Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Perspective
  • Published:

Male infertility and somatic health — insights into lipid damage as a mechanistic link

Abstract

Over the past decade, mounting evidence has shown an alarming association between male subfertility and poor somatic health, with substantial evidence supporting the increased incidence of oncological disease, cardiovascular disease, metabolic disorders and autoimmune diseases in men who have previously received a subfertility diagnosis. This paradigm is concerning, but might also provide a novel window for a crucial health reform in which the infertile phenotype could serve as an indication of potential pathological conditions. One of the major limiting factors in this association is the poor understanding of the molecular features that link infertility with comorbidities across the life course. Enzymes involved in the lipid oxidation process might provide novel clues to reconcile the mechanistic basis of infertility with incident pathological conditions. Building research capacity in this area is essential to enhance the early detection of disease states and provide crucial information about the disease risk of offspring conceived through assisted reproduction.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Incident comorbidities of male infertility and the role of lipid oxidation.
Fig. 2: Male infertility, incident comorbidities and lipid oxidation.
Fig. 3: Biosynthesis of oxidized phospholipids by ALOX15 and consequences for human health and fertility.
Fig. 4: Lipid damage as a molecular link between male infertility and somatic health.

Similar content being viewed by others

References

  1. Zegers-Hochschild, F. et al. International Committee for Monitoring Assisted Reproductive Technology (ICMART) and the World Health Organization (WHO) revised glossary of ART terminology, 2009. Fertil. Steril. 92, 1520–1524 (2009).

    CAS  PubMed  Google Scholar 

  2. Agarwal, A., Mulgund, A., Hamada, A. & Chyatte, M. R. A unique view on male infertility around the globe. Reprod. Biol. Endocrinol. 13, 37–37 (2015).

    PubMed  PubMed Central  Google Scholar 

  3. McLachlan, R. I. & de Kretser, D. M. Male infertility: the case for continued research. Med. J. Aust. 174, 116–117 (2001).

    CAS  PubMed  Google Scholar 

  4. Morris, J. K., Alberman, E., Scott, C. & Jacobs, P. Is the prevalence of Klinefelter syndrome increasing? Eur. J. Hum. Genet. 16, 163–170 (2008).

    CAS  PubMed  Google Scholar 

  5. Gunes, S., Al-Sadaan, M. & Agarwal, A. Spermatogenesis, DNA damage and DNA repair mechanisms in male infertility. Reprod. Biomed. Online 31, 309–319 (2015).

    CAS  PubMed  Google Scholar 

  6. Punjani, N. & Lamb, D. J. Male infertility and genitourinary birth defects: there is more than meets the eye. Fertil. Steril. 114, 209–218 (2020).

    CAS  PubMed  Google Scholar 

  7. Houston, B. J. et al. A systematic review of the validated monogenic causes of human male infertility: 2020 update and a discussion of emerging gene-disease relationships. Hum. Reprod. Update 28, 15–29 (2021).

    PubMed  PubMed Central  Google Scholar 

  8. Olesen, I. A. et al. Clinical, genetic, biochemical, and testicular biopsy findings among 1,213 men evaluated for infertility. Fertil. Steril. 107, 74–82.e77 (2017).

    PubMed  Google Scholar 

  9. Punab, M. et al. Causes of male infertility: a 9-year prospective monocentre study on 1737 patients with reduced total sperm counts. Hum. Reprod. 32, 18–31 (2017).

    CAS  PubMed  Google Scholar 

  10. Tüttelmann, F., Ruckert, C. & Röpke, A. Disorders of spermatogenesis: perspectives for novel genetic diagnostics after 20 years of unchanged routine. Med. Genet. 30, 12–20 (2018).

    PubMed  PubMed Central  Google Scholar 

  11. Oud, M. S. et al. Exome sequencing reveals novel causes as well as new candidate genes for human globozoospermia. Hum. Reprod. 35, 240–252 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Murshidi, M. M., Choy, J. T. & Eisenberg, M. L. Male infertility and somatic health. Urol. Clin. North. Am. 47, 211–217 (2020).

    PubMed  Google Scholar 

  13. Chen, T., Belladelli, F., Giudice, F. D. & Eisenberg, M. L. Male fertility as a marker for health. Reprod. Biomed. Online 44, 131–144 (2022).

    PubMed  Google Scholar 

  14. Thonneau, P. et al. Incidence and main causes of infertility in a resident population (1,850,000) of three French regions (1988–1989). Hum. Reprod. 6, 811–816 (1991).

    CAS  PubMed  Google Scholar 

  15. Hamada, A., Esteves, S. C., Nizza, M. & Agarwal, A. Unexplained male infertility: diagnosis and management. Int. Braz. J. Urol. 38, 576–594 (2012).

    PubMed  Google Scholar 

  16. Salonia, A. et al. Are infertile men less healthy than fertile men? Results of a prospective case-control survey. Eur. Urol. 56, 1025–1031 (2009).

    PubMed  Google Scholar 

  17. Eisenberg, M. L., Li, S., Behr, B., Pera, R. R. & Cullen, M. R. Relationship between semen production and medical comorbidity. Fertil. Steril. 103, 66–71 (2015).

    PubMed  Google Scholar 

  18. Latif, T. et al. Semen quality as a predictor of subsequent morbidity: a Danish cohort study of 4,712 men with long-term follow-up. Am. J. Epidemiol. 186, 910–917 (2017).

    PubMed  Google Scholar 

  19. Jensen, T. K., Jacobsen, R., Christensen, K., Nielsen, N. C. & Bostofte, E. Good semen quality and life expectancy: a cohort study of 43,277 men. Am. J. Epidemiol. 170, 559–565 (2009).

    PubMed  Google Scholar 

  20. Punjani, N. & Lamb, D. J. Canary in the coal mine? Male infertility as a marker of overall health. Annu. Rev. Genet. 54, 465–486 (2020).

    CAS  PubMed  Google Scholar 

  21. Glazer, C. H. et al. Male factor infertility and risk of death: a nationwide record-linkage study. Hum. Reprod. 34, 2266–2273 (2019).

    PubMed  Google Scholar 

  22. Eisenberg, M. L. et al. Semen quality, infertility and mortality in the USA. Hum. Reprod. 29, 1567–1574 (2014).

    PubMed  PubMed Central  Google Scholar 

  23. Del Giudice, F. et al. Increased mortality among men diagnosed with impaired fertility: analysis of US claims data. Urology 147, 143–149 (2021).

    PubMed  Google Scholar 

  24. Duffy, J. M. N. et al. Top 10 priorities for future infertility research: an international consensus development study. Hum. Reprod. 35, 2715–2724 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Kasman, A. M., Del Giudice, F. & Eisenberg, M. L. New insights to guide patient care: the bidirectional relationship between male infertility and male health. Fertil. Steril. 113, 469–477 (2020).

    CAS  PubMed  Google Scholar 

  26. Choy, J. T. & Eisenberg, M. L. Comprehensive men’s health and male infertility. Transl. Androl. Urol. 9, S239–S243 (2020).

    PubMed  PubMed Central  Google Scholar 

  27. Del Giudice, F. et al. Clinical correlation among male infertility and overall male health: a systematic review of the literature. Investig. Clin. Urol. 61, 355–371 (2020).

    PubMed  PubMed Central  Google Scholar 

  28. Hawksworth, D. J. & Burnett, A. L. Nonalcoholic fatty liver disease, male sexual dysfunction, and infertility: common links, common problems. Sex. Med. Rev. 8, 274–285 (2020).

    PubMed  Google Scholar 

  29. Del Giudice, F. et al. Association between male infertility and male-specific malignancies: systematic review and meta-analysis of population-based retrospective cohort studies. Fertil. Steril. 114, 984–996 (2020).

    PubMed  Google Scholar 

  30. Ahmadi, S., Bashiri, R., Ghadiri-Anari, A. & Nadjarzadeh, A. Antioxidant supplements and semen parameters: an evidence based review. Int. J. Reprod. Biomed. 14, 729–736 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Majzoub, A. & Agarwal, A. Systematic review of antioxidant types and doses in male infertility: benefits on semen parameters, advanced sperm function, assisted reproduction and live-birth rate. Arab. J. Urol. 16, 113–124 (2018).

    PubMed  PubMed Central  Google Scholar 

  32. Ross, C. et al. A systematic review of the effect of oral antioxidants on male infertility. Reprod. Biomed. Online 20, 711–723 (2010).

    CAS  PubMed  Google Scholar 

  33. Ventimiglia, E. et al. Infertility as a proxy of general male health: results of a cross-sectional survey. Fertil. Steril. 104, 48–55 (2015).

    PubMed  Google Scholar 

  34. Eisenberg, M. L., Li, S., Cullen, M. R. & Baker, L. C. Increased risk of incident chronic medical conditions in infertile men: analysis of United States claims data. Fertil. Steril. 105, 629–636 (2016).

    PubMed  Google Scholar 

  35. Fisher, J. R. W. & Hammarberg, K. Psychological and social aspects of infertility in men: an overview of the evidence and implications for psychologically informed clinical care and future research. Asian J. Androl. 14, 121–129 (2012).

    CAS  PubMed  Google Scholar 

  36. Hasanpoor-Azghdy, S. B., Simbar, M. & Vedadhir, A. The emotional-psychological consequences of infertility among infertile women seeking treatment: results of a qualitative study. Iran. J. Reprod. Med. 12, 131–138 (2014).

    PubMed  PubMed Central  Google Scholar 

  37. Schuppe, H.-C. et al. Urogenital infection as a risk factor for male infertility. Dtsch. Arztebl Int. 114, 339–346 (2017).

    PubMed  PubMed Central  Google Scholar 

  38. Cunningham, K. A. & Beagley, K. W. Male genital tract chlamydial infection: implications for pathology and infertility. Biol. Reprod. 79, 180–189 (2008).

    CAS  PubMed  Google Scholar 

  39. Cooper, T. G. et al. World Health Organization reference values for human semen characteristics. Hum. Reprod. Update 16, 231–245 (2010).

    PubMed  Google Scholar 

  40. World Health Organization. WHO laboratory manual for the examination and processing of human semen 5th edn (WHO, 2010).

