Next Article in Journal
Metabolic Flux Analysis of Catechin Biosynthesis Pathways Using Nanosensor
Previous Article in Journal
Antioxidants Properties of Natural Products: A Themed Issue in Honor of Professor Isabel C.F.R. Ferreira
Previous Article in Special Issue
Long-Term Adverse Effects of Oxidative Stress on Rat Epididymis and Spermatozoa
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Reactive Oxygen Species and Male Fertility

by
Cristian O’Flaherty
1,2,3
1
Department of Surgery (Urology Division), McGill University, Montréal, QC H4A 3J1, Canada
2
Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3G 1Y6, Canada
3
The Research Institute, McGill University Health Centre, Montréal, QC H4A 3J1, Canada
Antioxidants 2020, 9(4), 287; https://doi.org/10.3390/antiox9040287
Submission received: 17 March 2020 / Accepted: 25 March 2020 / Published: 29 March 2020
(This article belongs to the Special Issue Reactive Oxygen Species and Male Fertility)
Human infertility affects ~15% of couples worldwide, and it is now recognized that in half of these cases, the causes of infertility can be traced to men [1,2]. The spermatozoon is a terminal cell with the unique goal of delivering the paternal genome into the oocyte. This essential task for any species survival can be threatened by environmental pollutants, chemicals, drugs, smoke, toxins, radiation, diseases, and lifestyles. Oxidative stress is a common feature of the mechanism of action of these factors and conditions that negatively impact male fertility [3,4,5,6,7]. Indeed, the reactive oxygen species (ROS)-mediated damage to spermatozoa is a significant contributing factor to infertility in 30-80% of infertile men [8,9,10,11,12].
Spermatozoa are terminally differentiated cells produced in the testes during the hormone-regulated process of spermatogenesis. Two somatic cell types are critical to this process: Sertoli cells protect and support the germ cell development, whereas interstitial Leydig cells produce necessary intratesticular steroids (e.g., testosterone) [13,14]. After their release from the testis, spermatozoa complete their maturation during the transit through the epididymis. There, they acquire the potential for motility and fertility through extensive morphological and biochemical modifications [15]. After ejaculation, spermatozoa must undergo the complex and timely process of capacitation that involves ionic, metabolic, and membrane changes, including the production of ROS at low concentrations [16,17]. Capacitation allows spermatozoa to bind to the zona pellucida that surrounds the oocyte and induce the acrosome reaction [18], an exocytotic event by which proteolytic enzymes (e.g., acrosin and hyaluronidase) are released. Thus, the spermatozoon penetrates the zona pellucida and reaches and fuses with the oocyte. Failure to undergo sperm capacitation and/or acrosome reaction is associated with infertility [19,20].
A key feature of sperm capacitation is the production of ROS at very low and controlled levels by the spermatozoon. This essential phenomenon for the acquisition of the fertility ability was first reported in humans [21], bovine [22], and equine [23] spermatozoa, and then confirmed by others (see [24] for more information). ROS play the role of second messengers and act in most of the known signal transduction pathways involved in this complex phenomenon [25,26,27]. Sperm capacitation is a redox-regulated process. Peroxiredoxins (PRDXs) play a crucial role in maintaining low levels of intracellular ROS to allow the achievement of fertilizing ability by the spermatozoon [28,29].
Oxidative stress can promote detrimental changes during spermatogenesis, epididymal maturation, and sperm capacitation that can lead to infertility [30,31,32,33,34,35]. Lipid peroxidation of the sperm plasma membrane is one of the first described oxidative damage associated with low sperm quality and infertility [36,37]. 4-hydroxynonenal, a subproduct of lipid peroxidation, forms adducts with proteins and DNA, impairing sperm mitochondrial function and promoting mutations in the sperm genome [38]. PRDX6, a unique antioxidant with peroxidase and calcium-independent phospholipase A2, is a key element in the antioxidant defence to protect sperm membranes and DNA from oxidative damage [39,40]. Oxidative damage to the paternal genome has been reported in humans and animals and associated with fertility failure [41,42,43,44]. Redox-dependent protein modifications are related to low sperm quality and infertility [45,46,47]. The reproductive system is equipped with antioxidant enzymes to avoid the adverse effects of high levels of ROS during the production and maturation of spermatozoa. Different studies have addressed the consequences of the absence of antioxidant enzymes on male reproduction (for details see [48]), and from knockout mouse models, we learn the critical need of superoxide dismutase, glutathione peroxidases, thioredoxins, and PRDXs to produce a healthy and fertile spermatozoa in young adult and aging males [32,41,49,50,51,52].
