Iron and copper in male reproduction: a double-edged sword
详细信息    查看全文
  • 作者:Eva Tvrda (1) (2)
    Rohan Peer (1)
    Suresh C. Sikka (3)
    Ashok Agarwal (1)

    1. Center for Reproductive Medicine
    ; Cleveland Clinic ; Cleveland ; OH ; USA
    2. Department of Animal Physiology
    ; Slovak University of Agriculture ; Nitra ; Slovakia
    3. Department of Urology
    ; Tulane University School of Medicine ; New Orleans ; LA ; USA
  • 关键词:Iron ; Copper ; Male fertility ; Spermatozoa ; Oxidative stress ; ROS
  • 刊名:Journal of Assisted Reproduction and Genetics
  • 出版年:2015
  • 出版时间:January 2015
  • 年:2015
  • 卷:32
  • 期:1
  • 页码:3-16
  • 全文大小:3,531 KB
  • 参考文献:1. Paoli M, Marles-Wright J, Smith A. Structure-function relationships in heme-proteins. DNA Cell Biol. 2002;21(4):271鈥?0.
    2. Zhou SJ, Schilling MJ, Makrides M. Evaluation of an iron specific checklist for the assessment of dietary iron intake in pregnant and postpartum women. Nutrition. 2005;21:908鈥?3.
    3. Lahti-Koski M, Valsta LM, Alfthan G, Tapanainen H, Aro A. Iron status of adults in the capital area of Finland. Eur J Nutr. 2003;42:287鈥?2.
    4. Reimann C, de Caritat P. Chemical Elements in the Environment. 1st ed. Berlin: Springer Verlag; 1998. p. 124鈥?.
    5. Reimann C, Siewers U, Tarvainen T, Bityukova L, Eriksson J, Gilucis A, Gregorauskiene V, Lukashev V, Matinian N, Pasieczna A. / Agricultural Soils in Northern Europe: A Geochemical Atlas. Schweizerbartsche Verlagsbuchhandlung, Stuttgart; 2003. 279 p.
    6. Mackenzie EL, Iwasaki K, Tsuji Y. Intracellular iron transport and storage: from molecular mechanisms to health implications. Antiox Redox Signal. 2008;10:997鈥?030.
    7. Crichton RR, Charloteaux-Wauters M. Iron transport and storage. Eur J Biochem. 1987;164:485鈥?06.
    8. Wise T, Lunstra DD, Rohrer GA, Ford JJ. Relationships of testicular iron and ferritin concentrations with testicular weight and sperm production in boars. J Anim Sci. 2003;81:503鈥?1.
    9. Toebosch AM, Kroos MJ, Grootegoed A. Transport of transferrin-bound iron into rat Sertoli cells and spermatids. Int J Androl. 1987;10:753鈥?4.
    10. Aitken RJ, Harkiss D, Buckingham D. Relationship between iron-catalysed lipid peroxidation potential and human sperm function. J Reprod Fertil. 1993;98:257鈥?5.
    11. Lieu PT, Heiskala M, Peterson PA, Yang Y. The roles of iron in health and disease. Mol Asp Med. 2001;22:1鈥?7.
    12. Nemeth E, Tuttle MS, Powelson J, Vaughn MB, Donovan A, Ward DM, et al. Hepcidin regulates cellular iron efflux by binding to ferroportin and inducing its internalization. Science. 2004;306:2090鈥?.
    13. Morris MC, Evans DA, Tangney CC, Bienias JL, Schneider JA, Wilson RS, et al. Dietary copper and high saturated and trans fat intakes associated with cognitive decline. Arch Neurol. 2006;63:1085鈥?.
    14. Bertram M, Graedel TE, Rechberger H, Spatari S. The contemporary European copper cycle: waste management subsystem. Ecol Econ. 2002;42:3鈥?7.
    15. Arredondo M, Nunez MT. Iron and copper metabolism. Mol Asp Med. 2005;26:313鈥?7.
    16. L酶vstad RA. Copper catalyzed oxidation of ascorbate (vitamin C). Inhibitory effect of catalase, superoxide dismutase, serum proteins (ceruloplasmin, albumin, apotransferrin) and amino acids. Int J Biochem. 1987;19(4):309鈥?3.
    17. Kachur AV, Koch CJ, Biaglow JE. Mechanism of copper-catalyzed oxidation of glutathione. Free Radic Res. 1998;28(3):259鈥?9.
    18. Hellman NE, Gitlin JD. Ceruloplasmin metabolism and function. Annu Rev Nutr. 2002;22:439鈥?8.
    19. Orlando C, Caldini AL, Barni T, Wood WG, Strasburger CJ, Natali A, et al. Ceruloplasmin and transferrin in human seminal plasma: are they an index of seminiferous tubular function? Fertil Steril. 1985;43:290鈥?.
    20. Aldred AR, Grimes A, Schreiber G, Mercer JF. Rat ceruloplasmin. Molecular cloning and gene expression in liver, choroid plexus, yolk sac, placenta, and testis. J Biol Chem. 1987;262:2875鈥?.
    21. Krezel A, Maret W. Dual nanomolar and picomolar Zn(II) binding properties of metallothionein. J Am Chem Soc. 2007;129(35):10911鈥?1.
    22. Mukhopadhyay D, Mitra A, Nandi P, Varghese AC, Murmu N, Chowdhury R, et al. Expression of metallothionein-1 (MT-1) mRNA in the rat testes and liver after cadmium injection. Syst Biol Reprod Med. 2009;55:188鈥?2.
    23. Ren XY, Zhou Y, Zhang JP, Feng WH, Jiao BH. Expression of metallothionein gene at different time in testicular interstitial cells and liver of rats treated with cadmium. World J Gastroenterol. 2003;9:1554鈥?.
    24. Betka M, Callard GV. Stage-dependent accumulation of cadmium and induction of metallothionein-like binding activity in the testis of the Dogfish shark Squalus acanthias. Biol Reprod. 1999;60:14鈥?2.
    25. Sugihara T, Wadhwa R, Kaul SC, Mitsui Y. A novel testis-specific metallothionein-like protein, tesmin, is an early marker of male germ cell differentiation. Genomics. 1999;57:130鈥?.
    26. Fox PL. The copper-iron chronicles: The story of an intimate relationship. BioMetals. 2003;16:9鈥?0.
    27. Roeser HP, Lee GR, Nacht S, Cartwright GE. The role of ceruloplasmin in iron metabolism. J Clin Invest. 1970;49:2408鈥?7.
    28. Ranganathan PN, Lu Y, Jiang L, Kim C, Collins JF. Serum ceruloplasmin protein expression and activity increases in iron-deficient rats and is further enhanced by higher dietary copper intake. Blood. 2011;118(11):3146鈥?3.
    29. Halliwell B. Biochemistry of oxidative stress. Biochem Soc Trans. 2007;35:1147鈥?0.
    30. Agarwal A, Sekhon LH. The role of antioxidant therapy in the treatment of male infertility. Hum Fertil. 2010;13:217鈥?5.
    31. Peeker R, Abramsson L, Marklund SL. Superoxide dismutase isoenzymes in human seminal plasma and spermatozoa. Mol Hum Reprod. 1997;13:1061鈥?.
    32. Beutler E, Blaisdell RK. Iron enzymes in iron deficiency II. Catalase in human erythrocytes. J Clin Invest. 1958;37(6):833鈥?.
    33. Maneesh M, Jayalekshmi H. Role of reactive oxygen species and antioxidants on pathophysiology of male reproduction. Ind J Clin Chem. 2006;21:80鈥?0.
    34. Letelier ME, Sanchez-Jofre S, Peredo-Silva L, Cortes-Troncoso J, Aracena-Parks P. Mechanisms underlying iron and copper ions toxicity in biological systems: Pro-oxidant activity and protein-binding effects. Chem Biol Interact. 2010;188:220鈥?.
    35. Thomas C, Mackey MM, Diaz AA, Cox DP. Hydroxyl radical is produced via the Fenton reaction in submitochondrial particles under oxidative stress: implications for diseases associated with iron accumulation. Redox Rep. 2009;14:102鈥?.
    36. Kehrer JP. The Haber-Weiss reaction and mechanisms of toxicity. Toxicology. 2000;149:43鈥?0.
    37. Crane1 FL, L枚w H. The oxidative function of diferric transferring. Biochem Res Internat.2012;2012,1鈥?.
    38. Orino K, Lehman L, Tsuji Y, Ayaki H, Torti SV, Torti FM. Ferritin and the response to oxidative stress. Biochem J. 2001;357(Pt 1):241鈥?.
    39. Carlsen CU, M酶ller JKS, Skibsted LF. Heme-iron in lipid oxidation. Coord Chem Rev. 2005;249(3鈥?):485鈥?8.
    40. Rotem R, Paz GF, Homonnai ZT, Kalina M, Naor Z. Further studies on the involvement of protein kinase C in human sperm flagellar motility. Endocrinology. 1990;127:2571鈥?.
    41. Kalina M, Socher R, Rotem R, Naor Z. Ultrastructural localization of protein kinase C in human sperm. J Histochem Cytochem. 1995;43:439鈥?5.
    42. Upadhyaya M, Hibbard BM, Walker SM. Seminal acid phosphatase in relation to fertility. Acta Obstet Gynecol Scand. 1986;65:49鈥?2.
    43. Taira A, Merrick G, Wallner K, Dattoli M. Reviving the acid phosphatase test for prostate cancer. Phys Pract. 2013;1鈥?0.
    44. Tong WH, Rouault TA. Metabolic regulation of citrate and iron by aconitases: role of iron-sulfur cluster biogenesis. Biometals. 2007;20(3鈥?):549鈥?4.
    45. Stamler JS. Redox signaling: nitrosylation and related target interactions of nitric oxide. Cell. 1994;78(6):931鈥?.
    46. Loganathasamy K. Nitric oxide: a double edged weapon for sperm functions. Vet Sci Technol. 2012;3:6.
    47. Hales KG. Iron testes: sperm mitochondria as a context for dissecting iron metabolism. BMC Biol. 2010;8:79.
    48. Metzendorf C, Lind MI. Drosophila mitoferrin is essential for male fertility: evidence for a role of mitochondrial iron metabolism during spermatogenesis. BMC Dev Biol. 2010;10:68.
    49. Paradkar PN, Zumbrennen KB, Paw BH, Ward DM, Kaplan J. Regulation of mitochondrial iron import through differential turnover of mitoferrin 1 and mitoferrin 2. Mol Cell Biol. 2009;29:1007鈥?6.
    50. Zhang Y, Lyver ER, Knight SA, Pain D, Lesuisse E, Dancis A. Mrs3p, Mrs4p, and frataxin provide iron for Fe-S cluster synthesis in mitochondria. J Biol Chem. 2006;281:22493鈥?02.
    51. Nikolaev AA, Lutski沫 DL, Nikolaeva NN, Lozhkina LV. Iron and nonheme iron protein metabolism in ejaculates with varying degrees of fertility. Urol Nefrol. 1998;5:27鈥?1.
    52. Kanwal MR, Rehman NU, Ahmad N, Samad HA, Uh-Rehman Z, Akhtarand SA. Bulk cations and trace lements in the Nili-Ravi buffalo and crossbred cow bull semen. Int J Agric Biol. 2000;2:302鈥?.
    53. Tvrda E, Knazicka Z, Lukacova J, Schneidgenova M, Massanyi P, Goc Z, et al. Relationships between iron and copper content, motility characteristics, and antioxidant status in bovine seminal plasma. JMBFS. 2012;2:536鈥?7.
    54. Knazicka Z, Lukacova J, Tvrda E, Gren A, Goc Z, Massanyi P, et al. / In vitro assessment of iron effect on the spermatozoa motility parameters. JMBFS. 2012;2:414鈥?5.
    55. Uriu-Adams JY, Keen CL. Copper, oxidative stress, and human health. Mol Asp Med. 2005;6:268鈥?8.
    56. Denis M. Structure and function of cytochrome-c oxidase. Biochimie. 1986;68(3):459鈥?0.
    57. Yu MA, Egawa T, Shizawa-Itoh, Yoshikawa S, Yeh SR, Rousseau DL, et al. Radical formation in cytochrome c oxidase. Biochim Biophys Acta. 1807;2011:1295鈥?04.
    58. H眉ttemann M, Jaradat S, Grossman LI. Cytochrome c oxidase of mammals contains a testes-specific isoform of subunit VIb鈥搕he counterpart to testes-specific cytochrome c? Mol Reprod Dev. 2003;66:8鈥?6.
    59. Krzyzosiak J, McMillan G, Molan P, Vishwanath R. Protein tyrosine phosphorylation during prolonged / in vitro incubation of ejaculated bovine spermatozoa is regulated by the oxidative state of the medium. Biol Reprod. 2000;62:1615鈥?3.
    60. Le Calv茅 M, Segalen J, Quernee D, Lavault MT, Lescoat D. Diamine oxidase activity and biochemical markers in human seminal plasma. Hum Reprod. 1995;10:1141鈥?.
    61. Wolverkamp MCJ, Debriun RWF. Diamine oxidase 鈥?an overview of historical, biochemical and functional aspects. Dig Dis. 1994;12:2鈥?4.
    62. Saleh BOM, Hussain NK, Majid AY, Thabet B, Fadhil KA. Status of zinc and copper concentrations in seminal plasma of male infertility and their correlations with various sperm parameters. Iraq Postgrad Med J. 2008;7:76鈥?0.
    63. Akinloye O, Abbiyesukru FM, Oguntibeju OO, Arowojolu AO, Truter EJ. The impact of blood and seminal plasma zinc and copper concentrations on spermogram and hormonal changes in infertile Nigerian men. Reprod Biol. 2011;11:83鈥?7.
    64. Abdul-Rasheed OF. Association between seminal plasma copper and magnesium levels with oxidative stress in Iraqi infertile men. OMJ. 2010;25:168鈥?2.
    65. Wong WY, Flik G, Groenen PM, Swinkels DW, Thomas CM, Copius-Peereboom JH, et al. The impact of calcium, magnesium, zinc, and copper in blood and seminal plasma on semen parameters in men. Reprod Toxicol. 2001;15:131鈥?.
    66. Machal L, Chladek G, Strakova E. Copper, phosphorus and calcium in bovine blood and seminal plasma in relation to semen quality. J Anim Feed Sci. 2002;11:425鈥?5.
    67. Tabasomi M, Alavi-Shoushtari SM. Effects of / in vitro copper sulphate supplementation on the ejaculated sperm characteristics in water buffaloes ( / Bubalus bubalis). Vet Res Forum. 2013;4:31鈥?.
    68. Celino FT, Yamaguchi S, Miura C. Tolerance of spermatogonia to oxidative stress is due to high levels of Zn and Cu/Zn superoxide dismutase. PLoS One. 2011;6:1鈥?1.
    69. Kawakami E, Takemura A, Sakuma M, Takano M, Hirano T, Hori T, et al. Superoxide dismutase and catalase activities in the seminal plasma of normozoospermic and asthenozoospermic Beagles. J Vet Med Sci. 2007;69:133鈥?.
    70. Zini A, Fischer MA, Mak V, Phang D, Jarvi K. Catalase-like and superoxide dismutase-like activities in human seminal plasma. Urol Res. 2002;30:321鈥?.
    71. Marzec-Wr贸blewska U, Kami艅ski P, Lakota P, Szyma艅ski M, Wasilow K, Ludwikowski G, et al. Zinc and iron concentration and SOD activity in human semen and seminal plasma. Biol Trace Elem Res. 2011;143:167鈥?7.
    72. Comhaire FH, Mahmoud AMA, Depuydt CE, Zalata AA, Christophe AB. Mechanisms and effects of male genital tract infection on sperm quality and fertilizing potential: the andrologist's viewpoint. Hum Reprod Update. 1999;5(5):393鈥?.
    73. Eghbali M, Alavi-Shoushtari SM, Rezaii SA. Effects of copper and superoxide dismutase content of seminal plasma on buffalo semen characteristics. Pak J Biol Sci. 2008;11:1964鈥?.
    74. Khosrowbeygi A, Zarghami N, Deldar Y. Correlation between sperm quality parameters and seminal plasma antioxidants status. Iran J Reprod Med. 2004;2:58鈥?4.
    75. Murawski M, Saczko J, Marcinkowska A, Chwi艂kowska A, Grybo艣 M, Bana艣 T. Evaluation of superoxide dismutase activity and its impact on semen quality parameters of infertile men. Folia Histochem Cytobiol. 2007;45:123鈥?.
    76. Abd-Elmoaty MA, Saleh R, Sharma R, Agarwal A. Increased levels of oxidants and reduced antioxidants in semen of infertile men with varicocele. Fertil Steril. 2010;94:1531鈥?.
    77. Malkoc E, Tunckiran MA, Uguz S, Kocoglu S, Ates F, Muftuoglu T, et al. Evaluation of both malondialdehyde and catalase enzymes in semen, tissue and blood in adult men with grade 3 varicocele. Dis Molec Med. 2013;1:26鈥?0.
    78. Rossi T, Mazzilli F, Delfino M, Dondero F. Improved human sperm recovery using superoxide dismutase and catalase supplementation in semen cryopreservation procedure. Cell Tissue Bank. 2001;2:9鈥?3.
    79. Chaivechakarn A, Thuwnaut P, Ponglowhapan S, Chatdarong K. Effects of cold storage prior to freezing on superoxide dismutase and glutathione peroxidase activities, level of total reactive oxygen species and sperm quality in dogs. In: England G, Kutzler M, Comizzoli P, Nizanski W, Rijsselaere T, Concannon P, editors. Proceeding from the 7th International Symposium on Canine and Feline Reproduction: 26鈥?9 July 2012. Whistler: ISCFR; 2012. p. 1鈥?.
    80. Cocchia N, Pasolini MP, Mancini R, Petrazzuolo O, Cristofaro I, Rosapane I, et al. Effect of SOD (superoxide dismutase) protein supplementation in semen extenders on motility, viability, acrosome status and ERK (extracellular signal-regulated kinase) protein phosphorylation of chilled stallion spermatozoa. Theriogenology. 2011;75:1201鈥?0.
    81. Tvrda E, Lukac N, Schneidgenova M, Lukacova J, Szabo CS, Goc Z, et al. Impact of seminal chemical elements on the oxidative balance in bovine seminal plasma and spermatozoa. J Vet Med. 2013;2013:1鈥?.
    82. Tsunoda S, Kawano N, Kawano K, Kimura N, Fujii J. Impaired fertilitzing ability of superoxide dismutase 1-defficient mouse sperm during / in vitro fertilization. Biol Reprod. 2012;87:1鈥?.
    83. Garratt M, Bathgate R, de Graaf SP, Brooks RC. Copper-zinc superoxide dismutase deficiency impairs sperm motility and / in vivo fertility. Reproduction. 2013;146:297鈥?04.
    84. Evans JL, Abraham PA. Anemia, iron storage and ceruloplamsin in copper nutrition in the growing rat. J Nutr. 1973;103:196鈥?01.
    85. Boral MC, Kaul P, Dey SK, Deb C. Effect of experimentally induced anemia on the testicular activity of the toad ( / Bufo melanostictus). J Exp Zool. 1974;188(1):77鈥?7.
    86. Yassin MA, Soliman AT, Desanctis V. Anemia (IDA): Effects on pituitary gonadal axis and sperm parameters. Blood. 2013;122:967.
    87. Alleyne M, Horne MK, Miller JL. Individualized treatment for iron-deficiency anemia in adults. Am J Med. 2008;121:943鈥?.
    88. Davies S, Henthorn J, Brozovic M. Iron deficiency in sickle cell anaemia. J Clin Pathol. 1983;36:1012鈥?.
    89. Agbaraji VO, Scott RB, Leto S, Kingslow LW. Fertility studies in sickle cell disease: semen analysis in adult male patients. Int J Fertil. 1988;33:347鈥?2.
    90. Gerald F, Ruth F. Testicular function in sickle cell disease. Fertil Steril. 1974;25:243鈥?.
    91. Abbasi AA, Prasad AO, Ortega J, Conego E, Oberleas D. Gonadal function abnormalities in Sickle cell anaemia Studies in adult male patients. Ann Intern Med. 1976;85:601鈥?.
    92. Olatunji O, Frasier SD. Sexual Maturation in subject with sickle cell anaemia: Studies of serum gonadotropin concentration, height, weight and skeletal age. J Pediatr. 1975;87:459鈥?4.
    93. Abdulwaheed OO, Abdulrasaq AA, Sulaiman AK, Abdulgafar AJ, Munirdeen AI. The hormonal assessment of the infertile male in Ilorin Nigeria. Afri J Endocrinol Metab. 2002;3:62鈥?.
    94. Abudu EK, Akanmu SA, Soriyan OO, Akinbami AA, Adediran A, Adezemo TA, et al. Serum testosterone levels of HbSS (sickle cell disease) male subjects in Lagos, Nigeria. BMC Res Notes. 2011;4:298鈥?02.
    95. Dada OO, Nduka EU. Endocrine function and haemoglobinopathies: Relation between the sickle cell gene and circulatory levels of testosterone, LH and FSH in adult males. Clin Chim Acta. 1980;105:269鈥?3.
    96. Berthaut I, Guignedoux G, Kirsch-Noir F, De Larouziere V, Ravel C, Bachir D, et al. Influence of sickle cell disease and treatment with hydroxyurea on sperm parameters and fertility of human males. Haematologica. 2008;93:988鈥?3.
    97. Grigg A. Effect of hydroxyurea on sperm count, motility and morphology in adult men with sickle cell or myeloproliferative disease. Intern Med J. 2007;37:190鈥?.
    98. Chatterjee R, Katz M, Cox TF, Porter JB, Bantock H. Evaluation of GH in thalassaemic boys with failed puberty: spontaneous versus provocative test. Eur J Pediatr. 1993;152:721鈥?.
    99. Chatterjee R, Katz M, Cox TF, Porter JB. Prospective study of the hypothalamic鈥損ituitary axis in thalassaemic patients who developed secondary amenorrhoea. Clin Endocrinol. 1993;39:287鈥?4.
    100. Chatterjee R, Katz M. Reversible hypogonadotrophic hypogonadism in sexually infantile male thalassaemic patients with transfusional iron overload. Clin Endocrinol. 2000;53:33鈥?2.
    101. Chatterjee R, Katz M, Oatridge A, Bydder GM, Porter JB. Selective loss of anterior pituitary volume with severe pituitary鈥揼onadal insufficiency in poorly compliant male thalassaemic patients with pubertal arrest. Ann N Y Acad Sci. 1998;850:482鈥?.
    102. De Sanctis V, Pintor C, Gamberini MR, Ughi M, Pinamonti A, Aliquo MC, et al. Multicentric study of endocrine complications in thalassaemia major. Clin Endocrinol. 1995;42:581鈥?.
    103. Merchant RH, Shirodkar A, Ahmed J. Evaluation of growth, puberty and endocrine dysfunctions in relation to iron overload in multi transfused Indian thalassemia patients. Indian J Pediatr. 2011;78:679鈥?3.
    104. Noetzli LJ, Panigray A, Mittleman SD, Hyderi A, Dongelyan A, Coates TD, et al. Pituitary iron and volume predict hypogonadism in transfusional iron overload. Am J Hematol. 2007;87:167鈥?1.
    105. Cisternino M, Manzoni SM, Coslovich E, Autelli M. Hormonal replacement therapy with HCG and HU-FSH in thalassaemic patients affected by hypogonadotropic hypogonadism. J Pediatr Endocrinol Metab. 1998;11 Suppl 3:885鈥?0.
    106. Perera D, Pizzey A, Campbell A, Katz M, Porter J, Petrou M, et al. Sperm DNA damage in potentially fertile homozygous beta-thalassaemia patients with iron overload. Hum Reprod. 2002;17:1820鈥?.
    107. Van Niekerk FE, Van Niekerk CH. The influence of experimentally induced copper deficiency on the fertility of rams I. Semen parameters and peripheral plasma androgen concentration. J S Afr Vet Assoc. 1989;60:28鈥?1.
    108. Van Niekerk FE, Van Niekerk CH. The influence of experimentally induced copper deficiency on the fertility of rams II. Macro- and microscopic changes in the testes. J S Afr Vet Assoc. 1989;60:32鈥?.
    109. Lyubimov AV, Smith JA, Rousselle SD, Mercieca MD, Tomaszewski JE, Smith AC, et al. The effects of tetrathiomolybdate (TTM, NSC-714598) and copper supplementation on fertility and early embryonic development in rats. Reprod Toxicol. 2004;19:223鈥?3.
    110. Aupperle H, Schoon HA, Frank A. Experimental copper deficiency, chromium deficiency and additional molybdenum supplementation in goats-pathological findings. Acta Vet Scand. 2001;42:311鈥?1.
    111. Lee J, Petris MJ, Thiele DJ. Characterization of mouse embryonic cells deficient in the ctr1 high affinity copper transporter. Identification of a Ctr1-independent copper transport system. J Biol Chem. 2002;277(43):40253鈥?.
    112. Suzuki KT, Someya A, Komada Y, Ogra Y. Roles of metallothionein in copper homeostasis: responses to Cu-deficient diets in mice. J Inorg Biochem. 2002;88(2):173鈥?2.
    113. Jenkinson SG, Lawrence RA, Burk RF, Williams DM. Efects of copper deficiency on the activity of the selenoenzyme glutathione peroxidase and on excretion and tissue retention of 75SeO3 2鈥?,2. J Nutr. 1982;112:197鈥?04.
    114. Picco SJ, De Luca JC, Mattioli G, Dulout FN. DNA damage induced by copper deficiency in cattle assessed by the Comet assay. Mutat Res. 2001;498(1鈥?):1鈥?.
    115. Narisawa S, Hecht NB, Goldberg E, Boatright KM, Reed JC, Millan JL. Testis-specific cytochrome c-null mice produce functional sperm but undergo early testicular atrophy. Mol Cell Biol. 2002;22:5554鈥?2.
    116. Wang J, Pantopoulos K. Regulation of cellular iron metabolism. Biochem J. 2011;434:365鈥?1.
    117. Merker HJ, Baumgartner W, Kovac G, Bartko P, Rosival I, Zezula I. Iron-induced injury of rat testis. Andrologia. 1996;28:267鈥?3.
    118. Lucesoli F, Fraga CG. Oxidative damage to lipids and DNA concurrent with decrease of antioxidants in rat testes after acute iron intoxication. Arch Biochem Biophys. 1995;316:567鈥?1.
    119. De Lourdes MP, E Garcia FC. Spermatogenesis recovery in the mouse after iron injury. Hum Exp Toxicol. 2003;22(5):275鈥?.
    120. Buretic-Tomaljanovic A, Vlastelic I, Radojcic-Badovinac A, Starcevic-Cizmarevic N, Nadalin S, Ristic S. The impact of hemochromatosis mutations and transferrin genotype on gonadotropin serum levels in infertile men. Fertil Steril. 2009;91:1793鈥?00.
    121. Gottschalk R, Seidl C, Schilling S, Braner A, Seifried E, Hoelzer D, et al. Iron-overload and genotypic expression of HFE mutations H63D/C282Y and transferrin receptor Hin6I and BanI polymorphism in german patients with hereditary haemochromatosis. Eur J Immunogenet. 2000;27(3):129鈥?4.
    122. Gunel-Ozcan A, Basar MM, Kisa U, Ankarali HC. Hereditary haemochromatosis gene (HFE) H63D mutation shows an association with abnormal sperm motility. Mol Biol Rep. 2009;36:1709鈥?4.
    123. Anderson D, Schmid TE, Baumgartner A. Male-mediated developmental toxicity. Asian J Androl. 2014;16(1):81鈥?.
    124. Uitz PM, Hartleb S, Schaefer S, Al-Fakhri N, Kann PH. Pituitary function in patients with hereditary haemochromatosis. Horm Metab Res. 2013;45:54鈥?1.
    125. Eidi M, Eidi A, Pouyan O, Shahmohammadi P, Fazaeli R, Bahar M. Seminal plasma levels of copper and its relationship with seminal parameters. Iran J Reprod Med. 2010;8:6.
    126. Schmid TE, Grant PG, Marchetti F, Weldon RH, Eskenazi B, Wyrobek AJ. Elemental composition of human semen is associated with motility and genomic sperm defects among older men. Hum Reprod. 2013;28:274鈥?2.
    127. Babaei H, Kheirandish R, Ebrahimi L. The effects of copper toxicity on histopathological and morphometrical changes in the rat testes. Asian Pacif J Tropic Biomed. 2012;2012:615鈥?.
    128. Knazicka Z, Tvrda E, Bardos L, Lukac N. Dose- and time-dependent effect of copper ions on the viability of bull spermatozoa in different media. J Environ Sci Health. 2012;47:1294鈥?00.
    129. Roychoudhury S, Massanyi P, Bulla J, Choudhury MD, Straka L, Lukac N, et al. / In vitro copper toxicity on rabbit spermatozoa motility, morphology and cell membrane integrity. J Environ Sci Health A Tox Hazard Subst Environ Eng. 2010;A45:1482鈥?1.
    130. Rebrelo L, Guadarrama A, Lopez T, Zegers HF. Effect of Cu ion on the motility, viability, acrosome reaction and fertilizing capacity of human spermatozoa in vitro. Reprod Fertil Dev. 1996;8:871鈥?.
    131. Tarnacka B, Rodo M, Cichy S, Czlonkowska A. Procreation ability in Wilson's disease. Acta Neurol Scand. 2000;101:395鈥?.
    132. Tvrd谩 E, K艌a啪ick谩 Z, B谩rdos L, Mass谩nyi P, Luk谩膷 N. Impact of oxidative stress on male fertility - a review. Acta Vet Hung. 2011;59:465鈥?4.
    133. Huang YL, Tseng WC, Lin TH. / In vitro effects of metal ions (Fe2+, Mn2+, Pb2+) on sperm motility and lipid peroxidation in human semen. J Toxicol Environ Health A. 2001;62:259鈥?7.
    134. Lucesoli F, Caligiuri M, Roberti MF, Perazzo JC, Fraga CG. Dose-dependent increase of oxidative damage in the testes of rats subjected to acute iron overload. Arch Biochem Biophys. 1999;372:37鈥?3.
    135. Wellejus A, Poulsen HE, Loft S. Iron-induced oxidative DNA damage in rat sperm cells / in vivo and / in vitro. Free Radic Res. 2000;32:75鈥?3.
    136. Fraga CG, Oteiza PI. Iron toxicity and antioxidant nutrients. Toxicology. 2002;180(1):23鈥?2.
    137. Verma A, Kanwar KC. Human sperm motility and lipid proxidation in different ascorbic acid concentrations: an / in vitro analysis. Andrologia. 1998;30:325鈥?.
    138. Mojica-Villegas MA, Izuierdo-Vega JA, Chamorro-Cevallos G, Sanchez-Guiterrez M. Protective effects of resveratrol on biomarkers of oxidative stress induced by iron/ascorbate in mouse spermatozoa. Nutrients. 2014;6:489鈥?03.
    139. Murugan MA, Gangadharan B, Mathur PP. Antioxidative effect of fullerenol on goat epididymal spermatozoa. Asian J Androl. 2002;4:149鈥?2.
    140. Bansal AK, Bilaspuri GS. Effect of manganese on bovine sperm motility, viability, and lipid peroxidation / in vitro. Anim Reprod. 2008;5:90鈥?.
    141. Ball BA, Vo A. Detection of lipid peroxidation in equine spermatozoa based upon the lipophilic fluorescent dye C11-BODIPY581/591. J Androl. 2002;23:259鈥?9.
    142. Olivari FA, Hern谩ndez PP, Allende ML. Acute copper exposure induces oxidative stress and cell death in lateral line hair cells of zebrafish larvae. Brain Res. 2008;1244:1鈥?2.
    143. Vlarengo A, Pertica M, Mancinelli G, Zanicchi G, Orunesu M. Rapid induction of copper-binding proteins in the gills of metal exposed mussels. Comp Biochem Physiol. 1980;67:215鈥?.
    144. Wimalasena DS, Wiese TJ, Wimalasena K. Copper ions disrupt dopamine metabolism via inhibition of V-H鈥?鈥夆垝ATPase: a possible contributing factor to neurotoxicity. J Neurochem. 2007;101:313鈥?6.
    145. Earnshaw MJ, Wilson S, Akberali HB, Butler RD, Marriott KRM. The action of heavy metals on the gametes of the marine mussel, Mytilus edulis (L.) 鈥?III. The effect of applied copper and zinc on sperm motility in relation to ultrastructural damage and intracellular metal localization. Mar Environ Res. 1986;20:261鈥?8.
    146. Krumschnabel G, Manzl C, Berger C, Hofer B. Oxidative stress, mitochondrial permeability transition, and cell death in Cu-exposed trout hepatocytes. Toxicol Appl Pharmacol. 2005;209:62鈥?3.
    147. Brittenham GM, Griffith PM, Nienhuis AW. Efficacy of deferoxamine in preventing complications of iron overload in patients with thalassemia major. N Engl J Med. 1994;331:567鈥?3.
    148. Wali YA, Taqi A, Deghaidi A. Study of intermittent intravenous deferrioxamine high-dose therapy in heavily iron-loaded children with beta-thalassemia major poorly compliant to subcutaneous inject ions. Pediatr Hematol Oncol. 2004;21:453鈥?0.
    149. Tripathi N, Kannan GM, Pant BP, Jaiswal DK, Malhotra PR, Flora SJS. Arsenic induced changes in certain neurotransmitters levels and their recoveries following chelation in rat whole brain. Toxicol Lett. 1997;92:201鈥?.
    150. Pande M, Mehta A, Pant BP, Flora SJS. Combined administration of a chelating agent and an antioxidant in the prevention and treatment of acute lead intoxication in rats. Environ Toxicol Pharmacol. 2001;9:173鈥?4.
    151. Poggiali E, Cassinerio E, Zanaboni L, Cappellini MD. An update on iron chelation therapy. Blood Transf. 2012;10:411鈥?2.
    152. Kalinowski DS, Richardson DR. The evolution of iron chelators for the treatment of iron overload disease and cancer. Pharmacol Rev. 2005;57:547鈥?3.
    153. Kontoghiorghes GJ, Spyrou A, Kolnagou A. Iron chelation therapy in hereditary hemochromatosis and thalassemia intermedia: regulatory and non regulatory mechanisms of increased iron absorption. Hemoglobin. 2010;34(3):251鈥?4.
    154. Soliman A, Yassin M, De Sanctis V. Intravenous iron replacement therapy in eugonadal males with iron-deficiency anemia: Effects on pituitary gonadal axis and sperm parameters; A pilot study. Indian J Endocrinol Metab. 2014;18(3):310鈥?.
    155. 艠铆ha M, Karl铆膷kov谩 J, Filipsk媒 T, Mac谩kov谩 K, Hrdina R, Mlad臎nka P. Novel method for rapid copper chelation assessment confirmed low affinity of D-penicillamine for copper in comparison with trientine and 8-hydroxyquinolines. J Inorg Biochem. 2013;123:80鈥?.
    156. Rawy SM, Al Nassr S. Zinc sulphate and vitamin E alleviate reproductive toxicity caused by aluminium sulphate in male albino rats. Toxicol Ind Health. 2012;2012:1鈥?4.
    157. Sripetchwandee J, Pipatpiboon N, Chattipakorn N, Chattipakorn S. Combined therapy of iron chelator and antioxidant completely restores brain dysfunction induced by iron toxicity. PLoS One. 2014;9(1):e85115.
    158. Khanna AK, Xu J, Mehra MR. Antioxidant N-acetyl cysteine reverses cigarette smoke-induced myocardial infarction by inhibiting inflammation and oxidative stress in a rat model. Lab Invest. 2011;92:224鈥?5.
    159. Zhu Y, Zhang XL, Zhu BF, Ding YN. Effect of antioxidant N-acetylcysteine on diabetic retinopathy and expression of VEGF and ICAM-1 from retinal blood vessels of diabetic rats. Mol Biol Rep. 2012;39:3727鈥?5.
    160. Bansal AK, Bilaspuri GS. Antioxidant effect of vitamin E on motility, viability and lipid peroxidation of cattle spermatozoa under oxidative stress. Anim Sci Paper Rep. 2009;27(1):5鈥?4.
    161. Omara FO, Blakley BR. Vitamin E is protective against iron toxicity and iron-induced hepatic vitamin E depletion in mice. J Nutr. 1993;123(10):1649鈥?5.
    162. Campanella L, Gatta T, Ravera O. Relationship between antioxidant capacity and manganese accumulation in the soft tissues of two freshwater molluscs: / Unio pictorum mancus ( / Lamellibranchia, / Unionidae) and / Viviparous ater ( / Gastropoda, / Prosobranchia). J Limnol. 2005;64:153鈥?.
    163. Bansal AK. Manganese: a potent antioxidant in semen. Iran J Appl Anim Sci. 2013;3(2):217鈥?2.
    164. Bansal AK, Kaur AR. Cooperative functions of manganese and thiol redox system against oxidative stress in human spermatozoa. J Hum Reprod Sci. 2009;2(2):76鈥?0.
    165. VanLandingham JW, Fitch CA, Levenson CW. Zinc inhibits the nuclear translocation of the tumor suppressor protein p53 and protects cultured human neurons from copper-induced neurotoxicity. NeuroMolecular Med. 2002;1(3):171鈥?2.
    166. Lanno RP, Slinger SJ, Hilton JW. Effect of ascorbic acid on dietary copper toxicity in rainbow trout ( / Salmo gairdneri Richardson). Aquaculture. 1985;49(3鈥?):269鈥?7.
    167. Ounjaijean S, Thephinlap C, Khansuwan U, Phisalapong C, Fucharoen S, Porter JB, et al. Effect of green tea on iron status and oxidative stress in iron-loaded rats. Med Chem. 2008;4(4):365鈥?0.
    168. Khan N, Afaq F, Saleem M, Ahmad N, Mukhtar H. Targeting multiple signaling pathways by green tea polyphenol (鈭?-epigallocatechin-3-gallate. Cancer Res. 2006;66(5):2500鈥?.
    169. Kalpravidh RW, Siritanaratkul N, Insain P, Charoensakdi R, Panichkul N, Hatairaktham S, et al. Improvement in oxidative stress and antioxidant parameters in beta-thalassemia/Hb E patients treated with curcuminoids. Clin Biochem. 2010;43(4鈥?):424鈥?.
    170. Thephinlap C, Phisalaphong C, Fucharoen S, Porter JB, Srichairatanakool S. Efficacy of curcuminoids in alleviation of iron overload and lipid peroxidation in thalassemic mice. Med Chem. 2009;5(5):474鈥?2.
    171. Jiao Y, Wilkinson 4th J, Di X, Wang W, Hatcher H, Kock ND, et al. Curcumin, a cancer chemopreventive and chemotherapeutic agent, is a biologically active iron chelator. Blood. 2009;113(2):462鈥?.
    172. Juan ME, Gonz谩lez-Pons E, Munuera T, Ballester J, Rodr铆guez-Gil JE, Planas JM. Trans-resveratrol, a natural antioxidant from grapes, increases sperm output in healthy rats. J Nutr. 2005;135(4):757鈥?0.
    173. Shin S, Jeon JH, Park D, Jang MJ, Choi JH, Choi BH, et al. Trans-Resveratrol relaxes the corpus cavernosum ex vivo and enhances testosterone levels and sperm quality in vivo. Arch Pharm Res. 2008;31(1):83鈥?.
    174. Revel A, Raanani H, Younglai E, Xu J, Han R, Savouret JF, et al. Resveratrol, a natural aryl hydrocarbon receptor antagonist, protects sperm from DNA damage and apoptosis caused by benzo(a)pyrene. Reprod Toxicol. 2001;15(5):479鈥?6.
    175. Uguralp S, Mizrak B, Bay KA. Resveratrol reduces ischemia reperfusion injury after experimental testicular torsion. Eur J Pediatr Surg. 2005;15(2):114鈥?.
  • 刊物类别:Medicine
  • 刊物主题:Medicine & Public Health
    Gynecology
    Developmental Biology
    Human Genetics
  • 出版者:Springer Netherlands
  • ISSN:1573-7330
文摘
Iron and copper are essential trace nutrients playing important roles in general health and fertility. However, both elements are highly toxic when accumulating in large quantities. Their direct or indirect impact on the structure and function of male gonads and gametes is not completely understood yet. Excess or deficiency of either element may lead to defective spermatogenesis, reduced libido, and oxidative damage to the testicular tissue and spermatozoa, ultimately leading to fertility impairment. This review will detail the complex information currently available on the dual roles iron and copper play in male reproduction.

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700