  41. Maddatu, J., Anderson-Baucum, E. & Evans-Molina, C. Smoking and the risk of type 2 diabetes. Transl. Res. 184, 101–107 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Polsky, S. & Ellis, S. L. Obesity, insulin resistance, and type 1 diabetes mellitus. Curr. Opin. Endocrinol. Diabetes Obes. 22, 277–282 (2015).

    CAS  PubMed  Google Scholar 

  43. Jokinen, E. Obesity and cardiovascular disease. Minerva Pediatr. 67, 25–32 (2015).

    CAS  PubMed  Google Scholar 

  44. Ambrose, J. A. & Barua, R. S. The pathophysiology of cigarette smoking and cardiovascular disease: an update. J. Am. Coll. Cardiol. 43, 1731–1737 (2004).

    CAS  PubMed  Google Scholar 

  45. Samet, J. M. Tobacco smoking: the leading cause of preventable disease worldwide. Thorac. Surg. Clin. 23, 103–112 (2013).

    PubMed  Google Scholar 

  46. Kolb, R., Sutterwala, F. S. & Zhang, W. Obesity and cancer: inflammation bridges the two. Curr. Opin. Pharmacol. 29, 77–89 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Herlihy, A. S., Halliday, J. L., Cock, M. L. & McLachlan, R. I. The prevalence and diagnosis rates of Klinefelter syndrome: an Australian comparison. Med. J. Aust. 194, 24–28 (2011).

    PubMed  Google Scholar 

  48. Röpke, A. & Tüttelmann, F. Mechanisms in endocrinology: aberrations of the X chromosome as cause of male infertility. Eur. J. Endocrinol. 177, R249–R259 (2017).

    PubMed  Google Scholar 

  49. Ramakrishnan, V., Kumar, S. G. & Pandiyan, R. Klinefelter syndrome and its association with male infertility. Asian Pac. J. Reprod. 3, 77–79 (2014).

    Google Scholar 

  50. Bojesen, A., Juul, S., Birkebaek, N. H. & Gravholt, C. H. Morbidity in Klinefelter syndrome: a Danish register study based on hospital discharge diagnoses. J. Clin. Endocrinol. Metab. 91, 1254–1260 (2006).

    CAS  PubMed  Google Scholar 

  51. Bojesen, A., Juul, S., Birkebaek, N. & Gravholt, C. H. Increased mortality in Klinefelter syndrome. J. Clin. Endocrinol. Metab. 89, 3830–3834 (2004).

    CAS  PubMed  Google Scholar 

  52. Swerdlow, A. J., Higgins, C. D., Schoemaker, M. J., Wright, A. F. & Jacobs, P. A. Mortality in patients with Klinefelter syndrome in Britain: a cohort study. J. Clin. Endocrinol. Metab. 90, 6516–6522 (2005).

    CAS  PubMed  Google Scholar 

  53. Simon, L., Zini, A., Dyachenko, A., Ciampi, A. & Carrell, D. T. A systematic review and meta-analysis to determine the effect of sperm DNA damage on in vitro fertilization and intracytoplasmic sperm injection outcome. Asian J. Androl. 19, 80–90 (2017).

    PubMed  Google Scholar 

  54. Marchetti, F., Bishop, J. B., Cosentino, L., Moore, D. II & Wyrobek, A. J. Paternally transmitted chromosomal aberrations in mouse zygotes determine their embryonic fate. Biol. Reprod. 70, 616–624 (2004).

    CAS  PubMed  Google Scholar 

  55. Cheung, S., Parrella, A., Rosenwaks, Z. & Palermo, G. D. Genetic and epigenetic profiling of the infertile male. PLoS ONE 14, e0214275 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Choy, J. T. & Eisenberg, M. L. Male infertility as a window to health. Fertil. Steril. 110, 810–814 (2018).

    PubMed  Google Scholar 

  57. Baker, S. M. et al. Involvement of mouse Mlh1 in DNA mismatch repair and meiotic crossing over. Nat. Genet. 13, 336–342 (1996).

    CAS  PubMed  Google Scholar 

  58. Ji, G. et al. Common variants in mismatch repair genes associated with increased risk of sperm DNA damage and male infertility. BMC Med. 10, 49 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Sun, F. et al. Abnormal progression through meiosis in men with nonobstructive azoospermia. Fertil. Steril. 87, 565–571 (2007).

    PubMed  Google Scholar 

  60. Wang, C., Wang, Y., Hughes, K. S., Parmigiani, G. & Braun, D. Penetrance of colorectal cancer among mismatch repair gene mutation carriers: a meta-analysis. JNCI Cancer Spectr. 4, pkaa027–pkaa027 (2020).

    PubMed  PubMed Central  Google Scholar 

  61. Paul, C. et al. Deletion of genes implicated in protecting the integrity of male germ cells has differential effects on the incidence of DNA breaks and germ cell loss. PLoS ONE 2, e989 (2007).

    PubMed  PubMed Central  Google Scholar 

  62. Reitmair, A. H. et al. MSH2 deficient mice are viable and susceptible to lymphoid tumours. Nat. Genet. 11, 64–70 (1995).

    CAS  PubMed  Google Scholar 

  63. Zhao, L. Mismatch repair protein expression in patients with stage II and III sporadic colorectal cancer. Oncol. Lett. 15, 8053–8061 (2018).

    PubMed  PubMed Central  Google Scholar 

  64. Huang, Y.-L. et al. The role of ERCC1 and AFP gene polymorphism in hepatocellular carcinoma. Medicine 98, e15090 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Koutsoukos, K. et al. Clinical perspectives of ERCC1 in bladder cancer. Int. J. Mol. Sci. 21, 8829 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Miller, A. C. et al. Cystic fibrosis carriers are at increased risk for a wide range of cystic fibrosis-related conditions. Proc. Natl Acad. Sci. USA 117, 1621 (2020).

    CAS  PubMed  Google Scholar 

  67. Stoltz, D. A., Meyerholz, D. K. & Welsh, M. J. Origins of cystic fibrosis lung disease. N. Engl. J. Med. 372, 351–362 (2015).

    PubMed  PubMed Central  Google Scholar 

  68. Cutting, G. R. Cystic fibrosis genetics: from molecular understanding to clinical application. Nat. Rev. Genet. 16, 45–56 (2015).

    CAS  PubMed  Google Scholar 

  69. Elborn, J. S. Cystic fibrosis. Lancet 388, 2519–2531 (2016).

    CAS  PubMed  Google Scholar 

  70. Cuppens, H. & Cassiman, J. J. CFTR mutations and polymorphisms in male infertility. Int. J. Androl. 27, 251–256 (2004).

    CAS  PubMed  Google Scholar 

  71. Chen, H., Ruan, Y. C., Xu, W. M., Chen, J. & Chan, H. C. Regulation of male fertility by CFTR and implications in male infertility. Hum. Reprod. Update 18, 703–713 (2012).

    CAS  PubMed  Google Scholar 

  72. Ishiguro, H., Steward, M. & Naruse, S. Cystic fibrosis transmembrane conductance regulator and SLC26 transporters in HCO3 secretion by pancreatic duct cells. Sheng Li Xue Bao 59, 465–476 (2007).

    CAS  PubMed  Google Scholar 

  73. Patrizio, P., Ord, T., Silber, S. J. & Asch, R. H. Cystic fibrosis mutations impair the fertilization rate of epididymal sperm from men with congenital absence of the vas deferens. Hum. Reprod. 8, 1259–1263 (1993).

    CAS  PubMed  Google Scholar 

  74. Schulz, S. et al. Increased frequency of cystic fibrosis transmembrane conductance regulator gene mutations in infertile males. Fertil. Steril. 85, 135–138 (2006).

    CAS  PubMed  Google Scholar 

  75. Colaco, S. & Modi, D. Consequences of Y chromosome microdeletions beyond male infertility. J. Assist. Reprod. Genet. 36, 1329–1337 (2019).

    PubMed  PubMed Central  Google Scholar 

  76. Forsberg, L. A. et al. Mosaic loss of chromosome Y in peripheral blood is associated with shorter survival and higher risk of cancer. Nat. Genet. 46, 624–628 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. García Rodríguez, A., de la Casa, M., Johnston, S., Gosálvez, J. & Roy, R. Association of polymorphisms in genes coding for antioxidant enzymes and human male infertility. Ann. Hum. Genet. 83, 63–72 (2019).

    PubMed  Google Scholar 

  78. Nudell, D., Castillo, M., Turek, P. J. & Pera, R. R. Increased frequency of mutations in DNA from infertile men with meiotic arrest. Hum. Reprod. 15, 1289–1294 (2000).

    CAS  PubMed  Google Scholar 

  79. Behboudi-Gandevani, S., Bidhendi Yarandi, R., Rostami Dovom, M., Azizi, F. & Ramezani Tehrani, F. The association between male infertility and cardiometabolic disturbances: a population-based study. Int. J. Endocrinol. Metab. 19, e107418 (2021).

    PubMed  PubMed Central  Google Scholar 

  80. Cariati, F., D’Argenio, V. & Tomaiuolo, R. The evolving role of genetic tests in reproductive medicine. J. Transl. Med. 17, 267–267 (2019).

    PubMed  PubMed Central  Google Scholar 

  81. Bonde, J. P. et al. The epidemiologic evidence linking prenatal and postnatal exposure to endocrine disrupting chemicals with male reproductive disorders: a systematic review and meta-analysis. Hum. Reprod. Update 23, 104–125 (2016).

    PubMed  PubMed Central  Google Scholar 

  82. Wyrobek, A. J. Methods and concepts in detecting abnormal reproductive outcomes of paternal origin. Reprod. Toxicol. 7 (Suppl. 1), 3–16 (1993).

    PubMed  Google Scholar 

  83. Mehrpour, O., Karrari, P., Zamani, N., Tsatsakis, A. M. & Abdollahi, M. Occupational exposure to pesticides and consequences on male semen and fertility: a review. Toxicol. Lett. 230, 146–156 (2014).

    CAS  PubMed  Google Scholar 

  84. Krzastek, S. C., Farhi, J., Gray, M. & Smith, R. P. Impact of environmental toxin exposure on male fertility potential. Transl. Androl. Urol. 9, 2797–2813 (2020).

    PubMed  PubMed Central  Google Scholar 

  85. Wdowiak, A., Skrzypek, M., Stec, M. & Panasiuk, L. Effect of ionizing radiation on the male reproductive system. Ann. Agric. Env. Med. 26, 210–216 (2019).