The decline of fertility and the increase of abnormalities in the semen of men that is observed over more than two decades is worrisome. However, we still do not have sufficient information to understand why this is happening [53,54,55]. The exposure environmental toxicants could partly explain this phenomenon, but there is still a significant amount of uncertainty regarding the possible causes of the decrease in sperm quality over the years [53,55]. It is an established trend that men are delaying fatherhood due to professional reasons, and scientific data from studies in animals and humans revealed that sperm quality worsens as men age. There is increasing evidence that children fathered by 50-year-old-or older men are prone to manifest a variety of disorders that can be linked to significant mutations of the paternal genome [56].
This Special Issue on ROS and male fertility is composed of four original contributions that provide new data on the role of SOD [57] and PRDXs [58] as essential antioxidants in the epididymis, and PRDXs as novel players in the protection of gonocytes against oxidative stress [59] and the participation of glutathione-S transferase omega 2 in the regulation of sperm function during capacitation [60]. The Special Issue is completed with six review articles that provide an update on the molecular changes induced by oxidative stress that impairs human sperm motility [61], the importance of ROS in determining the functionality of spermatozoa and situations in which oxidative stress occurs and impacts on male fertility [24], the oxidation of the sperm nucleus and the impact of oxidative stress on the paternal genome [62], the importance of using appropriate tools to study the role of ROS in sperm and infertility [63], and an exhaustive evaluation of ROS production and its relevance in male fertility and antioxidant therapy [64]. The characterization of the redox regulation and effects of oxidative stress in equine spermatozoa is also included in this Special Issue [65].
Many groundbreaking studies have contributed to our understanding of the field of ROS in male fertility. It is undeniable that these reactive molecules play an essential role in both physiological and pathological mechanisms of the male reproductive system. Antioxidant therapy is a common clinical strategy to improve sperm quality and function in infertile men [66,67]. However, there are controversial results regarding the efficacy of these treatments. Some controlled trials suggested that such treatments are beneficial to achieve live births [66], whereas others did not show any benefit [68]. This discrepancy is likely based on the lack of tools in clinics to establish whether oxidative stress is responsible for the infertility of a given patient. Thus, more fundamental and clinical research is needed to comprehend how ROS modulate male fertility to design better diagnostic tools and therapeutic strategies to fight against male infertility.

Funding

Original research by the author of this editorial was supported by the Canadian Institutes of Health Research (CIHR), operating grant # MOP133661 to CO.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Bushnik, T.; Cook, J.L.; Yuzpe, A.; Tough, S.; Collins, J. Estimating the prevalence of infertility in Canada. Hum. Reprod. 2012, 7, 738–746. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. World Health Organization. Towards more objectivity in diagnosis and management of male fertility. Int. J. Androl. 1997, 7, 1–53. [Google Scholar]
  3. Anderson, J.B.; Williamson, R.C. Testicular torsion in Bristol: A 25-year review. Br.J. Surg. 1988, 75, 988–992. [Google Scholar] [CrossRef] [PubMed]
  4. Hasegawa, M.; Wilson, G.; Russell, L.D.; Meistrich, M.L. Radiation-induced cell death in the mouse testis: Relationship to apoptosis. Radiat. Res. 1997, 147, 457–467. [Google Scholar] [CrossRef] [PubMed]
  5. Brennemann, W.; Stoffel-Wagner, B.; Helmers, A.; Mezger, J.; Jager, N.; Klingmuller, D. Gonadal function of patients treated with cisplatin based chemotherapy for germ cell cancer. J. Urol. 1997, 158 Pt 1, 844–850. [Google Scholar] [CrossRef]
  6. Smith, R.; Kaune, H.; Parodi, D.; Madariaga, M.; Rios, R.; Morales, I.; Castro, A. Increased sperm DNA damage in patients with varicocele: Relationship with seminal oxidative stress. Hum. Reprod. 2006, 21, 986–993. [Google Scholar] [CrossRef] [Green Version]
  7. Turner, T.T. The study of varicocele through the use of animal models. Hum. Reprod. Update 2001, 7, 78–84. [Google Scholar] [CrossRef] [Green Version]
  8. Agarwal, A.; Gupta, S.; Sikka, S. The role of free radicals and antioxidants in reproduction. Curr. Opin. Obstet. Gynecol. 2006, 18, 325–332. [Google Scholar] [CrossRef]
  9. Aitken, R.J.; Baker, M.A. Oxidative stress, sperm survival and fertility control. Mol. Cell. Endocrinol. 2006, 250, 66–69. [Google Scholar] [CrossRef]
  10. Gagnon, C.; Iwasaki, A.; De Lamirande, E.; Kovalski, N. Reactive oxygen species and human spermatozoa. Ann. N. Y. Acad. Sci. 1991, 637, 436–444. [Google Scholar] [CrossRef]
  11. De Lamirande, E.; Gagnon, C. Impact of reactive oxygen species on spermatozoa: A balancing act between beneficial and detrimental effects. Hum. Reprod. 1995, 10 (Suppl. 1), 15–21. [Google Scholar] [CrossRef] [PubMed]
  12. Tremellen, K. Oxidative stress and male infertility: A clinical perspective. Hum. Reprod. Update 2008, 14, 243–258. [Google Scholar] [CrossRef] [PubMed]
  13. Clermont, Y. Kinetics of spermatogenesis in mammals: Seminiferous epithelium cycle and spermatogonial renewal. Physiol. Rev. 1972, 52, 198–236. [Google Scholar] [CrossRef] [PubMed]
  14. Stocco, D.M.; McPhaul, M.J. Physiology of Testicular Steroidogenesis. In Knobil and Neill’s Physiology of Reproduction, 3rd ed.; Jimmy, D.N., Tony, M.P., Donald, W.P., John, R.G.C., David, M.d.K., JoAnne, S.R., Paul, M.W., Eds.; Academic Press: St Louis, MO, USA, 2006; pp. 977–1016. [Google Scholar]
  15. Robaire, B.; Hinton, B.T. The Epididymis, in Knobil and Neill’s Physiology of Reproduction, 4th ed.; Plant, T.M., Zeleznik, A.J., Eds.; Academic Press: San Diego, CA, USA, 2015; pp. 691–771. [Google Scholar]
  16. De Lamirande, E.; O’Flaherty, C. Sperm activation: Role of reactive oxygen species and kinases. Biochim. Biophys. Acta 2008, 1784, 106–115. [Google Scholar] [CrossRef]
  17. De Lamirande, E.; O’Flaherty, C. Sperm Capacitation as An Oxidative event, in Studies on Men’s Health and Fertility, Oxidative Stress in Applied Basic Research and Clinical Practice; Aitken, J., Alvarez, J., Agawarl, A., Eds.; Springer Science: Berlin/Heidelber, Germany, 2012; pp. 57–94. [Google Scholar]
  18. Yanagimachi, R. Mammalian fertilization, in The Physiology of Reproduction; Knobil, E., Neill, D., Eds.; Raven Press: New York, NY, USA, 1994; pp. 189–318. [Google Scholar]
  19. Aitken, R.J. Sperm function tests and fertility. Int. J. Androl. 2006, 29, 69–75. [Google Scholar] [CrossRef]
  20. Buffone, M.G.; Calamera, J.C.; Verstraeten, S.V.; Doncel, G.F. Capacitation-associated protein tyrosine phosphorylation and membrane fluidity changes are impaired in the spermatozoa of asthenozoospermic patients. Reproduction 2005, 129, 697–705. [Google Scholar] [CrossRef] [Green Version]
  21. De Lamirande, E.; Gagnon, C. Capacitation-associated production of superoxide anion by human spermatozoa. Free Radic. Biol. Med. 1995, 18, 487–495. [Google Scholar] [CrossRef]
  22. O’Flaherty, C.; Beorlegui, N.; Beconi, M.T. Participation of superoxide anion in the capacitation of cryopreserved bovine sperm. Int. J. Androl. 2003, 26, 109–114. [Google Scholar] [CrossRef]
  23. Burnaugh, L.; Sabeur, K.; Ball, B.A. Generation of superoxide anion by equine spermatozoa as detected by dihydroethidium. Theriogenology 2007, 67, 580–589. [Google Scholar] [CrossRef]
  24. Aitken, R.J.; Drevet, J.R. The Importance of Oxidative Stress in Determining the Functionality of Mammalian Spermatozoa: A Two-Edged Sword. Antioxidants 2020, 9, 111. [Google Scholar] [CrossRef] [Green Version]
  25. Aitken, R.J.; Harkiss, D.; Knox, W.; Paterson, M.; Irvine, D.S. A novel signal transduction cascade in capacitating human spermatozoa characterised by a redox-regulated, cAMP-mediated induction of tyrosine phosphorylation. J. Cell Sci. 1998, 111 Pt 5, 645–656. [Google Scholar] [PubMed]
  26. O’Flaherty, C.; de Lamirande, E.; Gagnon, C. Reactive oxygen species modulate independent protein phosphorylation pathways during human sperm capacitation. Free Radic. Biol. Med. 2006, 40, 1045–1055. [Google Scholar] [CrossRef] [PubMed]
  27. O’Flaherty, C.; de Lamirande, E.; Gagnon, C. Positive role of reactive oxygen species in mammalian sperm capacitation: Triggering and modulation of phosphorylation events. Free Radic. Biol. Med. 2006, 41, 528–540. [Google Scholar] [CrossRef] [PubMed]
  28. Lee, D.; Moawad, A.; Morielli, T.; Fernandez, M.; O’Flaherty, C. Peroxiredoxins prevent oxidative stress during human sperm capacitation. Mol. Hum. Reprod. 2017, 23, 106–115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. O’Flaherty, C. Redox regulation of mammalian sperm capacitation. Asian J. Androl. 2015, 17, 583–590. [Google Scholar] [CrossRef] [PubMed]
  30. Morielli, T.; O’Flaherty, C. Oxidative stress impairs function and increases redox protein modifications in human spermatozoa. Reproduction 2015, 149, 113–123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Aitken, R.J.; Harkiss, D.; Buckingham, D. Relationship between iron-catalysed lipid peroxidation potential and human sperm function. J. Reporod. Fertil. 1993, 98, 257–265. [Google Scholar] [CrossRef]
  32. Moawad, A.R.; Fernandez, M.C.; Scarlata, E.; Dodia, C.; Feinstein, S.I.; Fisher, A.B.; O’Flaherty, C. Deficiency of peroxiredoxin 6 or inhibition of its phospholipase A2 activity impair the in vitro sperm fertilizing competence in mice. Sci. Rep. 2017, 7, 12994. [Google Scholar] [CrossRef] [Green Version]
  33. Aitken, R.J.; Gordon, E.; Harkiss, D.; Twigg, J.P.; Milne, P.; Jennings, Z.; Irvine, D.S. Relative impact of oxidative stress on the functional competence and genomic integrity of human spermatozoa. Biol. Reprod. 1998, 59, 1037–1046. [Google Scholar] [CrossRef]
  34. Noblanc, A.; Kocer, A.; Drevet, J.R. Post-testicular protection of male gametes from oxidative damage. The role of the epididymis. Med. Sci. 2012, 28, 519–525. [Google Scholar]
  35. Iwasaki, A.; Gagnon, C. Formation of reactive oxygen species in spermatozoa of infertile patients. Fertil. Steril. 1992, 57, 409–416. [Google Scholar] [CrossRef]
  36. Jones, R.; Mann, T.; Sherins, R. Peroxidative breakdown of phospholipids in human spermatozoa, spermicidal properties of fatty acid peroxides, and protective action of seminal plasma. Fertil. Steril. 1979, 31, 531–537. [Google Scholar] [CrossRef]
  37. Alvarez, J.G.; Touchstone, J.C.; Blasco, L.; Storey, B.T. Spontaneous lipid peroxidation and production of hydrogen peroxide and superoxide in human spermatozoa. Superoxide dismutase as major enzyme protectant against oxygen toxicity. J. Androl. 1987, 8, 338–348. [Google Scholar] [PubMed]
  38. Aitken, R.J.; Whiting, S.; De Iuliis, G.N.; McClymont, S.; Mitchell, L.A.; Baker, M.A. Electrophilic Aldehydes Generated by Sperm Metabolism Activate Mitochondrial Reactive Oxygen Species Generation and Apoptosis by Targeting Succinate Dehydrogenase. J. Biol. Chem. 2012, 287, 33048–33060. [Google Scholar] [CrossRef] [Green Version]
  39. O’Flaherty, C. Peroxiredoxin 6: The Protector of Male Fertility. Antioxidants 2018, 7, 173. [Google Scholar] [CrossRef] [Green Version]
  40. Fernandez, M.C.; O’Flaherty, C. Peroxiredoxin 6 activates maintenance of viability and DNA integrity in human spermatozoa. Hum. Reprod. 2018, 33, 1394–1407. [Google Scholar] [CrossRef]
  41. Ozkosem, B.; Feinstein, S.I.; Fisher, A.B.; O’Flaherty, C. Absence of Peroxiredoxin 6 Amplifies the Effect of Oxidant Stress on Mobility and SCSA/CMA3 Defined Chromatin Quality and Impairs Fertilizing Ability of Mouse Spermatozoa. Biol. Reprod. 2016, 94, 1–10. [Google Scholar] [CrossRef] [Green Version]
  42. Aitken, R.J.; De Iuliis, G.N.; Finnie, J.M.; Hedges, A.; McLachlan, R.I. Analysis of the relationships between oxidative stress, DNA damage and sperm vitality in a patient population: Development of diagnostic criteria. Hum. Reprod. 2010, 25, 2415–2426. [Google Scholar] [CrossRef] [Green Version]
  43. Noblanc, A.; Damon-Soubeyrand, C.; Karrich, B.; Henry-Berger, J.; Cadet, R.; Saez, F.; Guiton, R.; Janny, L.; Pons-Rejraji, H.; Alvarez, J.G.; et al. DNA oxidative damage in mammalian spermatozoa: Where and why is the male nucleus affected? Free Radic. Biol. Med. 2013, 65, 719–723. [Google Scholar] [CrossRef]
  44. Cambi, M.; Tamburrino, L.; Marchiani, S.; Olivito, B.; Azzari, C.; Forti, G.; Baldi, E.; Muratori, M. Development of a specific method to evaluate 8-hydroxy,2-deoxyguanosine in sperm nuclei: Relationship with semen quality in a cohort of 94 subjects. Reproduction 2013, 145, 227–235. [Google Scholar] [CrossRef] [Green Version]
  45. O’Flaherty, C.; Matsushita-Fournier, D. Reactive oxygen species and protein modifications in spermatozoa. Biol. Reprod. 2017, 97, 577–585. [Google Scholar] [CrossRef] [PubMed]
  46. Salvolini, E.; Buldreghini, E.; Lucarini, G.; Vignini, A.; Di Primio, R.; Balercia, G. Nitric oxide synthase and tyrosine nitration in idiopathic asthenozoospermia: An immunohistochemical study. Fertil. Steril. 2012, 97, 554–560. [Google Scholar] [CrossRef] [PubMed]
  47. Morielli, T.; O’Flaherty, C. Oxidative stress promotes protein tyrosine nitration and S-glutathionylation impairing motility and capacitation in human spermatozoa. Free Radic. Biol. Med. 2012, 53 (Suppl. 2), S137. [Google Scholar] [CrossRef]
  48. Scarlata, E.; O’Flaherty, C. Antioxidant Enzymes and Male Fertility: Lessons from Knockout Models. Antioxid. Redox Signal. 2020, 32, 569–580. [Google Scholar] [CrossRef] [PubMed]
  49. Selvaratnam, J.S.; Robaire, B. Effects of Aging and Oxidative Stress on Spermatozoa of Superoxide-Dismutase 1- and Catalase-Null Mice1. Biol. Reprod. 2016, 95, 60–61. [Google Scholar] [CrossRef] [PubMed]
  50. Smith, T.B.; Baker, M.A.; Connaughton, H.S.; Habenicht, U.; Aitken, R.J. Functional deletion of Txndc2 and Txndc3 increases the susceptibility of spermatozoa to age-related oxidative stress. Free Radic. Biol. Med. 2013, 65, 872–881. [Google Scholar] [CrossRef]
  51. Chabory, E.; Damon, C.; Lenoir, A.; Kauselmann, G.; Kern, H.; Zevnik, B.; Garrel, C.; Saez, F.; Cadet, R.; Henry-Berger, J.; et al. Epididymis seleno-independent glutathione peroxidase 5 maintains sperm DNA integrity in mice. J. Clin. Investig. 2009, 119, 2074–2085. [Google Scholar] [CrossRef]
  52. Schneider, M.; Forster, H.; Boersma, A.; Seiler, A.; Wehnes, H.; Sinowatz, F.; Neumuller, C.; Deutsch, M.J.; Walch, A.; Hrabe de Angelis, M.; et al. Mitochondrial glutathione peroxidase 4 disruption causes male infertility. FASEB J. 2009, 23, 3233–3242. [Google Scholar] [CrossRef] [Green Version]
  53. Smarr, M.M.; Sapra, K.J.; Gemmill, A.; Kahn, L.G.; Wise, L.A.; Lynch, C.D.; Factor-Litvak, P.; Mumford, S.L.; Skakkebaek, N.E.; Slama, R.; et al. Is human fecundity changing? A discussion of research and data gaps precluding us from having an answer. Hum. Reprod. 2017, 32, 499–504. [Google Scholar] [CrossRef]
  54. Levine, H.; Jørgensen, N.; Martino-Andrade, A.; Mendiola, J.; Weksler-Derri, D.; Mindlis, I.; Pinotti, R.; Swan, S.H. Temporal trends in sperm count: A systematic review and meta-regression analysis. Hum. Reprod. Update 2017, 23, 646–659. [Google Scholar] [CrossRef]
  55. Skakkebaek, N.E.; Meyts, E.R.-D.; Louis, G.M.B.; Toppari, J.; Andersson, A.-M.; Eisenberg, M.L.; Jensen, T.K.; Jørgensen, N.; Swan, S.H.; Sapra, K.J.; et al. Male Reproductive Disorders and Fertility Trends: Influences of Environment and Genetic Susceptibility. Physiol. Rev. 2016, 96, 55–97. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Paul, C.; Robaire, B. Ageing of the male germ line. Nat. Rev. Urol. 2013, 10, 227–234. [Google Scholar] [CrossRef] [PubMed]
  57. Noblanc, A.; Klaassen, A.; Robaire, B. The Exacerbation of Aging and Oxidative Stress in the Epididymis of Sod1 Null Mice. Antioxidants 2020, 9, 151. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Wu, P.Y.; Scarlata, E.; O’Flaherty, C. Long-Term Adverse Effects of Oxidative Stress on Rat Epididymis and Spermatozoa. Antioxidants 2020, 9, 170. [Google Scholar] [CrossRef] [Green Version]
  59. O’Flaherty, C.; Boisvert, A.; Manku, G.; Culty, M. Protective Role of Peroxiredoxins against Reactive Oxygen Species in Neonatal Rat Testicular Gonocytes. Antioxidants 2019, 9, 32. [Google Scholar] [CrossRef] [Green Version]
  60. Hamilton, L.E.; Zigo, M.; Mao, J.; Xu, W.; Sutovsky, P.; O’Flaherty, C.; Oko, R. GSTO2 Isoforms Participate in the Oxidative Regulation of the Plasmalemma in Eutherian Spermatozoa during Capacitation. Antioxidants 2019, 8, 601. [Google Scholar] [CrossRef] [Green Version]
  61. Nowicka-Bauer, K.; Nixon, B. Molecular Changes Induced by Oxidative Stress that Impair Human Sperm Motility. Antioxidants 2020, 9, 134. [Google Scholar] [CrossRef] [Green Version]
  62. Drevet, J.R.; Aitken, R.J. Oxidation of Sperm Nucleus in Mammals: A Physiological Necessity to Some Extent with Adverse Impacts on Oocyte and Offspring. Antioxidants 2020, 9, 95. [Google Scholar] [CrossRef] [Green Version]
  63. Villaverde, A.I.S.B.; Netherton, J.; Baker, M.A. From Past to Present: The Link Between Reactive Oxygen Species in Sperm and Male Infertility. Antioxidants 2019, 8, 616. [Google Scholar] [CrossRef] [Green Version]
  64. Martin-Hidalgo, D.; Bragado, M.J.; Batista, A.R.; Oliveira, P.F.; Alves, M.G. Antioxidants and Male Fertility: From Molecular Studies to Clinical Evidence. Antioxidants 2019, 8, 89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Peña, F.J.; O’Flaherty, C.; Ortiz Rodríguez, J.M.; Martín Cano, F.E.; Gaitskell-Phillips, G.L.; Gil, M.C.; Ortega Ferrusola, C. Redox Regulation and Oxidative Stress: The Particular Case of the Stallion Spermatozoa. Antioxidants 2019, 8, 567. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Showell, M.G.; Mackenzie-Proctor, R.; Brown, J.; Yazdani, A.; Stankiewicz, M.T.; Hart, R.J. Antioxidants for male subfertility. Cochrane Database Syst. Rev. 2014, 12, CD007411. [Google Scholar] [CrossRef] [PubMed]
  67. Windsor, B.; Popovich, I.; Jordan, V.; Showell, M.; Shea, B.; Farquhar, C. Methodological quality of systematic reviews in subfertility: A comparison of Cochrane and non-Cochrane systematic reviews in assisted reproductive technologies. Hum. Reprod. 2012, 27, 3460–3466. [Google Scholar] [CrossRef] [PubMed]
  68. Zini, A.; Al-Hathal, N. therapy in male infertility: Fact or fiction? Asian J. Androl. 2011, 13, 374–381. [Google Scholar] [CrossRef] [PubMed] [Green Version]

Share and Cite

MDPI and ACS Style

O’Flaherty, C. Reactive Oxygen Species and Male Fertility. Antioxidants 2020, 9, 287. https://doi.org/10.3390/antiox9040287

AMA Style

O’Flaherty C. Reactive Oxygen Species and Male Fertility. Antioxidants. 2020; 9(4):287. https://doi.org/10.3390/antiox9040287

Chicago/Turabian Style

O’Flaherty, Cristian. 2020. "Reactive Oxygen Species and Male Fertility" Antioxidants 9, no. 4: 287. https://doi.org/10.3390/antiox9040287

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop