. – P. 353–375.

Hopkins L.L., Mohr H.E. Vanadium as an essential nutrient // Fed. Proc. – 1974. – Vol. 33. – P. 1773–1175.

http://integrity.thomson-pharma.com.

http://minerals.usgs.gov/minerals/pubs/ commodity/vanadium/mcs-2012-vanad.pdf.

Hughes J.M., Schindler M., Rakovan J.F., Cureton F.E. The crystal structure of hummerite, KstrMucgt(uVr5aOl 1u4)n8itHs 2aOn:dBthoendiKng2Mbegt2w(Hee2On )t1h6e iVnt1e0rOs2t8i- tial complex // Can. Mineral. – 2002. – Vol. 40. – P. 1429–1435.

Hughes J.M., Schindler M., Francis C.A. The C2/m disordered structure of pascoite, Ca3V10O2817H2O: Bonding between structural units and interstitial complexes, in compounds containing the V10O286- decavanadanate polyanion // Can. Mineral. – 2005. – Vol. 43. – P. 1379– 1386.

Itkes A.V., Imamova L.R., Alexandrova N.M. et al. Expression of c-myc gene in human ovary carcinoma cells treated with vanadate // Exp Cell Res. – 1990. – Vol. 188. – P. 169–171.

Josephson L., Cantley L.C. Isolation of a potent (Na,K)-ATPase inhibitor from striated muscle // Biochemistry. – 1977. – Vol. 16. – С. 4572– 4578.

Kawakami N., Ueki T., Amata Y. et al. A novel vanadium reductase, Vanabin2, forms a possible cascade involved in electron transfer // Biochim. Biophys. Acta. – 2009. – Vol. 1794. – P. 674–679.

Khandelwal R., Pugazhenti S. In vivo effects of vanadate on hepatic glycogen metabolizing and lipogenic enzymes in insulin-dependent and insulin-resistant diabetic animals // Mol. Cell. Biochem. – 1995. – Vol. 153. – P. 87–94.

Korbecki J., Baranowska-Bosiacka I., Gutowska I., Chlubek D. Biochemical and medical importance of vanadium compounds // Acta Biochim. Polon. – 2012. – Vol. 59. – P. 195–200.

Kopf-Maier P., Krahl D. Tumor inhibition by metalocenes: ultrastructural localization of titanium and vanadium in treated tumor cells by electron energy loss spectroscopy // Chem. Biol. Interact. – 1983. – Vol. 44. – P. 317–328.

Koyuturk M., Tunali S., Bolkent S., Yanardag R. Biological effects of vanadyl sulfate on liver of streptozotocin-induced diabetic rats // Trace Elem. Res. – 2005. – Vol. 104. – P. 233–247.

Krady M.M., Freyermuth S., Rogue P., Malviya A.N. Pervanadate elicits proliferation and mediates mitogen-activated protein (MAP) // FEBS Lett. – 1997. – Vol. 412. – P. 420–424.

Krenn B.E., Tromp M.G., Wever R. The brown alga Ascophyllum nodosum contains two different vanadium bromoperoxidases // J. Biol. Chem. – 1989. – Vol. 264. – P. 19287–19292.

Kresja C. M., Nadler S. G., Esselstyn J. M. et al. Role of oxidative stress in the action of vanadium phosphotyrosine phosphatase inhibitors // J. Biol. Chem. – 1997. – Vol. 272. – P. 11541–11549.

Haldara K. A., Banerjee S., Naskar K. et al. Sub-optimal dose of Sodium 126. Antimony Gluconate (SAG)-diperoxovanadate combination clears organ parasites from BALB/c mice infected with antimony resistant Leishmania donovani by expanding antileishmanial T-cell repertoire and increasing IFN-gamma to IL-10 ratio // Exp. Parasitol. – 2009. – Vol. 122. – P. 145–154.

Lа Barre S., Potin P., Leblanc C., Delage L. The halogenated metabolism of brown algae (Phaeophyta), its biological importance and its environmental significance // Mar. Drugs. – 2010. – Vol. 8. – P. 988–1010.

Lambert L.A., Perri H., Meehan T.J. Evolution of duplications in the transferrin family of proteins // Comp. Biochem. Physiol. B. Biochem. Mol. Biol. – 2005. – Vol. 140. – P. 11–25.

Lambert L.A., Mitchell S.L. Molecular evolution of the transferrin receptor/glutamate carboxypeptidase II family // J. Mol. Evol. – 2007. – Vol. 64. – P. 113–128.

Leon-Lai C.H., Gresser M.J., Tracey A.S. Influence of vanadium(V) complexes on the catalytic activity of ribonuclease A. The role of vanadate complexes as transition state analogues to reactions at phosphate // Can. J. Chem. – 1996. – Vol. 74. – P. 38–48.

Liasko R., Karkabounas S., Kabanos Th. et al. Antitumor effects of a vanadium complex with cysteine on malignant cell lines and tumor-bearing Wistar rats // Metal Ions Biol. Med. – 2000. – Vol. 6. – P. 577–579.

Li S.H., McNeill J.H. In vivo effects of vanadium of GLUT-4 translocation in cardiac tissue of STZ-diabetic rats // Mol. Cell. Biochem. – 2001. – Vol. 212. – P. 121–129.

Li Z., Cartier J.D., Dailey L.A., Huang Y.C. Vanadyl sulfate inhibits NO production via threonine phosphorylation of eNOS // Environ. Health Perspect. – 2004. – Vol. 112. – P. 201–206.

Littlechild J. Haloperoxidases and their role in biotransformation reactions // Curr. Opin. Chem. Biol. – 1999. – Vol. 3. – P. 28–34.

Liu X., Kim C. N., Yang J. et al. Induction of apoptotic program in cell-free extracts: Requirements for dATP and cytochrome C // Cell. – 1996. – Vol. 86. – P. 147–157.

Lu B., Ennis D., Lai R. et al. Enhanced sensitivity of insulin-resistant adipocytes to vanadate is associated with oxidative stress and decreased reduction of vanadate (+5) to vanadyl (+4) // J. Biol. Chem. – 2001. – Vol. 276. – P. 35589–35598.

Maier R.H., Purser S.M., Nicholson D.L., Pories W.J. The cytotoxic interaction of inorganic trace elements with EDTA and cisplatin in sensitive and resistant human avarian cancer cells // In Vitro Cell. Dev. Biol. Anim. – 1997. – Vol. 33. – P. 218–221.

Mancini M., Nicholson D.W., Roy S. et al. The caspase-3 precursor has a cytosolic and mitochondrial distribution: implications for apoptotic signaling // J. Cell Biol. – 1998. – Vol. 140. – P. 1485–1495.

Martin J.H., Knauer G.A. The elemental composition of plankton // Geochim. Cosmochim. Acta. – 1973. – Vol. 37. – P. 1639–1653.

Maschek J.A., Baker B.J. The chemistry of algal secondary metabolism // Algal Chemical Ecology / Ed. by Amsler C.D. – Berlin, Heidelberg : Springer-Verlag, 2008. – P. 1–24. 93

Matsuda M., Mandarino L., DeFronzo R. Synergistic interaction of magnesium and vanadate on glucose metabolism in diabetic rats // Metabolism. – 1999. – Vol. 48. – P. 725–731.

Maurya M., Agarwal S., Abid M. et al. Dioxoand oxovanadium(V) complexes of thiohydrazone ONS donor ligands: synthesis, characterization, reactivity, and antiamoebic activity // Inorg. Chem. – 2006. – Vol. 45. – P. 1260–1269.

Maurya M.R., Agarwal S., Abid M. et al. Synthesis, characterisation, reactivity and in vitro antiamoebic activity of hydrazone based oxovanadium(IV), oxovanadium(V) and mubis(oxo)bisoxovanadium(V) complexes // Dalton Trans. – 2006. – Vol. 21. – P. 937–947.

Messerschmidt A., Wever R. X-ray structure of a vanadium-containing enzyme: chloroperoxidase from the fungus Curvularia inaequalis // Proc. Natl. Acad. Sci. U.S.A. – 1996. – Vol. 93. – P. 392–396.

Michibata H., Iwata Y., Hirata J. Isolation of highly acidic and vanadium-containing blood cells from among several types of blood cells in Ascidiidae species by density-gradient centrifugation // J. Exp. Zool. – 1991. – Vol. 257. – P. 306–313.

Michibata H., Terada T., Anada N et al.. The accumulation and distribution of vanadium, iron, and manganese in some solitary ascidians // Biol. Bull. – 1986. – Vol. 171. – P. 672–681.

Mohammad A., Sharma V., McNeill J.H. Vanadium increases GLUT4 in diabetic rat skeletal muscle // Mol. Cell. Biochem. – 2002. – Vol. 233. – P. 139–143.

Mohammadi M., Yazdanparast R. Methoxy VO-salen complex: in vitro antioxidant activity, cytotoxicity evaluation and protective effect on CCl4-induced oxidative stress in rats // Food Chem. Toxicol. – 2009. – Vol. 47. – P. 716–721.

Mongold J.J., Cros G.H., Vian L. et al. Toxicological aspects of vanadyl sulphate on diabetic rats: effects on vanadium levels and pancreatic Bcell morphology // Pharmacol. Toxicol. – 1990. – Vol. 67. – P. 192–198.

Montero M.R., Guerri C., Grisolia S. Vanadate, alcohol or both increase passive membrane permeability of neuro-2a cells; lesser sensitivity of Hep2 cells // Life Sci. – 1981. – Vol. 28. – P. 641– 646.

Motoyashiki T. et al. Involvement of the rapid increase in cAMP contentin the vanadyl-stmulated release of lipoprotein lipase activity from rat fat pads // Biol. Pharm. Bull. – 1996. – Vol. 19. – P. 1412–1416.

Motoyashiki T. et al. Involvement of adenosine in vanadate-stimulated release of lipoprotein lipase activity // Biol. Pharm. Bull. – 1998. – Vol. 21. – P. 889–892.

Mosseri R., Waner T., Shefi M. et al. Gluconeogenesis in non-obese diabetic (NOD) mice: in vivo effects of vandadate treatment on hepatic glucose6-phoshatase and phosphoenolpyruvate carboxykinase // Metabolism. – 2000. – Vol. 49. – P. 321–325.

Narla R., Dong Y., D’Cruz O.J. et al. Bis(4,7-dimethyl-1,10-phenanthroline) sulfatooxovanadium (IV) as a novel apoptosis-inducing anticancer agent // Clin. Cancer Res. – 2000. – Vol. 6. – P. 1546–1556.

Nechay B.R. Mechanisms of action of vanadium // Annu. Rev. Pharmacol.Toxicol. – 1984. – Vol. 24. – P. 501–524.

Nechay B.R., Nanninga L.B., Nechay P.S.E. et al. Role of vanadium in biology // Fed. Proc. – 1986. – Vol. 45. – P. 123–132.

Nevo Y., Nelson N. The NRAMP family of metal-ion transporters // Biochem. Biophys Acta. – 2006. – Vol. 1763. – P. 609–620.

Nicholls G.D., Curl H., Bowen V.T. Spectrographic analysis of marine plankton // Limnol. Oceanog. – 1959. – Vol. 4. – P. 472–478.

Nielsen F.H., Uthus E.O. Vanadium in Biological Systems: Physiology and Biochemistry / Ed. by N.D. Chasteen. – London : Kluwer Academic, 1990. – P. 51–62.

Nilsson J., Shteinman A. A., Degerman E. et al. Salicylamide and salicylglycine oxidovanadium complexes with insulin-mimetic properties // J. Inorg. Bioch. – 2011. – Vol. 105. – P. 1795–1800.

Noda C., Masuda T., Sato K. et al. Vanadate improves cardiac function and myocardial energy metabolism in diabetic rat hearts // Jpn. Heart J. – 2002. – Vol. 44. – P. 745–757.

Nour-Eldeen A.F., Craig M.M., Gresser M.J. Interaction of inorganic vanadate with glucose-6-phosphate dehydrogenase, nonenzymatic formation of glucose 6-vanadate // J. Biol. Chem. – 1985. – Vol. 260. – P. 6836–6842.

Ozaki H., Urakawa N. Effects of vanadate on mechanical responses and Na-K pump in vascular smooth muscle // Eur. J. Pharmacol. – 1980. – Vol. 68. – P. 339–347.

Patterson B.W., Hansard II S.L., Ammerman C.B. et al. Kinetic model of whole body vanadium metabolism: studies in sheep // Am. J. Physiol. – 1986. – Vol. 251. – P. R325–R332.

Plat H., Krenn B.E., Wever R. The bromoperoxidase from the lichen Xanthoria parietina is a novel vanadium enzyme // Biochem J. – 1987. – Vol. 248. – P. 277–279.

Pugazhenti S., Angel J., Khandelwal R. Long-term effects of vanadate treatment on glycogen metabolizing and lipogenic enzymes of liver in genetically diabetic (db/db) mice // Metabolism. – 1991. – Vol. 40. – P. 941–946.

Pyle T.E., Tieh T.T. Strontium, vanadium and zinc in the shells of pteropods // Limnol. Oceanogr. – 1970. – Vol. 15. – P. 153–154.

Ramanadham M., Kern M. Differential effects of vanadate on DNA synthesis induced by mitogens in T and B lymphocytes // Mol. Cell Biochem. – 1983. – Vol. 5. – P. 67–71.

Ramanadham S., Brownsey R.W., Cros G. H. et al. Sustained prevention of myocardial and metabolic abnormalities in diabetic rats following withdrawal from oral vanadyl treatment // Metabolism. – 1989. – Vol. 38. – P. 1022–1028.

Ray W.J., Burgner J.W., Post C.B. Characterization of vanadate-based transitionstate-analog complexes of phosphoglucomutase by spectral and NMR techniques // Biochemistry. – 1990. – Vol. 29. – P. 2770–2778.

Ray W.J., Post C.B. The oxyvanadium constellation in transition-state-analog complexes of phosphoglucomutase and ribonuclease – structural deductions from electron-transfer spectra // Biochemistry. – 1990. – Vol. 29. – P. 2779–2789.

Robinson J.D. Vanadate inhibition of brain (Ca 2+ Mg2+)-ATPase // Neurochem. Res. – 1981. – Vol. 6. – P. 225–232.

Robson R.L., Eady R.E., Richardson T.H. et al. The alternative nitrogenase of Azotobacter chroococcum is a vanadium enzyme // Nature. – 1986. – Vol. 322. – P. 388–390.

Roshchin A.V., Ordzhonikidze E.K., Shalganova I.V. Vanadium – toxicity, metabolism, carrier state // J. Hyg. Epidemiol. Microbiol. Immunol. – 1980. – Vol. 24. – P. 377–383.

Rossetti L., Laughlin M. Correction of chronic hyperglycemia with vanadate, but not with phlorizin, normalizes in vivo glycogen repletion and in vitro glycogen synthase activity in diabetic skeletal muscle // J. Clin. Invest. – 1989. – Vol. 84. – P. 892–899.

Rowley A.F. Preliminary investigations on the possible antimicrobial properties of tunicate blood cell vanadium // J. Exp. Zool. – 1983. – Vol. 227. – P. 319–322.

Rychcik M., Skyllas-Kazacos M. Characteristics of a new all-vanadium redox flow battery // J. Power Sources. – 1988. – № 1. – P. 59– 67.

Sabbioni E., Marrafante E. Metabollic pattern of vanadium in the rat // Bioinorg. Chem. – 1978. – Vol. 9. – P. 389–407.

Sakurai H., Tsuchiya K., Nakatsuka M. et al. Insulin-like effect of vanadyl ion on streptozocininduced diabetic rats // J. Endocrinol. – 1990. – Vol. 126. – P. 451–459.

Sandirasegarane L, Gopalakrishnan V. Vanadate increases cytosolic free calcium in rat aortic smooth muscle cells // Life Sci. – 1995. – Vol. 56. – P. PL169–PL174.

Sanna D., Garribba E., Micera G. Interaction of VO2+ ion with human serum transferrin and albumin // J. Inorg. Biochem. – 2009. – Vol. 103. – P. 648–655.

Sardar S., Mondal A., Chatterjee M. Protective role of vanadium in the survival of hosts during the growth of atransplantable murine lymphoma and its profound effects on the rates and patterns of biotransformation // Neoplasma. – 1993. – Vol. 40. – P. 27–30.

Schlake H.P., Bertram H.P., Husstedt I.W. Acute systemic vanadate poisoning presenting as cerebrovascular ischemia with prolonged reversible neurological deficits (PRIND) // Clin. Neurol. Neurosurg. – 1994. – Vol. 96. – P. 92–95.

Sera M., Tanaka K., Morita T., Ueki H. Increasing effect of vanadate on lipoprotein lipase activity in isolated rat fat pads // Arch. Biochem. Biophys. – 1990. – Vol. 279. – P. 291–297.

Serra M., Sabbioni E., Pintar A., Casella L. Vanadium effect on the activity of horseradish peroxidase, catalase, glutathione peroxidase, and superoxide dismutase in vitro // J. Inorg. Chem. – 1992. – Vol. 46. – P. 161–174.

Shah K.R., Matsubara T., Foerster D.R. Mechanisms of inotropic responses of the isolated rat hearts to vanadate // Int. J. Cardiol. – 1995. – Vol. 52. – P. 101–113.

Shi X., Dalai N.S. Vanadate-mediated hydroxyl radical generation from superoxide radical in the presence of NADH: Haber-Weiss vs. Fenton mechanism // Arch. Biochem. Biophys. – 1993. – Vol. 3. – P. 336–341.

Skulachev V.P. Cytochrome C in the apoptotic and antioxidant cascades // FEBS Letters. – 1998. – Vol. 423. – P. 275–280.

Soedjak H.S., Butler A. Characterization of vanadium bromoperoxidase from Macrocystis and Fucus: reactivity of vanadium bromoperoxidase toward acyl and alkyl peroxides and bromination of amines // Biochemistry. – 1990. – Vol. 29. – P. 7974–7981.

Sremark R., Ullberg S., Appelgren L.E. Autoradiographic localization of V48-labelled vanadium pentoxide V2O in developing teeth and bones of rats // Acta Odontol. Scand. – 1962. – Vol. 20. – P. 225–232.

Sremark R., Ullberg S. Distribution and Rkiandeitoicissootofp4e8Vs i2nOA5 innimmailcBe i/o/loPgryoca.nSdytmhepM. Uesdeicoafl Sciences. – Mexico City, New York : Academic Press, 1962. – Vol. 2. – P. 103–114.

Srivastava C., Srivastava A. Vanadium and the cardiovascular functions // Can. J. Physiol. Pharmacol. – 2004. – Vol. 82. – P. 833–839.

Stacey N.H., Klaassen C.D. Inhibition of lipid peroxidation without prevention of cellular injury in isolated rat hepatocytes // Toxicol. Appl. Pharmacol. – 1981. – Vol. 58. – P. 8–15.

Stacey N.H., Kappus H. Comparison of methods of assessment of metal-induced lipid peroxidation in isolated rat hepatocytes // J. Toxicol. Environ. Health. – 1982. – Vol. 9. – P. 277–281.

Stankiewicz P.J., Gresser M.J., Tracey A.S., Hass L.F. 2,3-Diphosphoglycerate phosphatase activity of phosphoglycerate mutase – stimulation by vanadate and phosphate // Biochemistry. – 1987. – Vol. 26. – P. 1264–1269.

Stoecker D. Distribution of ascid and vanadium in Rhopalaea birkelandi Tokioka // J. Exp. Mar. Biol. Ecol. – 1980. – Vol. 48. – P. 277– 281.

Stone J.R., Marletta M.A. The ferrous heme of soluble guanylate cyclase: formation of hexacoordinate complexes with carbon monoxide and nitrosomethane // Biochemistry. – 1995. – Vol. 34. – P. 16397–16403.

Strasia C.A. Vanadium: essentiality and toxicity in the laboratory rat. Ph.D. Thesis. – Ann Arbor: University Microfilms, 1971.

Sugiyama H., Matsugo S., Misu H. et al. Regulation of the physiological effects of peroxidovanadium(V) complexes by the electronic nature of ligands // J. Inorg. Biochem. – 2013. – Vol. 121. – P. 66–76.

Susin S. A., Lorenzo H. K., Zamzami N. et al. Molecular characterization of mitochondrial apoptosis-inducing factor // Nature. – 1999. – Vol. 397. – P. 441–446.

Szabo C., Dawson V.L. Role of poly(ADPribose) synthetase in inflammation and ischaemiareperfusion // Trends Pharmacol. Sci. – 1998. – Vol. 19. – P. 287–298. 95

Takenaga K. Suppression of metastatic potential of highmetastatic Lewis lung sarcoma cells by vanadate, an inhibitor of tyrosine phosphotase, through inhibiting cell-substrate adhesion // Invasion Metastasis. – 1996. – Vol. 16. – P. 97–106.

Tamura S., Brown T.A., Whipple J.H. et al. Novel mechanism for the insulin-like effect of vanadate on glycogen synthase in rat adipocytes // J. Biol. Chem. – 1984. – Vol. 259. – P. 6650–6658.

Tawa R., Uchida K., Taniyama J. et al. A new insulin-mimetic vanadyl complex (Npyridylmethylaspartate) oxovanadium (IV) with VO(N2O2) coordination mode, and evaluation of its effects on uptake of D-glucose by Ehrlich ascites tumor cells // J. Pharm. Pharmacol. – 1999. – Vol. 51. – P. 119–124.

Taylor D.M., Williams D.R. Trace Element Medicine and Chelation Therapy. – Cambridge : Royal Society of Chemistry, 1995. – 138 p.

Thompson H.J., Chasteen D.N., Neeker L. Dietary vanadyl (IV) sulfate inhibits chemicallyinduced mammary carcinogenesis // Carcinogenesis. – 1984. – Vol. 5. – P. 849–851.

Thompson K.H., McNeill J.H. Effect of vanadyl sulfate feeding on susceptibility to peroxidative change in diabetic rats // Res. Commun. Chem. Pathol. Pharmacol. – 1993. – Vol. 80. – P. 187–200.

Thompson K.H., Lichter J., LeBel C. et al. Vanadium treatment of type 2 diabetes: a view to the future // J. Inorg. Biochem. – 2009. – Vol. 103. – P. 554–558.

Tracey A.S., Leon-Lai C.H. 1H- and 51V-NMR investigation of the complexes formed between vanadate and nucleosides // Inorg. Chem. – 1991. – Vol. 30. – P. 3200–3204.

Tromp M.G.M., Olafsson G., Krenn B.E., Wever R. Some structural aspects of vanadium bromoperoxidase from Ascophyllum nodosum // Biochim. Biophys. Acta. – 1990. – Vol. 1040. – P. 192–198.

Tunali S., Yanardag R. Effect of vanadyl sulfate on the status of lipid parameters and on stomach and spleen tissues of streptozotocin-induced diabetic rats // Pharmacol. Res. – 2006. – Vol. 53. – P. 271–277.

Ueki T., Takemoto K., Fayard B. et al. Scanning X-ray microscopy of living and freeze-dried blood cells in two vanadium-rich ascidian species, Phallusia mammillata and Ascidia sydneiensis samea // Zool. Sci. – 2002. – Vol. 19. – P. 27–35.

Ueki T., Michibata H. Molecular mechanism of the transport and reduction pathway of vanadium in ascidians // Coordination Chem. Rev. – 2011. – Vol. 255. – P. 2249– 2257.

Underwood E. Trace Elements in Human and Animal Nutrition. – 4th ed. – New York : Acad. Press, 1977. – 402 p.

Uyama T., Nose Y., Wuchiyama J. Finding of the same antigens in the polychaete, Pseudopotamilla occelata, as those in the vanadium-rich ascidian, Ascidia sydneiensis Samea // Zool. Sci. – 1997. – Vol. 14. – P. 43–47.

Vaillancourt F. H., Yeh E., Vosburg D.A. Nature’s inventory of halogenation catalysts: oxidative strategies predominate // Chem. Rev. – 2006. – Vol. 106. – P. 3364–3378.

Valera A., Rodriguez-Gil J., Bosch F. Vanadate treatment restores the expression of genes for key enzymes in the glucose and ketone bodies metabolism in the liver of diabetic rats // J. Clin. Invest. – 1993. – Vol. 92. – P. 4–11.

Whitfield F.B., Helidoniotis F., Shaw K.J., Svoronos D. Distribution of bromophenols in species of marine algae from Eastern Australia // J. Agric. Food. Chem. – 1999. – Vol. 47. – P. 2367– 2373.

Wei Y., Zhang C., Zhao P. A new salicylic acid-erivatized kojic acid vanadyl complex: Synthesis, characterization and anti-diabetic therapeutic potential // J. Inorg. Biochem. – 2011. – Vol. 105. – P. 1081–1085.

Werner H. A crystal-chemical approach to the composition and occurrence of vanadium minerals // The Canadian Mineralogist. – 2000. – Vol. 38. – P. 1443–1446.

WHO. International Programme on Chemical Safety, Environmental Health Criteria 61. – Geneva : World Health Organisation, 1988.

Woo E.S., Rice R.L., Lazo J.S. Cell cycle dependent subcellular distribution of Cdc25B subtypes // Oncogene. – 1999. – Vol. 18. – P. 2770– 2776.

Yamaguchi N., Togi A., Ueki T. Expressed sequence tag analysis of blood cells in the vanadium-rich ascidian, Ascidia sydneiensis Samea – a survey of genes for metal accumulation // Zool. Sci. – 2002. – Vol. 19. – P. 1001–1008.

Yanardag R., Bolkent S., Karabulut- Bulan O., Tunali S. Effects of vanadyl sulfate on kidney in experimental diabetes // Biol. Trace Elem. Res. – 2003. – Vol. 95. – P. 73–85.

Yang Z. Z., Tschopp O., Hemmings-Mieszc- zak M. et al. Protein kinase B alpha/Akt1 regulates placental development and fetal growth // J. Biol. Chem. – 2003. – Vol. 278. – P. 32124–32131.

Yoshihara M., Ueki T., Watanabe T. et al. Vanabin P, a novel vanadium-binding protein in the blood plasma of an ascidian, Ascidia sydnei- ensis Samea // Biochim. Biophys. Acta. – 2005. – Vol. 1730. – P. 206–214.

Young-Jin K., Young-Inn K., Sung-Nak C. Magnetic properties and coordination environ- ments of oxovanadium(IV) within VO(IV)N2S2 a2n0d00V. –OV(IoVl.)1S94. chromophores – P. 2155–2161. Polyhedron.

Yuen V.G., Vera E., Battell M.L. et al. Acute and chronic oral administration of bis(maltolato) oxovanadium(IV) in Zucker diabetic fatty (ZDF) rats. // Diab. Res. Clin. Prac. – 1999. – Vol. 43. – P. 9–19.

Zaporowska H. Effect of vanadium on L- ascorbic acid concentration in rat tissues // Gen Pharmacol. – 1994. – Vol. 25. – P. 467–470.

Zhang Z., Leonard S. S., Huang C. et al. Role of reactive oxygen species and MAPKs in vanadate-induced G2/M phase arrest // Free Rad. Biol. Med. – 2003. – Vol. 34. – P. 1333–1342.

Zick Y., Sager-Eisenberg R. A combination of H2O2 and vanadate concomitantly stimulates protein tyrosine phosphorylation and polyphos- phoinositide breakdown in different cell lines // Biochemistry. – 1990. – Vol. 29. – P. 10240–10245.




DOI: http://dx.doi.org/10.24855/biosfera.v5i1.314

© ФОНД НАУЧНЫХ ИССЛЕДОВАНИЙ "XXI ВЕК"