    CAS  Google Scholar 

  86. De Felice, F. et al. Radiation effects on male fertility. Andrology 7, 2–7 (2019).

    PubMed  Google Scholar 

  87. Adams, J. A., Galloway, T. S., Mondal, D., Esteves, S. C. & Mathews, F. Effect of mobile telephones on sperm quality: a systematic review and meta-analysis. Environ. Int. 70, 106–112 (2014).

    PubMed  Google Scholar 

  88. Kovac, J. R., Khanna, A. & Lipshultz, L. I. The effects of cigarette smoking on male fertility. Postgrad. Med. 127, 338–341 (2015).

    PubMed  PubMed Central  Google Scholar 

  89. Office of the Surgeon General; Office on Smoking and Health (US). The Health Consequences of Smoking: A Report of the Surgeon General (Centers for Disease Control and Prevention, 2004).

  90. Katib, A. Mechanisms linking obesity to male infertility. Cent. European J. Urol. 68, 79–85 (2015).

    PubMed  PubMed Central  Google Scholar 

  91. Sermondade, N. et al. Obesity and increased risk for oligozoospermia and azoospermia. Arch. Intern. Med. 172, 440–442 (2012).

    PubMed  PubMed Central  Google Scholar 

  92. Eisenberg, M. L. et al. The relationship between male BMI and waist circumference on semen quality: data from the LIFE study. Hum. Reprod. 29, 193–200 (2014).

    PubMed  Google Scholar 

  93. Hammoud, A. O., Gibson, M., Peterson, C. M., Meikle, A. W. & Carrell, D. T. Impact of male obesity on infertility: a critical review of the current literature. Fertil. Steril. 90, 897–904 (2008).

    PubMed  Google Scholar 

  94. Hammoud, A. O. et al. Male obesity and alteration in sperm parameters. Fertil. Steril. 90, 2222–2225 (2008).

    PubMed  Google Scholar 

  95. Magnusdottir, E. V., Thorsteinsson, T., Thorsteinsdottir, S., Heimisdottir, M. & Olafsdottir, K. Persistent organochlorines, sedentary occupation, obesity and human male subfertility. Hum. Reprod. 20, 208–215 (2005).

    PubMed  Google Scholar 

  96. Jensen, T. K. et al. Body mass index in relation to semen quality and reproductive hormones among 1,558 Danish men. Fertil. Steril. 82, 863–870 (2004).

    CAS  PubMed  Google Scholar 

  97. Chavarro, J. E., Toth, T. L., Wright, D. L., Meeker, J. D. & Hauser, R. Body mass index in relation to semen quality, sperm DNA integrity, and serum reproductive hormone levels among men attending an infertility clinic. Fertil. Steril. 93, 2222–2231 (2010).

    CAS  PubMed  Google Scholar 

  98. Abdelaal, M., le Roux, C. W. & Docherty, N. G. Morbidity and mortality associated with obesity. Ann. Transl. Med. 5, 161–161 (2017).

    PubMed  PubMed Central  Google Scholar 

  99. Håkonsen, L. B. et al. Does weight loss improve semen quality and reproductive hormones? Results from a cohort of severely obese men. Reprod. Health 8, 24–24 (2011).

    PubMed  PubMed Central  Google Scholar 

  100. Crisóstomo, L. et al. Diet during early life defines testicular lipid content and sperm quality in adulthood. Am. J. Physiol. Endocrinol. Metab. 319, E1061–E1073 (2020).

    PubMed  Google Scholar 

  101. Aitken, R. J. & Baker, M. A. The role of genetics and oxidative stress in the etiology of male infertility — a unifying hypothesis? Front. Endocrinol. 11, 581838 (2020).

    Google Scholar 

  102. Brubaker, W. D., Li, S., Baker, L. C. & Eisenberg, M. L. Increased risk of autoimmune disorders in infertile men: analysis of US claims data. J. Androl. 6, 94–98 (2018).

    CAS  Google Scholar 

  103. Guo, D., Li, S., Behr, B. & Eisenberg, M. L. Hypertension and male fertility. World J. Mens Health 35, 59–64 (2017).

    PubMed  PubMed Central  Google Scholar 

  104. Walsh, T. J., Croughan, M. S., Schembri, M., Chan, J. M. & Turek, P. J. Increased risk of testicular germ cell cancer among infertile men. Arch. Intern. Med. 169, 351–356 (2009).

    PubMed  PubMed Central  Google Scholar 

  105. Hernández, B., Reilly, R. B. & Kenny, R. A. Investigation of multimorbidity and prevalent disease combinations in older Irish adults using network analysis and association rules. Sci. Rep. 9, 14567 (2019).

    PubMed  PubMed Central  Google Scholar 

  106. Huang, Y.-Q. et al. Charlson comorbidity index helps predict the risk of mortality for patients with type 2 diabetic nephropathy. J. Zhejiang Univ. Sci. B 15, 58–66 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Fraccaro, P. et al. Predicting mortality from change-over-time in the Charlson Comorbidity Index: a retrospective cohort study in a data-intensive UK health system. Medicine 95, e4973 (2016).

    PubMed  PubMed Central  Google Scholar 

  108. Shiraishi, K. & Matsuyama, H. Effects of medical comorbidity on male infertility and comorbidity treatment on spermatogenesis. Fertil. Steril. 110, 1006–1011 (2018).

    PubMed  Google Scholar 

  109. Muciaccia, B. et al. Higher clusterin immunolabeling and sperm DNA damage levels in hypertensive men compared with controls. Hum. Reprod. 27, 2267–2276 (2012).

    CAS  PubMed  Google Scholar 

  110. Glazer, C. H. et al. Male factor infertility and risk of multiple sclerosis: a register-based cohort study. Mult. Scler. 24, 1835–1842 (2018).

    PubMed  Google Scholar 

  111. Xu, R., Centola, G. M. & Tanrikut, C. Genitourinary cancer patients have worse baseline semen parameters than healthy sperm bankers. J. Androl. 7, 449–453 (2019).

    CAS  Google Scholar 

  112. Ferlin, A. et al. Sperm count and hypogonadism as markers of general male health. Eur. Urol. Focus 7, 205–213 (2019).

    PubMed  Google Scholar 

  113. American Diabetes Association. Diagnosis and classification of diabetes mellitus. Diabetes Care 37 (Suppl. 1), S81–S90 (2014).

    Google Scholar 

  114. Boeri, L. et al. Undiagnosed prediabetes is highly prevalent in primary infertile men — results from a cross-sectional study. BJU Int. 123, 1070–1077 (2019).

    CAS  PubMed  Google Scholar 

  115. Alberti, K. G. M. M. & Zimmet, P. Z. Definition, diagnosis and classification of diabetes mellitus and its complications. Part 1: diagnosis and classification of diabetes mellitus. Provisional report of a WHO Consultation. Diabet. Med. 15, 539–553 (1998).

    CAS  PubMed  Google Scholar 

  116. Kim, S. H., Chunawala, L., Linde, R. & Reaven, G. M. Comparison of the 1997 and 2003 American Diabetes Association classification of impaired fasting glucose: impact on prevalence of impaired fasting glucose, coronary heart disease risk factors, and coronary heart disease in a community-based medical practice. J. Am. Coll. Cardiol. 48, 293–297 (2006).

    PubMed  Google Scholar 

  117. Bener, A., Al-Ansari, A. A., Zirie, M. & Al-Hamaq, A. O. Is male fertility associated with type 2 diabetes mellitus? Int. Urol. Nephrol. 41, 777–784 (2009).

    PubMed  Google Scholar 

  118. Glazer, C. H. et al. Risk of diabetes according to male factor infertility: a register-based cohort study. Hum. Reprod. 32, 1474–1481 (2017).

    PubMed  PubMed Central  Google Scholar 

  119. Green, D. M. et al. Cumulative alkylating agent exposure and semen parameters in adult survivors of childhood cancer: a report from the St Jude Lifetime Cohort Study. Lancet Oncol. 15, 1215–1223 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Wang, N. N., Dallas, K., Li, S., Baker, L. & Eisenberg, M. L. The association between varicocoeles and vascular disease: an analysis of US claims data. J. Androl. 6, 99–103 (2018).

    CAS  Google Scholar 

  121. Pakpoor, J., Goldacre, R., Schmierer, K., Giovannoni, G. & Goldacre, M. J. Testicular hypofunction and multiple sclerosis risk: a record-linkage study. Ann. Neurol. 76, 625–628 (2014).

    PubMed  Google Scholar 

  122. Ferguson, L. & Agoulnik, A. I. Testicular cancer and cryptorchidism. Front. Endocrinol. 4, 32–32 (2013).

    Google Scholar 

  123. Hanson, H. A. et al. Subfertility increases risk of testicular cancer: evidence from population-based semen samples. Fertil. Steril. 105, 322–328 (2016).

    PubMed  Google Scholar 

  124. Jacobsen, R. et al. Risk of testicular cancer in men with abnormal semen characteristics: cohort study. BMJ 321, 789–792 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Raman, J. D., Nobert, C. F. & Goldstein, M. Increased incidence of testicular cancer in men presenting with infertility and abnormal semen analysis. J. Urol. 174, 1819–1822; discussion 1822 (2005).

    PubMed  Google Scholar 

  126. Zheng, J. F. et al. ICSI treatment of severe male infertility can achieve prospective embryo quality compared with IVF of fertile donor sperm on sibling oocytes. Asian J. Androl. 17, 845–849 (2015).

    PubMed  PubMed Central  Google Scholar 

  127. Crawford, G. E. & Ledger, W. L. In vitro fertilisation/intracytoplasmic sperm injection beyond 2020. BJOG 126, 237–243 (2019).

    CAS  PubMed  Google Scholar 

  128. Al-Jebari, Y. et al. Risk of prostate cancer for men fathering through assisted reproduction: nationwide population based register study. BMJ 366, l5214 (2019).

    PubMed  PubMed Central  Google Scholar 

  129. Walsh, T. J. et al. Increased risk of high-grade prostate cancer among infertile men. Cancer 116, 2140–2147 (2010).

    PubMed  Google Scholar 

  130. Eisenberg, M. L., Li, S., Brooks, J. D., Cullen, M. R. & Baker, L. C. Increased risk of cancer in infertile men: analysis of US claims data. J. Urol. 193, 1596–1601 (2015).

    PubMed  Google Scholar 

  131. Eisenberg, M. L., Betts, P., Herder, D., Lamb, D. J. & Lipshultz, L. I. Increased risk of cancer among azoospermic men. Fertil. Steril. 100, 681–685 (2013).

    PubMed  PubMed Central  Google Scholar 

  132. Agarwal, A. et al. Male oxidative stress infertility (MOSI): proposed terminology and clinical practice guidelines for management of idiopathic male infertility. World J. Mens Health 37, 296–312 (2019).

    PubMed  PubMed Central  Google Scholar 

  133. Oborna, I. et al. Increased lipid peroxidation and abnormal fatty acid profiles in seminal and blood plasma of normozoospermic males from infertile couples. Hum. Reprod. 25, 308–316 (2010).

    CAS  PubMed  Google Scholar 

  134. Pasqualotto, F. F. et al. Oxidative stress in normospermic men undergoing infertility evaluation. J. Androl. 22, 316–322 (2001).

    CAS  PubMed  Google Scholar 

  135. Aitken, R. J., Clarkson, J. S. & Fishel, S. Generation of reactive oxygen species, lipid peroxidation, and human sperm function. Biol. Reprod. 41, 183–197 (1989).

    CAS  PubMed  Google Scholar 

  136. Aitken, R. J., Gibb, Z., Baker, M. A., Drevet, J. & Gharagozloo, P. Causes and consequences of oxidative stress in spermatozoa. Reprod. Fertil. Dev. 28, 1–10 (2016).

    CAS  PubMed  Google Scholar 

  137. Aitken, R. J., Wingate, J. K., De Iuliis, G. N. & McLaughlin, E. A. Analysis of lipid peroxidation in human spermatozoa using BODIPY C11. Mol. Hum. Reprod. 13, 203–211 (2007).

    CAS  PubMed  Google Scholar 

  138. Bromfield, E. G., Aitken, R. J., Anderson, A. L., McLaughlin, E. A. & Nixon, B. The impact of oxidative stress on chaperone-mediated human sperm-egg interaction. Hum. Reprod. 30, 2597–2613 (2015).

    CAS  PubMed  Google Scholar 

  139. Walters, J. L. H. et al. Pharmacological inhibition of arachidonate 15-lipoxygenase protects human spermatozoa against oxidative stress. Biol. Reprod. 98, 784–794 (2018).

    PubMed  Google Scholar 

  140. Murphy, M. P. How mitochondria produce reactive oxygen species. Biochem. J. 417, 1–13 (2009).

    CAS  PubMed  Google Scholar 

  141. Förstermann, U. & Sessa, W. C. Nitric oxide synthases: regulation and function. Eur. Heart J. 33, 829–837 (2012). 837a–837d.

    PubMed  Google Scholar 

  142. Marnett, L. J. Oxy radicals, lipid peroxidation and DNA damage. Toxicology 181–182, 219–222 (2002).

    PubMed  Google Scholar 

  143. Aitken, R. J., Baker, M. A. & Nixon, B. Are sperm capacitation and apoptosis the opposite ends of a continuum driven by oxidative stress? Asian J. Androl. 17, 633–639 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  144. Aitken, R. J. et al. Relative impact of oxidative stress on the functional competence and genomic integrity of human spermatozoa. Biol. Reprod. 59, 1037–1046 (1998).

    CAS  PubMed  Google Scholar 

  145. Aitken, R. J. & Nixon, B. Sperm capacitation: a distant landscape glimpsed but unexplored. Mol. Hum. Reprod. 19, 785–793 (2013).

    CAS  PubMed  Google Scholar 

  146. O’Flaherty, C. M., Beorlegui, N. B. & Beconi, M. T. Reactive oxygen species requirements for bovine sperm capacitation and acrosome reaction. Theriogenology 52, 289–301 (1999).

    PubMed  Google Scholar 

  147. Leclerc, P., de Lamirande, E. & Gagnon, C. Regulation of protein-tyrosine phosphorylation and human sperm capacitation by reactive oxygen derivatives. Free Radic. Biol. Med. 22, 643–656 (1997).

    CAS  PubMed  Google Scholar 

  148. Boerke, A. et al. Involvement of bicarbonate-induced radical signaling in oxysterol formation and sterol depletion of capacitating mammalian sperm during in vitro fertilization. Biol. Reprod. 88, 21 (2013).

    PubMed  Google Scholar 

  149. Aitken, R. J. Impact of oxidative stress on male and female germ cells: implications for fertility. Reproduction 159, R189–R201 (2020).

    CAS  PubMed  Google Scholar 

  150. Otasevic, V., Stancic, A., Korac, A., Jankovic, A. & Korac, B. Reactive oxygen, nitrogen, and sulfur species in human male fertility. A crossroad of cellular signaling and pathology. Biofactors 46, 206–219 (2020).

    CAS  PubMed  Google Scholar 

  151. Carrageta, D. F. et al. Mitochondrial activation and reactive oxygen-species overproduction during sperm capacitation are independent of glucose stimuli. Antioxidants 9, 750 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  152. Noblanc, A., Kocer, A. & Drevet, J. R. Recent knowledge concerning mammalian sperm chromatin organization and its potential weaknesses when facing oxidative challenge. Basic. Clin. Androl. 24, 6 (2014).

    PubMed  PubMed Central  Google Scholar 

  153. Noblanc, A. et al. Glutathione peroxidases at work on epididymal spermatozoa: an example of the dual effect of reactive oxygen species on mammalian male fertilizing ability. J. Androl. 32, 641–650 (2011).

    CAS  PubMed  Google Scholar 

  154. Oliva, R. Protamines and male infertility. Hum. Reprod. Update 12, 417–435 (2006).

    CAS  PubMed  Google Scholar 

  155. Vertika, S., Singh, K. K. & Rajender, S. Mitochondria, spermatogenesis, and male infertility — an update. Mitochondrion 54, 26–40 (2020).

    CAS  PubMed  Google Scholar 

  156. Aitken, R. J. & Baker, M. A. Oxidative stress, sperm survival and fertility control. Mol. Cell Endocrinol. 250, 66–69 (2006).

    CAS  PubMed  Google Scholar 

  157. Marchetti, C., Obert, G., Deffosez, A., Formstecher, P. & Marchetti, P. Study of mitochondrial membrane potential, reactive oxygen species, DNA fragmentation and cell viability by flow cytometry in human sperm. Hum. Reprod. 17, 1257–1265 (2002).

    PubMed  Google Scholar 

  158. de Lamirande, E. & Gagnon, C. Reactive oxygen species and human spermatozoa. I. Effects on the motility of intact spermatozoa and on sperm axonemes. J. Androl. 13, 368–378 (1992).

    PubMed  Google Scholar 

  159. Chianese, R. & Pierantoni, R. Mitochondrial reactive oxygen species (ROS) production alters sperm quality. Antioxidants 10, 92 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  160. Aitken, R. J. et al. Electrophilic aldehydes generated by sperm metabolism activate mitochondrial reactive oxygen species generation and apoptosis by targeting succinate dehydrogenase. J. Biol. Chem. 287, 33048–33060 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  161. Lefièvre, L. et al. Human spermatozoa contain multiple targets for protein S-nitrosylation: an alternative mechanism of the modulation of sperm function by nitric oxide? Proteomics 7, 3066–3084 (2007).

    PubMed  PubMed Central  Google Scholar 

  162. O’Flaherty, C. & Matsushita-Fournier, D. Reactive oxygen species and protein modifications in spermatozoa. Biol. Reprod. 97, 577–585 (2017).

    PubMed  Google Scholar 

  163. Santiago, J., Santos, M. A. S., Fardilha, M. & Silva, J. V. Stress response pathways in the male germ cells and gametes. Mol. Hum. Reprod. 26, 1–13 (2020).

    CAS  PubMed  Google Scholar 

  164. Liu, C., Li, X. & Lu, B. The Immp2l mutation causes age-dependent degeneration of cerebellar granule neurons prevented by antioxidant treatment. Aging Cell 15, 167–176 (2016).

    CAS  PubMed  Google Scholar 

  165. George, S. K., Jiao, Y., Bishop, C. E. & Lu, B. Oxidative stress is involved in age-dependent spermatogenic damage of Immp2l mutant mice. Free Radic. Biol. Med. 52, 2223–2233 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  166. Soler, R., Füllhase, C., Lu, B., Bishop, C. E. & Andersson, K. E. Bladder dysfunction in a new mutant mouse model with increased superoxide-lack of nitric oxide? J. Urol. 183, 780–785 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  167. George, S. K., Jiao, Y., Bishop, C. E. & Lu, B. Mitochondrial peptidase IMMP2L mutation causes early onset of age-associated disorders and impairs adult stem cell self-renewal. Aging Cell 10, 584–594 (2011).

    CAS  PubMed  Google Scholar 

  168. Jiang, Y., Liu, C., Lei, B., Xu, X. & Lu, B. Mitochondria-targeted antioxidant SkQ1 improves spermatogenesis in Immp2l mutant mice. Andrologia 50, e12848 (2018).

    Google Scholar 

  169. Gianazza, E., Brioschi, M., Fernandez, A. M. & Banfi, C. Lipoxidation in cardiovascular diseases. Redox Biol. 23, 101119 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  170. Skulachev, V. P. et al. Prevention of cardiolipin oxidation and fatty acid cycling as two antioxidant mechanisms of cationic derivatives of plastoquinone (SkQs). Biochim. Biophys. Acta 1797, 878–889 (2010).

    CAS  PubMed  Google Scholar 

  171. Anisimov, V. N. et al. Effects of the mitochondria-targeted antioxidant SkQ1 on lifespan of rodents. Aging 3, 1110–1119 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  172. Kolosova, N. G., Stefanova, N. A., Muraleva, N. A. & Skulachev, V. P. The mitochondria-targeted antioxidant SkQ1 but not N-acetylcysteine reverses aging-related biomarkers in rats. Aging 4, 686–694 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  173. Skulachev, M. V. & Skulachev, V. P. Programmed aging of mammals: proof of concept and prospects of biochemical approaches for anti-aging therapy. Biochemistry 82, 1403–1422 (2017).

    CAS  PubMed  Google Scholar 

  174. Tsybul’ko, E. et al. The mitochondria-targeted plastoquinone-derivative SkQ1 promotes health and increases drosophila melanogaster longevity in various environments. J. Gerontol. A Biol. Sci. Med. Sci. 72, 499–508 (2017).

    PubMed  Google Scholar 

  175. Shabalina, I. G. et al. Improved health-span and lifespan in mtDNA mutator mice treated with the mitochondrially targeted antioxidant SkQ1. Aging 9, 315–339 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  176. Kurutas, E. B. The importance of antioxidants which play the role in cellular response against oxidative/nitrosative stress: current state. Nutr. J. 15, 71 (2016).

    PubMed  PubMed Central  Google Scholar 

  177. Cooper, T. G., Yeung, C.-H., Fetic, S., Sobhani, A. & Nieschlag, E. Cytoplasmic droplets are normal structures of human sperm but are not well preserved by routine procedures for assessing sperm morphology. Hum. Reprod. 19, 2283–2288 (2004).

    PubMed  Google Scholar 

  178. Fischer, M. A., Willis, J. & Zini, A. Human sperm DNA integrity: correlation with sperm cytoplasmic droplets. Urology 61, 207–211 (2003).

    PubMed  Google Scholar 

  179. Aitken, R. J. & Drevet, J. R. The importance of oxidative stress in determining the functionality of mammalian spermatozoa: a two-edged sword. Antioxidants 9, 111 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  180. Subramanian, V. et al. Seminal reactive oxygen species and total antioxidant capacity: correlations with sperm parameters and impact on male infertility. Clin. Exp. Reprod. Med. 45, 88–93 (2018).

    PubMed  PubMed Central  Google Scholar 

  181. Gaschler, M. M. & Stockwell, B. R. Lipid peroxidation in cell death. Biochem. Biophys. Res. Commun. 482, 419–425 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  182. Walters, J. L. H., Gadella, B. M., Sutherland, J. M., Nixon, B. & Bromfield, E. G. Male infertility: shining a light on lipids and lipid-modulating enzymes in the male germline. J. Clin. Med. 9, 372 (2020).

    Google Scholar 

  183. Ayala, A., Muñoz, M. F. & Argüelles, S. Lipid peroxidation: production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal. Oxid. Med. Cell Longev. 2014, 360438 (2014).

    PubMed  PubMed Central  Google Scholar 

  184. Chng, C. P., Sadovsky, Y., Hsia, K. J. & Huang, C. Site-specific peroxidation modulates lipid bilayer mechanics. Extrem. Mech. Lett. 42, 101148 (2021).

    Google Scholar 

  185. Balogh, E. et al. Oxidative stress impairs energy metabolism in primary cells and synovial tissue of patients with rheumatoid arthritis. Arthritis Res. Ther. 20, 95 (2018).

    PubMed  PubMed Central  Google Scholar 

  186. Camaré, C. et al. 4-Hydroxynonenal contributes to angiogenesis through a redox-dependent sphingolipid pathway: prevention by hydralazine derivatives. Oxid. Med. Cell Longev. 2017, 9172741 (2017).

    PubMed  PubMed Central  Google Scholar 

  187. Diana, T. et al. Stimulatory TSH-receptor antibodies and oxidative stress in Graves disease. J. Clin. Endocrinol. Metab. 103, 3668–3677 (2018).

    PubMed  PubMed Central  Google Scholar 

  188. Gęgotek, A., Domingues, P., Wroński, A., Wójcik, P. & Skrzydlewska, E. Proteomic plasma profile of psoriatic patients. J. Pharm. Biomed. Anal. 155, 185–193 (2018).

    PubMed  Google Scholar 

  189. Guéraud, F. 4-Hydroxynonenal metabolites and adducts in pre-carcinogenic conditions and cancer. Free Radic. Biol. Med. 111, 196–208 (2017).

    PubMed  Google Scholar 

  190. Martín-Sierra, C., Laranjeira, P., Domingues, M. R. & Paiva, A. Lipoxidation and cancer immunity. Redox Biol. 23, 101103 (2019).

    PubMed  PubMed Central  Google Scholar 

  191. Pillon, N. J. et al. The lipid peroxidation by-product 4-hydroxy-2-nonenal (4-HNE) induces insulin resistance in skeletal muscle through both carbonyl and oxidative stress. Endocrinology 153, 2099–2111 (2012).

    CAS  PubMed  Google Scholar 

  192. Soulage, C. O. et al. Two toxic lipid aldehydes, 4-hydroxy-2-hexenal (4-HHE) and 4-hydroxy-2-nonenal (4-HNE), accumulate in patients with chronic kidney disease. Toxins 12, 567 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  193. Tanaka, M. & Vécsei, L. Monitoring the redox status in multiple sclerosis. Biomedicines 8, 406 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  194. Hwang, H. V. et al. Transcriptomic and functional analyses of mitochondrial dysfunction in pressure overload-induced right ventricular failure. J. Am. Heart Assoc. 10, e017835 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  195. Bromfield, E. G. et al. Differential cell death decisions in the testis: evidence for an exclusive window of ferroptosis in round spermatids. Mol. Hum. Reprod. 25, 241–256 (2019).

    CAS  PubMed  Google Scholar 

  196. Mihalas, B. P. et al. Oxidative damage in naturally aged mouse oocytes is exacerbated by dysregulation of proteasomal activity. J. Biol. Chem. 293, 18944–18964 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  197. Mayorga-Torres, B. J. M., Camargo, M., Cadavid, Á. P., du Plessis, S. S. & Cardona Maya, W. D. Are oxidative stress markers associated with unexplained male infertility? Andrologia 49, e12659 (2017).

    Google Scholar 

  198. Turner, K. A. et al. Male infertility is a women’s health issue — research and clinical evaluation of male infertility is needed. Cells 9, 990 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  199. Walters, J. L. H. et al. Mechanistic insight into the regulation of lipoxygenase-driven lipid peroxidation events in human spermatozoa and their impact on male fertility. Antioxidants 10, 43 (2020).

    PubMed  PubMed Central  Google Scholar 

  200. Bisht, S., Faiq, M., Tolahunase, M. & Dada, R. Oxidative stress and male infertility. Nat. Rev. Urol. 14, 470–485 (2017).

    CAS  PubMed  Google Scholar 

  201. Moazamian, R. et al. Oxidative stress and human spermatozoa: diagnostic and functional significance of aldehydes generated as a result of lipid peroxidation. Mol. Hum. Reprod. 21, 502–515 (2015).

    CAS  PubMed  Google Scholar 

  202. Takeshima, T. et al. Oxidative stress and male infertility. Reprod. Med. Biol. 20, 41–52 (2021).

    CAS  PubMed  Google Scholar 

  203. Aksoy, Y., Aksoy, H., Altinkaynak, K., Aydin, H. R. & Ozkan, A. Sperm fatty acid composition in subfertile men. Prostaglandins Leukot. Essent. Fatty Acids 75, 75–79 (2006).

    CAS  PubMed  Google Scholar 

  204. Safarinejad, M. R., Hosseini, S. Y., Dadkhah, F. & Asgari, M. A. Relationship of omega-3 and omega-6 fatty acids with semen characteristics, and anti-oxidant status of seminal plasma: a comparison between fertile and infertile men. Clin. Nutr. 29, 100–105 (2010).

    CAS  PubMed  Google Scholar 

  205. Koppers, A. J., Garg, M. L. & Aitken, R. J. Stimulation of mitochondrial reactive oxygen species production by unesterified, unsaturated fatty acids in defective human spermatozoa. Free Radic. Biol. Med. 48, 112–119 (2010).

    CAS  PubMed  Google Scholar 

  206. Das, U. N. Folic acid and polyunsaturated fatty acids improve cognitive function and prevent depression, dementia, and Alzheimer’s disease-but how and why? Prostaglandins Leukot. Essent. Fatty Acids 78, 11–19 (2008).

    CAS  PubMed  Google Scholar 

  207. Guilbault, C. et al. Cystic fibrosis fatty acid imbalance is linked to ceramide deficiency and corrected by fenretinide. Am. J. Respir. Cell Mol. Biol. 41, 100–106 (2009).

    CAS  PubMed  Google Scholar 

  208. Kiziler, A. R. et al. Comparison of before and after varicocelectomy levels of trace elements, nitric oxide, asymmetric dimethylarginine and malondialdehyde in the seminal plasma and peripheral and spermatic veins. Biol. Trace Elem. Res. 167, 172–178 (2015).

    CAS  PubMed  Google Scholar 

  209. Benedetti, S. et al. Differences in blood and semen oxidative status in fertile and infertile men, and their relationship with sperm quality. Reprod. Biomed. Online 25, 300–306 (2012).

    CAS  PubMed  Google Scholar 

  210. Shamsi, M. B. et al. Antioxidant levels in blood and seminal plasma and their impact on sperm parameters in infertile men. Indian. J. Biochem. Biophys. 47, 38–43 (2010).

    CAS  PubMed  Google Scholar 

  211. Kazakov, Y., Khodos, M., Vidrevich, M. & Brainina, K. Potentiometry as a tool for monitoring of antioxidant activity and oxidative stress estimation in medicine. Crit. Rev. Anal. Chem. 49, 150–159 (2019).

    CAS  PubMed  Google Scholar 

  212. Ursini, F. & Maiorino, M. Lipid peroxidation and ferroptosis: the role of GSH and GPx4. Free Radic. Biol. Med. 152, 175–185 (2020).

    CAS  PubMed  Google Scholar 

  213. Hajeyah, A. A., Griffiths, W. J., Wang, Y., Finch, A. J. & O’Donnell, V. B. The biosynthesis of enzymatically oxidized lipids. Front. Endocrinol. 11, 591819 (2020).

    Google Scholar 

  214. Perrotta, I. et al. Expression of cyclooxygenase-1 (COX-1) and COX-2 in human male gametes from normal patients, and those with varicocele and diabetes: a potential molecular marker for diagnosing male infertility disorders. J. Anat. 221, 209–220 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  215. Matzkin, M. E. et al. Cyclooxygenase-2 in testes of infertile men: evidence for the induction of prostaglandin synthesis by interleukin-1β. Fertil. Steril. 94, 1933–1936 (2010).

    CAS  PubMed  Google Scholar 

  216. Nixon, B. J. et al. Mouse spermatocytes express CYP2E1 and respond to acrylamide exposure. PLoS ONE 9, e94904 (2014).

    PubMed  PubMed Central  Google Scholar 

  217. Lu, J. et al. New insights of CYP1A in endogenous metabolism: a focus on single nucleotide polymorphisms and diseases. Acta Pharm. Sin. B 10, 91–104 (2020).

    CAS  PubMed  Google Scholar 

  218. Brütsch, S. H. et al. Male subfertility induced by heterozygous expression of catalytically inactive glutathione peroxidase 4 is rescued in vivo by systemic inactivation of the Alox15 gene. J. Biol. Chem. 291, 23578–23588 (2016).

    PubMed  PubMed Central  Google Scholar 

  219. Bromfield, E. G. et al. Inhibition of arachidonate 15-lipoxygenase prevents 4-hydroxynonenal-induced protein damage in male germ cells. Biol. Reprod. 96, 598–609 (2017).

    PubMed  Google Scholar 

  220. Pizzimenti, S. et al. Interaction of aldehydes derived from lipid peroxidation and membrane proteins. Front. Physiol. 4, 242 (2013).

    PubMed  PubMed Central  Google Scholar 

  221. Cafe, S. L. et al. Oxidative stress dysregulates protein homeostasis within the male germ line. Antioxid. Redox Signal. 32, 487–503 (2020).

    CAS  PubMed  Google Scholar 

  222. Uchida, K. 4-Hydroxy-2-nonenal: a product and mediator of oxidative stress. Prog. Lipid Res. 42, 318–343 (2003).

    CAS  PubMed  Google Scholar 

  223. Baker, M. A. et al. Defining the mechanisms by which the reactive oxygen species by-product, 4-hydroxynonenal, affects human sperm cell function. Biol. Reprod. 92, 108 (2015).

    PubMed  Google Scholar 

  224. Nixon, B. et al. A kinase anchor protein 4 is vulnerable to oxidative adduction in male germ cells. Front. Cell Dev. Biol. 7, 319 (2019).

    PubMed  PubMed Central  Google Scholar 

  225. Koppers, A. J., De Iuliis, G. N., Finnie, J. M., McLaughlin, E. A. & Aitken, R. J. Significance of mitochondrial reactive oxygen species in the generation of oxidative stress in spermatozoa. J. Clin. Endocrinol. Metab. 93, 3199–3207 (2008).

    CAS  PubMed  Google Scholar 

  226. Aitken, R. J. & De Iuliis, G. N. On the possible origins of DNA damage in human spermatozoa. Mol. Hum. Reprod. 16, 3–13 (2010).

    CAS  PubMed  Google Scholar 

  227. Mitchell, L. A., De Iuliis, G. N. & Aitken, R. J. The TUNEL assay consistently underestimates DNA damage in human spermatozoa and is influenced by DNA compaction and cell vitality: development of an improved methodology. Int. J. Androl. 34, 2–13 (2011).

    CAS  PubMed  Google Scholar 

  228. Espinoza, J. A., Schulz, M. A., Sánchez, R. & Villegas, J. V. Integrity of mitochondrial membrane potential reflects human sperm quality. Andrologia 41, 51–54 (2009).

    CAS  PubMed  Google Scholar 

  229. Aitken, R. J., Findlay, J. K., Hutt, K. J. & Kerr, J. B. Apoptosis in the germ line. Reprod 141, 139–150 (2011).

    CAS  Google Scholar 

  230. Lord, T., Martin, J. H. & Aitken, R. J. Accumulation of electrophilic aldehydes during postovulatory aging of mouse oocytes causes reduced fertility, oxidative stress, and apoptosis. Biol. Reprod. 92, 33 (2015).

    PubMed  Google Scholar 

  231. Redgrove, K. A. et al. The molecular chaperone HSPA2 plays a key role in regulating the expression of sperm surface receptors that mediate sperm-egg recognition. PLoS ONE 7, e50851 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  232. Bromfield, E. G., Aitken, R. J., McLaughlin, E. A. & Nixon, B. Proteolytic degradation of heat shock protein A2 occurs in response to oxidative stress in male germ cells of the mouse. Mol. Hum. Reprod. 23, 91–105 (2017).

    CAS  PubMed  Google Scholar 

  233. Maniti, O. et al. Protein “amyloid-like” networks at the phospholipid membrane formed by 4-hydroxy-2-nonenal-modified mitochondrial creatine kinase. Mol. Membr. Biol. 32, 1–10 (2015).

    CAS  PubMed  Google Scholar 

  234. Li, D. & Li, Y. The interaction between ferroptosis and lipid metabolism in cancer. Signal. Transduct. Target. Ther. 5, 108 (2020).

    PubMed  PubMed Central  Google Scholar 

  235. Dixon, S. J. et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell 149, 1060–1072 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  236. Hirschhorn, T. & Stockwell, B. R. The development of the concept of ferroptosis. Free. Radic. Biol. Med. 133, 130–143 (2019).

    CAS  PubMed  Google Scholar 

  237. Stockwell, B. R., Jiang, X. & Gu, W. Emerging mechanisms and disease relevance of ferroptosis. Trends Cell Biol. 30, 478–490 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  238. Doll, S. et al. ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat. Chem. Biol. 13, 91–98 (2017).

    CAS  PubMed  Google Scholar 

  239. Zhang, Q. et al. The structural basis for the phospholipid remodeling by lysophosphatidylcholine acyltransferase 3. Nat. Commun. 12, 6869 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  240. Kagan, V. E. et al. Oxidized arachidonic and adrenic PEs navigate cells to ferroptosis. Nat. Chem. Biol. 13, 81–90 (2017).

    CAS  PubMed  Google Scholar 

  241. Wenzel, S. E. et al. PEBP1 wardens ferroptosis by enabling lipoxygenase generation of lipid death signals. Cell 171, 628–641.e26 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  242. Chu, B. et al. ALOX12 is required for p53-mediated tumour suppression through a distinct ferroptosis pathway. Nat. Cell Biol. 21, 579–591 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  243. Zhao, X. et al. Inhibition of ferroptosis attenuates busulfan-induced oligospermia in mice. Toxicology 440, 152489 (2020).

    CAS  PubMed  Google Scholar 

  244. Meng, P. et al. Arsenite induces testicular oxidative stress in vivo and in vitro leading to ferroptosis. Ecotoxicol. Environ. Saf. 194, 110360 (2020).

    CAS  PubMed  Google Scholar 

  245. Ou, Z. et al. Cigarette smoking is associated with high level of ferroptosis in seminal plasma and affects semen quality. Reprod. Biol. Endocrinol. 18, 55 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  246. Li, L. et al. Ferroptosis is associated with oxygen-glucose deprivation/reoxygenation-induced Sertoli cell death. Int. J. Mol. Med. 41, 3051–3062 (2018).

    CAS  PubMed  Google Scholar 

  247. Newcomer, M. E. & Brash, A. R. The structural basis for specificity in lipoxygenase catalysis. Protein Sci. 24, 298–309 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  248. Ivanov, I., Kuhn, H. & Heydeck, D. Structural and functional biology of arachidonic acid 15-lipoxygenase-1 (ALOX15). Gene 573, 1–32 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  249. Piñero, J. et al. The DisGeNET knowledge platform for disease genomics: 2019 update. Nucleic Acids Res. 48, D845–D855 (2019).

    PubMed Central  Google Scholar 

  250. Dobrian, A. D. et al. Functional and pathological roles of the 12- and 15-lipoxygenases. Prog. Lipid Res. 50, 115–131 (2011).

    CAS  PubMed  Google Scholar 

  251. Martínez-Clemente, M. et al. Disruption of the 12/15-lipoxygenase gene (Alox15) protects hyperlipidemic mice from nonalcoholic fatty liver disease. Hepatology 52, 1980–1991 (2010).

    PubMed  Google Scholar 

  252. Singh, N. K. & Rao, G. N. Emerging role of 12/15-Lipoxygenase (ALOX15) in human pathologies. Prog. Lipid Res. 73, 28–45 (2019).

    CAS  PubMed  Google Scholar 

  253. Weinstock-Guttman, B. et al. Serum lipid profiles are associated with disability and MRI outcomes in multiple sclerosis. J. Neuroinflammation 8, 127 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  254. Zhang, B. et al. Depletion of regulatory T cells facilitates growth of established tumors: a mechanism involving the regulation of myeloid-derived suppressor cells by lipoxin A4. J. Immunol. 185, 7199–7206 (2010).

    CAS  PubMed  Google Scholar 

  255. Kutzner, L. et al. Mammalian ALOX15 orthologs exhibit pronounced dual positional specificity with docosahexaenoic acid. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 1862, 666–675 (2017).

    CAS  PubMed  Google Scholar 

  256. Wisastra, R. & Dekker, F. J. Inflammation, cancer and oxidative lipoxygenase activity are intimately linked. Cancers 6, 1500–1521 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  257. Walters, J. L. H., De Iuliis, G. N., Nixon, B. & Bromfield, E. G. Oxidative stress in the male germline: a review of novel strategies to reduce 4-hydroxynonenal production. Antioxidants 7, 132 (2018).

    PubMed  PubMed Central  Google Scholar 

  258. Brash, A. R. Lipoxygenases: occurrence, functions, catalysis, and acquisition of substrate. J. Biol. Chem. 274, 23679–23682 (1999).

    CAS  PubMed  Google Scholar 

  259. Ivanov, I. et al. Molecular enzymology of lipoxygenases. Arch. Biochem. Biophys. 503, 161–174 (2010).

    CAS  PubMed  Google Scholar 

  260. Fischer, K. A. et al. 15-Lipoxygenase is a component of the mammalian sperm cytoplasmic droplet. Reproduction 130, 213–222 (2005).

    CAS  PubMed  Google Scholar 

  261. Moore, K. et al. Altered epididymal sperm maturation and cytoplasmic droplet migration in subfertile male Alox15 mice. Cell Tissue Res. 340, 569–581 (2010).

    PubMed  Google Scholar 

  262. Lax, Y., Grossman, S., Rubinstein, S., Magid, N. & Breitbart, H. Role of lipoxygenase in the mechanism of acrosome reaction in mammalian spermatozoa. Biochim. Biophys. Acta 1043, 12–18 (1990).

    CAS  PubMed  Google Scholar 

  263. Li, K. P., Yang, X. S. & Wu, T. The effect of antioxidants on sperm quality parameters and pregnancy rates for idiopathic male infertility: a network meta-analysis of randomized controlled trials. Front. Endocrinol. 13, 810242 (2022).

    Google Scholar 

  264. Steiner, A. Z. et al. The effect of antioxidants on male factor infertility: the males, antioxidants, and infertility (MOXI) randomized clinical trial. Fertil. Steril. 113, 552–560.e553 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  265. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/study/NCT04256278 (2020).

  266. Aitken, R. J. Antioxidant trials-the need to test for stress. Hum. Reprod. Open. 2021, hoab007 (2021).

    PubMed  PubMed Central  Google Scholar 

  267. Kayama, Y. et al. Cardiac 12/15 lipoxygenase-induced inflammation is involved in heart failure. J. Exp. Med. 206, 1565–1574 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  268. Higgs, G. A., Salmon, J. A. & Spayne, J. A. The inflammatory effects of hydroperoxy and hydroxy acid products of arachidonate lipoxygenase in rabbit skin. Br. J. Pharmacol. 74, 429–433 (1981).

    CAS  PubMed  PubMed Central  Google Scholar 

  269. Kühn, H. & O’Donnell, V. B. Inflammation and immune regulation by 12/15-lipoxygenases. Prog. Lipid Res. 45, 334–356 (2006).

    PubMed  Google Scholar 

  270. Cole, B. K., Lieb, D. C., Dobrian, A. D. & Nadler, J. L. 12- and 15-lipoxygenases in adipose tissue inflammation. Prostaglandins Other Lipid Mediat. 104-105, 84–92 (2013).

    CAS  PubMed  Google Scholar 

  271. Chakrabarti, S. K., Cole, B. K., Wen, Y., Keller, S. R. & Nadler, J. L. 12/15-lipoxygenase products induce inflammation and impair insulin signaling in 3T3-L1 adipocytes. Obesity 17, 1657–1663 (2009).

    CAS  PubMed  Google Scholar 

  272. Anderson, S. D. & Kippelen, P. Airway injury as a mechanism for exercise-induced bronchoconstriction in elite athletes. J. Allergy Clin. Immunol. 122, 225–235 (2008).

    PubMed  Google Scholar 

  273. Xu, X., Li, J., Zhang, Y. & Zhang, L. Arachidonic acid 15-lipoxygenase: effects of its expression, metabolites, and genetic and epigenetic variations on airway inflammation. Allergy Asthma Immunol. Res. 13, 684–696 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  274. Feltenmark, S. et al. Eoxins are proinflammatory arachidonic acid metabolites produced via the 15-lipoxygenase-1 pathway in human eosinophils and mast cells. Proc. Natl Acad. Sci. USA 105, 680–685 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  275. Serhan, C. N. Pro-resolving lipid mediators are leads for resolution physiology. Nature 510, 92–101 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  276. Kühn, H. Biosynthesis, metabolization and biological importance of the primary 15-lipoxygenase metabolites 15-hydro(pero)XY-5Z,8Z,11Z,13E-eicosatetraenoic acid and 13-hydro(pero)XY-9Z,11E-octadecadienoic acid. Prog. Lipid Res. 35, 203–226 (1996).

    PubMed  Google Scholar 

  277. Ziboh, V. A. The significance of polyunsaturated fatty acids in cutaneous biology. Lipids 31, S249–S253 (1996).

    CAS  PubMed  Google Scholar 

  278. Huang, J. T. et al. Interleukin-4-dependent production of PPAR-gamma ligands in macrophages by 12/15-lipoxygenase. Nature 400, 378–382 (1999).

    CAS  PubMed  Google Scholar 

  279. Sun, L. et al. 12/15-Lipoxygenase metabolites of arachidonic acid activate PPARγ: a possible neuroprotective effect in ischemic brain. J. Lipid Res. 56, 502–514 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  280. McMahon, B. & Godson, C. Lipoxins: endogenous regulators of inflammation. Am. J. Physiol. Renal Physiol. 286, F189–F201 (2004).

    CAS  PubMed  Google Scholar 

  281. Serhan, C. N., Hamberg, M. & Samuelsson, B. Lipoxins: novel series of biologically active compounds formed from arachidonic acid in human leukocytes. Proc. Natl Acad. Sci. USA 81, 5335–5339 (1984).

    CAS  PubMed  PubMed Central  Google Scholar 

  282. Krönke, G. et al. 12/15-Lipoxygenase counteracts inflammation and tissue damage in arthritis. J. Immunol. 183, 3383–3389 (2009).

    PubMed  Google Scholar 

  283. Marcheselli, V. L. et al. Novel docosanoids inhibit brain ischemia-reperfusion-mediated leukocyte infiltration and pro-inflammatory gene expression. J. Biol. Chem. 278, 43807–43817 (2003).

    CAS  PubMed  Google Scholar 

  284. Takano, T., Clish, C. B., Gronert, K., Petasis, N. & Serhan, C. N. Neutrophil-mediated changes in vascular permeability are inhibited by topical application of aspirin-triggered 15-epi-lipoxin A4 and novel lipoxin B4 stable analogues. J. Clin. Invest. 101, 819–826 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  285. Pakiet, A. et al. The effect of a high-fat diet on the fatty acid composition in the hearts of mice. Nutrients 12, 824 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  286. Saini, V. Molecular mechanisms of insulin resistance in type 2 diabetes mellitus. World J. Diabetes 1, 68–75 (2010).

    PubMed  PubMed Central  Google Scholar 

  287. Shetty, S. S. & Kumari, S. Fatty acids and their role in type-2 diabetes (Review). Exp. Ther. Med. 22, 706–706 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  288. Ferré, N. et al. Increased susceptibility to exacerbated liver injury in hypercholesterolemic ApoE-deficient mice: potential involvement of oxysterols. Am. J. Physiol. Gastrointest. Liver Physiol. 296, G553–G562 (2009).

    PubMed  Google Scholar 

  289. Chakrabarti, S. K. et al. Evidence for activation of inflammatory lipoxygenase pathways in visceral adipose tissue of obese Zucker rats. Am. J. Physiol. Endocrinol. Metab. 300, E175–E187 (2011).

    CAS  PubMed  Google Scholar 

  290. Obrosova, I. G. et al. High-fat diet induced neuropathy of pre-diabetes and obesity: effects of “healthy” diet and aldose reductase inhibition. Diabetes 56, 2598–2608 (2007).

    CAS  PubMed  Google Scholar 

  291. Nunemaker, C. S. et al. 12-Lipoxygenase-knockout mice are resistant to inflammatory effects of obesity induced by Western diet. Am. J. Physiol. Endocrinol. Metab. 295, E1065–E1075 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  292. Schnurr, K., Belkner, J., Ursini, F., Schewe, T. & Kühn, H. The selenoenzyme phospholipid hydroperoxide glutathione peroxidase controls the activity of the 15-lipoxygenase with complex substrates and preserves the specificity of the oxygenation products. J. Biol. Chem. 271, 4653–4658 (1996).

    CAS  PubMed  Google Scholar 

  293. Heydeck, D. et al. Interleukin-4 and -13 induce upregulation of the murine macrophage 12/15-lipoxygenase activity: evidence for the involvement of transcription factor STAT6. Blood 92, 2503–2510 (1998).

    CAS  PubMed  Google Scholar 

  294. Dobrian, A. D. et al. Differential expression and localization of 12/15 lipoxygenases in adipose tissue in human obese subjects. Biochem. Biophys. Res. Commun. 403, 485–490 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  295. Zhang, H. J. et al. 12S-hydroxyeicosatetraenoic acid levels link to coronary artery disease in Type 2 diabetic patients. J. Endocrinol. Invest. 36, 385–389 (2013).

    CAS  PubMed  Google Scholar 

  296. Ma, K. et al. 12-Lipoxygenase products reduce insulin secretion and β-cell viability in human islets. J. Clin. Endocrinol. Metab. 95, 887–893 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  297. Tersey, S. A. et al. 12-lipoxygenase promotes obesity-induced oxidative stress in pancreatic islets. Mol. Cell Biol. 34, 3735–3745 (2014).

    PubMed  PubMed Central  Google Scholar 

  298. McDuffie, M. et al. Nonobese diabetic (NOD) mice congenic for a targeted deletion of 12/15-lipoxygenase are protected from autoimmune diabetes. Diabetes 57, 199–208 (2008).

    CAS  PubMed  Google Scholar 

  299. Panth, N., Paudel, K. R. & Parajuli, K. Reactive oxygen species: a key hallmark of cardiovascular disease. Adv. Med. 2016, 9152732–9152732 (2016).

    PubMed  PubMed Central  Google Scholar 

  300. Mezentsev, A. et al. Eicosanoid regulation of vascular endothelial growth factor expression and angiogenesis in microvessel endothelial cells. J. Biol. Chem. 277, 18670–18676 (2002).

    CAS  PubMed  Google Scholar 

  301. Pfister, S. L. & Campbell, W. B. Arachidonic acid- and acetylcholine-induced relaxations of rabbit aorta. Hypertension 20, 682–689 (1992).

    CAS  PubMed  Google Scholar 

  302. Pfister, S. L. et al. Identification of the 11,14,15- and 11,12, 15-trihydroxyeicosatrienoic acids as endothelium-derived relaxing factors of rabbit aorta. J. Biol. Chem. 273, 30879–30887 (1998).

    CAS  PubMed  Google Scholar 

  303. Rubbo, H. & O’Donnell, V. Nitric oxide, peroxynitrite and lipoxygenase in atherogenesis: mechanistic insights. Toxicology 208, 305–317 (2005).

    CAS  PubMed  Google Scholar 

  304. Srivastava, K., Kundumani-Sridharan, V., Zhang, B., Bajpai, A. K. & Rao, G. N. 15(S)-hydroxyeicosatetraenoic acid-induced angiogenesis requires STAT3-dependent expression of VEGF. Cancer Res. 67, 4328–4336 (2007).

    CAS  PubMed  Google Scholar 

  305. Zhang, B., Cao, H. & Rao, G. N. 15(S)-hydroxyeicosatetraenoic acid induces angiogenesis via activation of PI3K-Akt-mTOR-S6K1 signaling. Cancer Res. 65, 7283–7291 (2005).

    CAS  PubMed  Google Scholar 

  306. Rosolowsky, M. & Campbell, W. B. Role of PGI2 and epoxyeicosatrienoic acids in relaxation of bovine coronary arteries to arachidonic acid. Am. J. Physiol. 264, H327–H335 (1993).

    CAS  PubMed  Google Scholar 

  307. Kriska, T. et al. Mice lacking macrophage 12/15-lipoxygenase are resistant to experimental hypertension. Am. J. Physiol. Heart Circ. Physiol. 302, H2428–H2438 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  308. Sigal, E., Dicharry, S., Highland, E. & Finkbeiner, W. E. Cloning of human airway 15-lipoxygenase: identity to the reticulocyte enzyme and expression in epithelium. Am. J. Physiol. 262, L392–L398 (1992).

    CAS  PubMed  Google Scholar 

  309. Sigal, E. et al. Arachidonate 15-lipoxygenase from human eosinophil-enriched leukocytes: partial purification and properties. Biochem. Biophys. Res. Commun. 150, 376–383 (1988).

    CAS  PubMed  Google Scholar 

  310. Lam, G. Y., Huang, J. & Brumell, J. H. The many roles of NOX2 NADPH oxidase-derived ROS in immunity. Semin. Immunopathol. 32, 415–430 (2010).

    CAS  PubMed  Google Scholar 

  311. Tavassolifar, M. J., Vodjgani, M., Salehi, Z. & Izad, M. The influence of reactive oxygen species in the immune system and pathogenesis of multiple sclerosis. Autoimmune Dis. 2020, 5793817–5793817 (2020).

    PubMed  PubMed Central  Google Scholar 

  312. Di Dalmazi, G., Hirshberg, J., Lyle, D., Freij, J. B. & Caturegli, P. Reactive oxygen species in organ-specific autoimmunity. Auto. Immun. Highlights 7, 11 (2016).

    PubMed  PubMed Central  Google Scholar 

  313. van Horssen, J., Witte, M. E., Schreibelt, G. & de Vries, H. E. Radical changes in multiple sclerosis pathogenesis. Biochim. Biophys. Acta 1812, 141–150 (2011).

    PubMed  Google Scholar 

  314. Xu, J. et al. Inhibition of 12/15-lipoxygenase by baicalein induces microglia PPARβ/δ: a potential therapeutic role for CNS autoimmune disease. Cell Death Dis. 4, e569 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  315. Emerson, M. R. & LeVine, S. M. Experimental allergic encephalomyelitis is exacerbated in mice deficient for 12/15-lipoxygenase or 5-lipoxygenase. Brain Res. 1021, 140–145 (2004).

    CAS  PubMed  Google Scholar 

  316. Wang, H. et al. 12-Lipoxygenase plays a key role in cell death caused by glutathione depletion and arachidonic acid in rat oligodendrocytes. Eur. J. Neurosci. 20, 2049–2058 (2004).

    PubMed  Google Scholar 

  317. Yao, Y., Clark, C. M., Trojanowski, J. Q., Lee, V. M. & Praticò, D. Elevation of 12/15 lipoxygenase products in AD and mild cognitive impairment. Ann. Neurol. 58, 623–626 (2005).

    CAS  PubMed  Google Scholar 

  318. Coussens, L. M. & Werb, Z. Inflammation and cancer. Nature 420, 860–867 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  319. Chiang, A. C. & Massagué, J. Molecular basis of metastasis. N. Engl. J. Med. 359, 2814–2823 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  320. Facchini, G. et al. Exploring the molecular aspects associated with testicular germ cell tumors: a review. Oncotarget 9, 1365–1379 (2017).

    PubMed  PubMed Central  Google Scholar 

  321. Fares, J., Fares, M. Y., Khachfe, H. H., Salhab, H. A. & Fares, Y. Molecular principles of metastasis: a hallmark of cancer revisited. Signal Transduct. Target. Ther. 5, 28 (2020).

    PubMed  PubMed Central  Google Scholar 

  322. Klil-Drori, A. J. & Ariel, A. 15-Lipoxygenases in cancer: a double-edged sword? Prostaglandins Other Lipid Mediat. 106, 16–22 (2013).

    CAS  PubMed  Google Scholar 

  323. Schneider, M. et al. Absence of glutathione peroxidase 4 affects tumor angiogenesis through increased 12/15-lipoxygenase activity. Neoplasia 12, 254–263 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  324. Seiler, A. et al. Glutathione peroxidase 4 senses and translates oxidative stress into 12/15-lipoxygenase dependent- and AIF-mediated cell death. Cell Metab. 8, 237–248 (2008).

    CAS  PubMed  Google Scholar 

  325. Yoshinaga, M., Buchanan, F. G. & DuBois, R. N. 15-LOX-1 inhibits p21 (Cip/WAF 1) expression by enhancing MEK-ERK 1/2 signaling in colon carcinoma cells. Prostaglandins Other Lipid Mediat. 73, 111–122 (2004).

    CAS  PubMed  Google Scholar 

  326. Cimen, I., Tunçay, S. & Banerjee, S. 15-Lipoxygenase-1 expression suppresses the invasive properties of colorectal carcinoma cell lines HCT-116 and HT-29. Cancer Sci. 100, 2283–2291 (2009).

    CAS  PubMed  Google Scholar 

  327. Cimen, I., Astarci, E. & Banerjee, S. 15-lipoxygenase-1 exerts its tumor suppressive role by inhibiting nuclear factor-kappa B via activation of PPAR gamma. J. Cell Biochem. 112, 2490–2501 (2011).

    CAS  PubMed  Google Scholar 

  328. Bürger, F., Krieg, P., Marks, F. & Fürstenberger, G. Positional- and stereo-selectivity of fatty acid oxygenation catalysed by mouse (12S)-lipoxygenase isoenzymes. Biochem. J. 348 (Pt 2), 329–335 (2000).

    PubMed  PubMed Central  Google Scholar 

  329. Tang, D. G. et al. 12(S)-HETE is a mitogenic factor for microvascular endothelial cells: its potential role in angiogenesis. Biochem. Biophys. Res. Commun. 211, 462–468 (1995).

    CAS  PubMed  Google Scholar 

  330. Janakiram, N. B., Mohammed, A. & Rao, C. V. Role of lipoxins, resolvins, and other bioactive lipids in colon and pancreatic cancer. Cancer Metastasis Rev. 30, 507–523 (2011).

    CAS  PubMed  Google Scholar 

  331. Wendel, M. & Heller, A. R. Anticancer actions of omega-3 fatty acids — current state and future perspectives. Anticancer Agents Med. Chem. 9, 457–470 (2009).

    CAS  PubMed  Google Scholar 

  332. Mangino, M. J., Brounts, L., Harms, B. & Heise, C. Lipoxin biosynthesis in inflammatory bowel disease. Prostaglandins Other Lipid Mediat. 79, 84–92 (2006).

    CAS  PubMed  Google Scholar 

  333. Kelavkar, U., Cohen, C., Eling, T. & Badr, K. 15-lipoxygenase-1 overexpression in prostate adenocarcinoma. Adv. Exp. Med. Biol. 507, 133–145 (2002).

    CAS  PubMed  Google Scholar 

  334. Kelavkar, U. P., Cohen, C., Kamitani, H., Eling, T. E. & Badr, K. F. Concordant induction of 15-lipoxygenase-1 and mutant p53 expression in human prostate adenocarcinoma: correlation with Gleason staging. Carcinogenesis 21, 1777–1787 (2000).

    CAS  PubMed  Google Scholar 

  335. Kelavkar, U. P., Parwani, A. V., Shappell, S. B. & Martin, W. D. Conditional expression of human 15-lipoxygenase-1 in mouse prostate induces prostatic intraepithelial neoplasia: the FLiMP mouse model. Neoplasia 8, 510–522 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  336. Harats, D. et al. Inhibition of carcinogenesis in transgenic mouse models over-expressing 15-lipoxygenase in the vascular wall under the control of murine preproendothelin-1 promoter. Cancer Lett. 229, 127–134 (2005).

    CAS  PubMed  Google Scholar 

  337. Germolec, D. R., Shipkowski, K. A., Frawley, R. P. & Evans, E. Markers of inflammation. Methods Mol. Biol. 1803, 57–79 (2018).

    CAS  PubMed  Google Scholar 

  338. Chung, H. Y. et al. Molecular inflammation: underpinnings of aging and age-related diseases. Ageing Res. Rev. 8, 18–30 (2009).

    CAS  PubMed  Google Scholar 

  339. Shah, M. S. & Brownlee, M. Molecular and cellular mechanisms of cardiovascular disorders in diabetes. Circ. Res. 118, 1808–1829 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors gratefully acknowledge the research contributions of all laboratories working on the ‘canary in the coal mine — male reproductive health’ paradigm and M. O’Bryan for bringing this issue to the attention of the Society for Reproductive Biology (SRB), Australia and New Zealand. The authors also acknowledge the work of the Male Reproductive Health Initiative of the European Society of Human Reproduction and Embryology. The authors acknowledge funding from the National Health and Medical Research Council (NHMRC), Australia, awarded to B.N., E.G.B. and R.J.A. (APP1163319).

Author information

Authors and Affiliations

Authors

Contributions

E.G.B., N.D.B. and R.J.A. researched data for the article. E.G.B., N.D.B., B.N., S.D.R., J.E.S. and J.L.H.W contributed substantially to discussion of the content. E.G.B. and N.D.B. wrote the article. All authors reviewed and edited the manuscript before submission.

Corresponding author

Correspondence to Elizabeth G. Bromfield.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Reviews Urology thanks M. Kurpisz and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Springer Nature or its licensor holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Burke, N.D., Nixon, B., Roman, S.D. et al. Male infertility and somatic health — insights into lipid damage as a mechanistic link. Nat Rev Urol 19, 727–750 (2022). https://doi.org/10.1038/s41585-022-00640-y

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41585-022-00640-y

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing