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.

  • Review Article
  • Published:

Advances in stem cell research for the treatment of primary hypogonadism

Abstract

In Leydig cell dysfunction, cells respond weakly to stimulation by pituitary luteinizing hormone, and, therefore, produce less testosterone, leading to primary hypogonadism. The most widely used treatment for primary hypogonadism is testosterone replacement therapy (TRT). However, TRT causes infertility and has been associated with other adverse effects, such as causing erythrocytosis and gynaecomastia, worsening obstructive sleep apnoea and increasing cardiovascular morbidity and mortality risks. Stem-cell-based therapy that re-establishes testosterone-producing cell lineages in the body has, therefore, become a promising prospect for treating primary hypogonadism. Over the past two decades, substantial advances have been made in the identification of Leydig cell sources for use in transplantation surgery, including the artificial induction of Leydig-like cells from different types of stem cells, for example, stem Leydig cells, mesenchymal stem cells, and pluripotent stem cells (PSCs). PSC-derived Leydig-like cells have already provided a powerful in vitro model to study the molecular mechanisms underlying Leydig cell differentiation and could be used to treat men with primary hypogonadism in a more specific and personalized approach.

Key points

  • Primary hypogonadism is mainly treated using testosterone replacement therapy (TRT). However, TRT has adverse effects and is unsuitable for men with hypogonadism wishing to maintain fertility.

  • Stem-cell-based therapy, in which a cell lineage can be re-established in human bodies to produce testosterone normally, would be the ideal choice for treating primary hypogonadism.

  • Stem Leydig cells, mesenchymal stromal cells, pluripotent stem cells and fibroblasts are newly discovered sources of Leydig cells.

  • Stem-cell-derived Leydig cells have many potential applications, including understanding the underlying mechanisms of primary hypogonadism, treating primary hypogonadism using transplantation therapy, and discovering drugs aimed at recovering Leydig cell function.

  • Research is needed in the applications of Leydig cells, including constructing 3D testicular organoids, promoting in vitro culture conditions of Leydig cells, and exploring the in vivo transplantation locations of Leydig cells.

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: The hypothalamus–pituitary–gonadal axis and pathophysiology of male hypogonadism.
Fig. 2: The development of human Leydig cells.
Fig. 3: Induction of human Leydig-like cells from different cell types.
Fig. 4: Applications of stem cell-derived, Leydig-like cells for the study and treatment of primary hypogonadism.

Similar content being viewed by others

References

  1. Zirkin, B. R. & Papadopoulos, V. Leydig cells: formation, function, and regulation. Biol. Reprod. 99, 101–111 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Cattabiani, C. et al. Relationship between testosterone deficiency and cardiovascular risk and mortality in adult men. J. Endocrinol. Invest. 35, 104–120 (2012).

    CAS  PubMed  Google Scholar 

  3. Morris, P. D. & Channer, K. S. Testosterone and cardiovascular disease in men. Asian J. Androl. 14, 428–435 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Belchetz, P. E., Barth, J. H. & Kaufman, J. M. Biochemical endocrinology of the hypogonadal male. Ann. Clin. Biochem. 47, 503–515 (2010).

    Article  CAS  PubMed  Google Scholar 

  5. Labrie, F. Blockade of testicular and adrenal androgens in prostate cancer treatment. Nat. Rev. Urol. 8, 73–85 (2011).

    Article  CAS  PubMed  Google Scholar 

  6. Salonia, A. et al. Paediatric and adult-onset male hypogonadism. Nat. Rev. Dis. Prim. 5, 38 (2019).

    Article  PubMed  Google Scholar 

  7. Patel, A. S., Leong, J. Y., Ramos, L. & Ramasamy, R. testosterone is a contraceptive and should not be used in men who desire fertility. World J. Mens. Health 37, 45–54 (2019).

    Article  PubMed  Google Scholar 

  8. Grech, A., Breck, J. & Heidelbaugh, J. Adverse effects of testosterone replacement therapy: an update on the evidence and controversy. Ther. Adv. Drug Saf. 5, 190–200 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Kimbrel, E. A. & Lanza, R. Next-generation stem cells - ushering in a new era of cell-based therapies. Nat. Rev. Drug Discov. 19, 463–479 (2020).

    Article  CAS  PubMed  Google Scholar 

  10. Zheng, C., Chen, J., Liu, S. & Jin, Y. Stem cell-based bone and dental regeneration: a view of microenvironmental modulation. Int. J. Oral. Sci. 11, 23 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Griswold, S. L. & Behringer, R. R. Fetal Leydig cell origin and development. Sex. Dev. 3, 1–15 (2009).

    Article  CAS  PubMed  Google Scholar 

  12. Martin, L. J. Cell interactions and genetic regulation that contribute to testicular Leydig cell development and differentiation. Mol. Reprod. Dev. 83, 470–487 (2016).

    Article  CAS  PubMed  Google Scholar 

  13. Rey, R., Josso, N. & Racine, C. in Endotext (eds Feingold, K. R. et al.) (MDText.com, Inc., 2000).

  14. Pelliniemi, L. J., Kuopio, T. & Fröjdman, K. in The Leydig Cell. 143-158 (Cache River Press, 1996).

  15. Voutilainen, R. Differentiation of the fetal gonad. Horm. Res. 38, 66–71 (1992).

    Article  CAS  PubMed  Google Scholar 

  16. Svechnikov, K. et al. Origin, development and regulation of human Leydig cells. Horm. Res. Paediatr. 73, 93–101 (2010).

    Article  CAS  PubMed  Google Scholar 

  17. Murray, T. J., Fowler, P. A., Abramovich, D. R., Haites, N. & Lea, R. G. Human fetal testis: second trimester proliferative and steroidogenic capacities. J. Clin. Endocrinol. Metab. 85, 4812–4817 (2000).

    CAS  PubMed  Google Scholar 

  18. Pelliniemi, L. J. & Niemi, M. Fine structure of the human foetal testis. I. The interstitial tissue. Z. Zellforsch. Mikrosk. Anat. 99, 507–522 (1969).

    Article  CAS  PubMed  Google Scholar 

  19. Haider, S. G. Cell biology of Leydig cells in the testis. Int. Rev. Cytol. 233, 181–241 (2004).

    Article  CAS  PubMed  Google Scholar 

  20. Mendis-Handagama, S. M. & Ariyaratne, H. B. Differentiation of the adult Leydig cell population in the postnatal testis. Biol. Reprod. 65, 660–671 (2001).

    Article  CAS  PubMed  Google Scholar 

  21. Shima, Y. Development of fetal and adult Leydig cells. Reprod. Med. Biol. 18, 323–330 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Wen, Q., Cheng, C. Y. & Liu, Y. X. Development, function and fate of fetal Leydig cells. Semin. Cell Dev. Biol. 59, 89–98 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Petersen, C. & Soder, O. The sertoli cell–a hormonal target and ‘super’ nurse for germ cells that determines testicular size. Horm. Res. 66, 153–161 (2006).

    CAS  PubMed  Google Scholar 

  24. Marx, J. Snaring the genes that divide the sexes for mammals. Science 269, 1824–1825 (1995).

    Article  CAS  PubMed  Google Scholar 

  25. McLaren, A. Sex determination. Makes a man. a man? Nature 346, 216–217 (1990).

    Article  CAS  PubMed  Google Scholar 

  26. McLaren, A. Sex determination. The making of male mice. Nature 351, 96 (1991).

    Article  CAS  PubMed  Google Scholar 

  27. Schimmer, B. P. & White, P. C. Minireview: steroidogenic factor 1: its roles in differentiation, development, and disease. Mol. Endocrinol. 24, 1322–1337 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Baker, P. J., Sha, J. H. & O’Shaughnessy, P. J. Localisation and regulation of 17beta-hydroxysteroid dehydrogenase type 3 mRNA during development in the mouse testis. Mol. Cell Endocrinol. 133, 127–133 (1997).

    Article  CAS  PubMed  Google Scholar 

  29. O’Shaughnessy, P. J., Baker, P. J. & Johnston, H. The foetal Leydig cell– differentiation, function and regulation. Int. J. Androl. 29, 90–95; discussion 105–108 (2006).

    Article  PubMed  CAS  Google Scholar 

  30. Shima, Y. et al. Contribution of Leydig and Sertoli cells to testosterone production in mouse fetal testes. Mol. Endocrinol. 27, 63–73 (2013).

    Article  CAS  PubMed  Google Scholar 

  31. Alexander, G. M. Postnatal testosterone concentrations and male social development. Front. Endocrinol. 5, 15 (2014).

    Article  Google Scholar 

  32. Ye, L., Li, X., Li, L., Chen, H. & Ge, R. S. Insights into the development of the adult leydig cell lineage from stem leydig cells. Front. Physiol. 8, 430 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Chen, H., Stanley, E., Jin, S. & Zirkin, B. R. Stem Leydig cells: from fetal to aged animals. Birth Defects Res. C Embryo Today 90, 272–283 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Chen, H., Wang, Y., Ge, R. & Zirkin, B. R. Leydig cell stem cells: Identification, proliferation and differentiation. Mol. Cell Endocrinol. 445, 65–73 (2017).

    Article  CAS  PubMed  Google Scholar 

  35. Chemes, H. Leydig cell development in humans. In The Leydig Cell. 175-202 (Cache River Press, 1996).

  36. Kumar, D. L. & DeFalco, T. A perivascular niche for multipotent progenitors in the fetal testis. Nat. Commun. 9, 4519 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Davidoff, M. S. et al. Progenitor cells of the testosterone-producing Leydig cells revealed. J. Cell Biol. 167, 935–944 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Davidoff, M. S., Middendorff, R., Muller, D. & Holstein, A. F. The neuroendocrine Leydig cells and their stem cell progenitors, the pericytes. Adv. Anat. Embryol. Cell Biol. 205, 1–107 (2009).

    Article  PubMed  Google Scholar 

  39. O’Shaughnessy, P. J., Morris, I. D. & Baker, P. J. Leydig cell re-generation and expression of cell signaling molecules in the germ cell-free testis. Reproduction 135, 851–858 (2008).

    Article  PubMed  CAS  Google Scholar 

  40. Chemes, H. E. in The Leydig Cell. 175-202 (Cache River Press, 1996).

  41. Li, X. et al. Regulation of seminiferous tubule-associated stem Leydig cells in adult rat testes. Proc. Natl Acad. Sci. USA 113, 2666–2671 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Ge, R. S. et al. In search of rat stem Leydig cells: identification, isolation, and lineage-specific development. Proc. Natl Acad. Sci. USA 103, 2719–2724 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Haider, S. G., Passia, D. & Overmeyer, G. Studies on the fetal and postnatal development of rat Leydig cells employing 3 beta-hydroxysteroid dehydrogenase activity. Acta Histochem. Suppl. 32, 197–202 (1986).

    CAS  PubMed  Google Scholar 

  44. Dupont, E., Labrie, F., Luu-The, V. & Pelletier, G. Ontogeny of 3 beta-hydroxysteroid dehydrogenase/delta 5-delta 4 isomerase (3 beta-HSD) in rat testis as studied by immunocytochemistry. Anat. Embryol. 187, 583–589 (1993).

    Article  CAS  Google Scholar 

  45. Shan, L. X., Phillips, D. M., Bardin, C. W. & Hardy, M. P. Differential regulation of steroidogenic enzymes during differentiation optimizes testosterone production by adult rat Leydig cells. Endocrinology 133, 2277–2283 (1993).

    Article  CAS  PubMed  Google Scholar 

  46. Zirkin, B. R. & Ewing, L. L. Leydig cell differentiation during maturation of the rat testis: a stereological study of cell number and ultrastructure. Anat. Rec. 219, 157–163 (1987).

    Article  CAS  PubMed  Google Scholar 

  47. Zimmerman, R. A. et al. 5alpha-Androstane-3alpha,17beta-diol activates pathway that resembles the epidermal growth factor responsive pathways in stimulating human prostate cancer LNCaP cell proliferation. Prostate Cancer Prostatic Dis. 7, 364–374 (2004).

    Article  CAS  PubMed  Google Scholar 

  48. Prince, F. P. Ultrastructure of immature Leydig cells in the human prepubertal testis. Anat. Rec. 209, 165–176 (1984).

    Article  CAS  PubMed  Google Scholar 

  49. Inano, H. & Tamaoki, B. I. Bioconversion of steroids in immature rat testes in vitro. Endocrinology 79, 579–590 (1966).

    Article  CAS  PubMed  Google Scholar 

  50. Steinberger, E. & Ficher, M. Differentiation of steroid biosynthetic pathways in developing testes. Biol. Reprod. 1, 119–133 (1969).

    Article  Google Scholar 

  51. Murono, E. P. Maturational changes in steroidogenic enzyme activities metabolizing testosterone and dihydrotestosterone in two populations of testicular interstitial cells. Acta Endocrinol. 121, 477–483 (1989).

    CAS  Google Scholar 

  52. Rommerts, F. F., Teerds, K., Themmen, A. P. & van Noort, M. Multiple regulation of testicular steroidogenesis. J. Steroid Biochem. 27, 309–316 (1987).

    Article  CAS  PubMed  Google Scholar 

  53. Teerds, K. J. et al. Effects of pure FSH and LH preparations on the number and function of Leydig cells in immature hypophysectomized rats. J. Endocrinol. 120, 97–106 (1989).

    Article  CAS  PubMed  Google Scholar 

  54. Chemes, H. et al. Isolation of human Leydig cell mesenchymal precursors from patients with the androgen insensitivity syndrome: testosterone production and response to human chorionic gonadotropin stimulation in culture. Biol. Reprod. 46, 793–801 (1992).

    Article  CAS  PubMed  Google Scholar 

  55. Hardy, M. P., Kelce, W. R., Klinefelter, G. R. & Ewing, L. L. Differentiation of Leydig cell precursors in vitro: a role for androgen. Endocrinology 127, 488–490 (1990).

    Article  CAS  PubMed  Google Scholar 

  56. Murono, E. P. & Washburn, A. L. Regulation of 5 alpha-reductase activity in cultured immature leydig cells by human chorionic gonadotropin. J. Steroid Biochem. 35, 715–721 (1990).

    Article  CAS  PubMed  Google Scholar 

  57. Murono, E. P., Washburn, A. L. & Goforth, D. P. Enhanced stimulation of 5 alpha-reductase activity in cultured Leydig cell precursors by human chorionic gonadotropin. J. Steroid Biochem. Mol. Biol. 48, 377–384 (1994).

    Article  CAS  PubMed  Google Scholar 

  58. Hardy, M. P., Kirby, J. D., Hess, R. A. & Cooke, P. S. Leydig cells increase their numbers but decline in steroidogenic function in the adult rat after neonatal hypothyroidism. Endocrinology 132, 2417–2420 (1993).

    Article  CAS  PubMed  Google Scholar 

  59. Mendis-Handagama, S. M. & Sharma, O. P. Effects of neonatal administration of the reversible goitrogen propylthiouracil on the testis interstitium in adult rats. J. Reprod. Fertil. 100, 85–92 (1994).

    Article  CAS  PubMed  Google Scholar 

  60. Gaytan, F., Pinilla, L., Romero, J. L. & Aguilar, E. Differential effects of the administration of human chorionic gonadotropin to postnatal rats. J. Endocrinol. 142, 527–534 (1994).

    Article  CAS  PubMed  Google Scholar 

  61. Chemes, H. E. et al. Response to acute hCG stimulation and steroidogenic potential of Leydig cell fibroblastic precursors in humans. J. Androl. 6, 102–112 (1985).

    Article  CAS  PubMed  Google Scholar 

  62. Khan, S., Teerds, K. & Dorrington, J. Growth factor requirements for DNA synthesis by Leydig cells from the immature rat. Biol. Reprod. 46, 335–341 (1992).

    Article  CAS  PubMed  Google Scholar 

  63. Moore, A. & Morris, I. D. The involvement of insulin-like growth factor-I in local control of steroidogenesis and DNA synthesis of Leydig and non-Leydig cells in the rat testicular interstitium. J. Endocrinol. 138, 107–114 (1993).

    Article  CAS  PubMed  Google Scholar 

  64. van Haren, L., Flinterman, J. F., Orly, J. & Rommerts, F. F. Luteinizing hormone induction of the cholesterol side-chain cleavage enzyme in cultured immature rat Leydig cells: no role of insulin-like growth factor-I? Mol. Cell Endocrinol. 87, 57–67 (1992).

    Article  PubMed  Google Scholar 

  65. Lin, T., Blaisdell, J. & Haskell, J. F. Type I IGF receptors of Leydig cells are upregulated by human chorionic gonadotropin. Biochem. Biophys. Res. Commun. 149, 852–858 (1987).

    Article  CAS  PubMed  Google Scholar 

  66. Lin, T., Blaisdell, J. & Haskell, J. F. Hormonal regulation of type I insulin-like growth factor receptors of Leydig cells in hypophysectomized rats. Endocrinology 123, 134–139 (1988).

    Article  CAS  PubMed  Google Scholar 

  67. Nagpal, M. L., Wang, D., Calkins, J. H., Chang, W. W. & Lin, T. Human chorionic gonadotropin up-regulates insulin-like growth factor-I receptor gene expression of Leydig cells. Endocrinology 129, 2820–2826 (1991).

    Article  CAS  PubMed  Google Scholar 

  68. Gnessi, L. et al. Testicular development involves the spatiotemporal control of PDGFs and PDGF receptors gene expression and action. J. Cell Biol. 131, 1105–1121 (1995).

    Article  CAS  PubMed  Google Scholar 

  69. Song, T. et al. Parathyroid hormone-related protein promotes rat stem leydig cell differentiation. Front. Physiol. 8, 911 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  70. Mendis-Handagama, S. M. & Siril Ariyaratne, H. B. Leydig cells, thyroid hormones and steroidogenesis. Indian. J. Exp. Biol. 43, 939–962 (2005).

    CAS  PubMed  Google Scholar 

  71. Zhang, M. et al. Transplanted human p75-positive stem Leydig cells replace disrupted Leydig cells for testosterone production. Cell Death Dis. 8, e3123 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Basaria, S. Male hypogonadism. Lancet 383, 1250–1263 (2014).

    Article  CAS  PubMed  Google Scholar 

  73. Bhasin, S. et al. Testosterone therapy in men with androgen deficiency syndromes: an Endocrine Society clinical practice guideline. J. Clin. Endocrinol. Metab. 95, 2536–2559 (2010).

    Article  CAS  PubMed  Google Scholar 

  74. Payne, A. H. & Hardy, M. P. The Leydig cell in health and disease (Springer Science & Business Media, 2007).

  75. Fraietta, R., Zylberstejn, D. S. & Esteves, S. C. Hypogonadotropic hypogonadism revisited. Clinics 68, 81–88 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  76. Skakkebaek, N. E. et al. Male reproductive disorders and fertility trends: influences of environment and genetic susceptibility. Physiol. Rev. 96, 55–97 (2016).

    Article  CAS  PubMed  Google Scholar 

  77. Radke, E. G., Braun, J. M., Meeker, J. D. & Cooper, G. S. Phthalate exposure and male reproductive outcomes: A systematic review of the human epidemiological evidence. Env. Int. 121, 764–793 (2018).

    Article  CAS  Google Scholar 

  78. Viswanathan, V. & Eugster, E. A. Etiology and treatment of hypogonadism in adolescents. Pediatr. Clin. North. Am. 58, 1181–1200 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  79. Travison, T. G., Araujo, A. B., O’Donnell, A. B., Kupelian, V. & McKinlay, J. B. A population-level decline in serum testosterone levels in American men. J. Clin. Endocrinol. Metab. 92, 196–202 (2007).

    Article  CAS  PubMed  Google Scholar 

  80. Nieschlag, E. Late-onset hypogonadism: a concept comes of age. Andrology 8, 1506–1511 (2019).

    Article  PubMed  Google Scholar 

  81. Midzak, A. S., Chen, H., Papadopoulos, V. & Zirkin, B. R. Leydig cell aging and the mechanisms of reduced testosterone synthesis. Mol. Cell Endocrinol. 299, 23–31 (2009).

    Article  CAS  PubMed  Google Scholar 

  82. Masterson, J. M., Soodana-Prakash, N., Patel, A. S., Kargi, A. Y. & Ramasamy, R. Elevated body mass index is associated with secondary hypogonadism among men presenting to a tertiary academic medical center. World J. Mens. Health 37, 93–98 (2019).

    Article  PubMed  Google Scholar 

  83. Wu, F. C. et al. Identification of late-onset hypogonadism in middle-aged and elderly men. N. Engl. J. Med. 363, 123–135 (2010).

    Article  CAS  PubMed  Google Scholar 

  84. Lawaetz, J. G. et al. Evaluation of 451 Danish boys with delayed puberty: diagnostic use of a new puberty nomogram and effects of oral testosterone therapy. J. Clin. Endocrinol. Metab. 100, 1376–1385 (2015).

    Article  CAS  PubMed  Google Scholar 

  85. Seftel, A. D. Male hypogonadism. Part I: Epidemiology of hypogonadism. Int. J. Impot. Res. 18, 115–120 (2006).

    Article  CAS  PubMed  Google Scholar 

  86. Barbonetti, A., D’Andrea, S. & Francavilla, S. Testosterone replacement therapy. Andrology 8, 1551–1566 (2020).

    Article  CAS  PubMed  Google Scholar 

  87. Westfield, G., Kaiser, U. B., Lamb, D. J. & Ramasamy, R. Short-acting testosterone: more physiologic? Front. Endocrinol. 11, 572465 (2020).

    Article  Google Scholar 

  88. McBride, J. A. & Coward, R. M. Recovery of spermatogenesis following testosterone replacement therapy or anabolic-androgenic steroid use. Asian J. Androl. 18, 373–380 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Dohle, G. R., Smit, M. & Weber, R. F. Androgens and male fertility. World J. Urol. 21, 341–345 (2003).

    Article  CAS  PubMed  Google Scholar 

  90. Anawalt, B. D. et al. Serum inhibin B levels reflect Sertoli cell function in normal men and men with testicular dysfunction. J. Clin. Endocrinol. Metab. 81, 3341–3345 (1996).

    CAS  PubMed  Google Scholar 

  91. Finkel, D. M., Phillips, J. L. & Snyder, P. J. Stimulation of spermatogenesis by gonadotropins in men with hypogonadotropic hypogonadism. N. Engl. J. Med. 313, 651–655 (1985).

    Article  CAS  PubMed  Google Scholar 

  92. Nieschlag, E., Simoni, M., Gromoll, J. & Weinbauer, G. F. Role of FSH in the regulation of spermatogenesis: clinical aspects. Clin. Endocrinol. 51, 139–146 (1999).

    Article  CAS  Google Scholar 

  93. Ramasamy, R. et al. Effect of natesto on reproductive hormones, semen parameters and hypogonadal symptoms: a single center, open label, single arm trial. J. Urol. 204, 557–563 (2020).

    Article  PubMed  Google Scholar 

  94. Rogol, A. D., Tkachenko, N. & Bryson, N. Natesto, a novel testosterone nasal gel, normalizes androgen levels in hypogonadal men. Andrology 4, 46–54 (2016).

    Article  CAS  PubMed  Google Scholar 

  95. Gagliano-Juca, T. & Basaria, S. Testosterone replacement therapy and cardiovascular risk. Nat. Rev. Cardiol. 16, 555–574 (2019).

    Article  PubMed  Google Scholar 

  96. Vigen, R. et al. Association of testosterone therapy with mortality, myocardial infarction, and stroke in men with low testosterone levels. JAMA 310, 1829–1836 (2013).

    Article  CAS  PubMed  Google Scholar 

  97. Finkle, W. D. et al. Increased risk of non-fatal myocardial infarction following testosterone therapy prescription in men. PLoS One 9, e85805 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  98. Bhasin, S. et al. Testosterone therapy in men with hypogonadism: an endocrine society clinical practice guideline. J. Clin. Endocrinol. Metab. 103, 1715–1744 (2018).

    Article  PubMed  Google Scholar 

  99. Goodman, N. et al. American Association of Clinical Endocrinologists and American College of endocrinology position statement on the association of testosterone and cardiovascular risk. Endocr. Pract. 21, 1066–1073 (2015).

    Article  PubMed  Google Scholar 

  100. Calof, O. M. et al. Adverse events associated with testosterone replacement in middle-aged and older men: a meta-analysis of randomized, placebo-controlled trials. J. Gerontol. A Biol. Sci. Med. Sci 60, 1451–1457 (2005).

    Article  PubMed  Google Scholar 

  101. Constantinou, J. & Feneley, M. R. PSA testing: an evolving relationship with prostate cancer screening. Prostate Cancer Prostatic Dis. 9, 6–13 (2006).

    Article  CAS  PubMed  Google Scholar 

  102. Cui, Y., Zong, H., Yan, H. & Zhang, Y. The effect of testosterone replacement therapy on prostate cancer: a systematic review and meta-analysis. Prostate Cancer Prostatic Dis. 17, 132–143 (2014).

    Article  CAS  PubMed  Google Scholar 

  103. Bell, M. A., Campbell, J. D., Joice, G., Sopko, N. A. & Burnett, A. L. Shifting the paradigm of testosterone replacement therapy in prostate cancer. World J. Mens. Health 36, 103–109 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  104. Lewis, T. & Goldstein, I. Transfer of topical testosterone preparations to children or spouses. J. Sex. Med. 6, 2649–2652 (2009).

    Article  PubMed  Google Scholar 

  105. Wittert, G. The relationship between sleep disorders and testosterone in men. Asian J. Androl. 16, 262–265 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Trost, L. W. & Mulhall, J. P. Challenges in testosterone measurement, data interpretation, and methodological appraisal of interventional trials. J. Sex. Med. 13, 1029–1046 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  107. Lee, J. A. & Ramasamy, R. Indications for the use of human chorionic gonadotropic hormone for the management of infertility in hypogonadal men. Transl. Androl. Urol. 7, S348–S352 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  108. Krzastek, S. C. & Smith, R. P. Non-testosterone management of male hypogonadism: an examination of the existing literature. Transl. Androl. Urol. 9, S160–S170 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  109. Surampudi, P., Swerdloff, R. S. & Wang, C. An update on male hypogonadism therapy. Expert. Opin. Pharmacother. 15, 1247–1264 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Liu, P. Y. et al. Induction of spermatogenesis and fertility during gonadotropin treatment of gonadotropin-deficient infertile men: predictors of fertility outcome. J. Clin. Endocrinol. Metab. 94, 801–808 (2009).

    Article  CAS  PubMed  Google Scholar 

  111. Carrasquillo, R., Chu, K. & Ramasamy, R. Novel therapy for male hypogonadism. Curr. Urol. Rep. 19, 63 (2018).

    Article  PubMed  Google Scholar 

  112. Larijani, B. et al. Stem cell therapy in treatment of different diseases. Acta Med. Iran. 50, 79–96 (2012).

    PubMed  Google Scholar 

  113. Evans, M. J. & Kaufman, M. H. Establishment in culture of pluripotential cells from mouse embryos. Nature 292, 154–156 (1981).

    Article  CAS  PubMed  Google Scholar 

  114. Martin, G. R. Isolation of a pluripotent cell line from early mouse embryos cultured in medium conditioned by teratocarcinoma stem cells. Proc. Natl Acad. Sci. USA 78, 7634–7638 (1981).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Thomson, J. A. et al. Embryonic stem cell lines derived from human blastocysts. Science 282, 1145–1147 (1998).

    Article  CAS  PubMed  Google Scholar 

  116. Takahashi, K. & Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663–676 (2006).

    Article  CAS  PubMed  Google Scholar 

  117. Takahashi, K. et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131, 861–872 (2007).

    Article  CAS  PubMed  Google Scholar 

  118. Morizane, A. et al. Direct comparison of autologous and allogeneic transplantation of iPSC-derived neural cells in the brain of a non-human primate. Stem Cell Rep. 1, 283–292 (2013).

    Article  CAS  Google Scholar 

  119. Bellin, M., Marchetto, M. C., Gage, F. H. & Mummery, C. L. Induced pluripotent stem cells: the new patient? Nat. Rev. Mol. Cell Biol. 13, 713–726 (2012).

    Article  PubMed  CAS  Google Scholar 

  120. Braganca, J., Lopes, J. A., Mendes-Silva, L. & Almeida Santos, J. M. Induced pluripotent stem cells, a giant leap for mankind therapeutic applications. World J. Stem Cell 11, 421–430 (2019).

    Article  Google Scholar 

  121. Lo, K. C., Lei, Z., Rao Ch, V., Beck, J. & Lamb, D. J. De novo testosterone production in luteinizing hormone receptor knockout mice after transplantation of Leydig stem cells. Endocrinology 145, 4011–4015 (2004).

    Article  CAS  PubMed  Google Scholar 

  122. Chen, P., Zirkin, B. R. & Chen, H. Stem Leydig cells in the adult testis: characterization, regulation and potential applications. Endocr. Rev. 41, 22–32 (2020).

    Article  Google Scholar 

  123. Zhang, L. et al. NGF induces adult stem Leydig cells to proliferate and differentiate during Leydig cell regeneration. Biochem. Biophys. Res. Commun. 436, 300–305 (2013).

    Article  CAS  PubMed  Google Scholar 

  124. Odeh, H. M., Kleinguetl, C., Ge, R., Zirkin, B. R. & Chen, H. Regulation of the proliferation and differentiation of Leydig stem cells in the adult testis. Biol. Reprod. 90, 123 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  125. Landreh, L. et al. Human testicular peritubular cells host putative stem Leydig cells with steroidogenic capacity. J. Clin. Endocrinol. Metab. 99, E1227–E1235 (2014).

    Article  CAS  PubMed  Google Scholar 

  126. Xia, K. et al. Restorative functions of Autologous Stem Leydig Cell transplantation in a Testosterone-deficient non-human primate model. Theranostics 10, 8705–8720 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Xia, K. et al. Endosialin defines human stem Leydig cells with regenerative potential. Hum. Reprod. 35, 2197–2212 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Jiang, M. H. et al. Characterization of Nestin-positive stem Leydig cells as a potential source for the treatment of testicular Leydig cell dysfunction. Cell Res. 24, 1466–1485 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  129. Zang, Z. J. et al. Transplantation of CD51+ stem Leydig cells: a new strategy for the treatment of testosterone deficiency. Stem Cell 35, 1222–1232 (2017).

    Article  CAS  Google Scholar 

  130. Zirkin, B. R. & Chen, H. Regulation of Leydig cell steroidogenic function during aging. Biol. Reprod. 63, 977–981 (2000).

    Article  CAS  PubMed  Google Scholar 

  131. Curley, M. et al. A young testicular microenvironment protects Leydig cells against age-related dysfunction in a mouse model of premature aging. FASEB J. 33, 978–995 (2019).

    Article  CAS  PubMed  Google Scholar 

  132. Frungieri, M. B., Calandra, R. S., Bartke, A. & Matzkin, M. E. Ageing and inflammation in the male reproductive tract. Andrologia 50, e13034 (2018).

    Article  CAS  PubMed  Google Scholar 

  133. Chen, H., Ge, R. S. & Zirkin, B. R. Leydig cells: from stem cells to aging. Mol. Cell Endocrinol. 306, 9–16 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Papadopoulos, V. & Zirkin, B. R. Leydig cell aging: molecular mechanisms and treatments. Vitam. Horm. 115, 585–609 (2021).

    Article  CAS  PubMed  Google Scholar 

  135. Arora, H. et al. Subcutaneous Leydig stem cell autograft: a promising strategy to increase serum testosterone. Stem Cell Transl. Med. 8, 58–65 (2019).

    Article  CAS  Google Scholar 

  136. Nauta, A. J. & Fibbe, W. E. Immunomodulatory properties of mesenchymal stromal cells. Blood 110, 3499–3506 (2007).

    Article  CAS  PubMed  Google Scholar 

  137. Gauthier-Fisher, A., Kauffman, A. & Librach, C. L. Potential use of stem cells for fertility preservation. Andrology 8, 862–878 (2019).

    Article  PubMed  Google Scholar 

  138. Peak, T. C., Haney, N. M., Wang, W., DeLay, K. J. & Hellstrom, W. J. Stem cell therapy for the treatment of Leydig cell dysfunction in primary hypogonadism. World J. Stem Cell 8, 306–315 (2016).

    Article  Google Scholar 

  139. Yazawa, T. et al. Differentiation of adult stem cells derived from bone marrow stroma into Leydig or adrenocortical cells. Endocrinology 147, 4104–4111 (2006).

    Article  CAS  PubMed  Google Scholar 

  140. Tanaka, T. et al. Steroidogenic factor 1/adrenal 4 binding protein transforms human bone marrow mesenchymal cells into steroidogenic cells. J. Mol. Endocrinol. 39, 343–350 (2007).

    Article  CAS  PubMed  Google Scholar 

  141. Yazawa, T. et al. Liver receptor homolog-1 regulates the transcription of steroidogenic enzymes and induces the differentiation of mesenchymal stem cells into steroidogenic cells. Endocrinology 150, 3885–3893 (2009).

    Article  CAS  PubMed  Google Scholar 

  142. Ju, Y. et al. Nuclear receptor 5A (NR5A) family regulates 5-aminolevulinic acid synthase 1 (ALAS1) gene expression in steroidogenic cells. Endocrinology 153, 5522–5534 (2012).

    Article  CAS  PubMed  Google Scholar 

  143. Wei, X., Peng, G., Zheng, S. & Wu, X. Differentiation of umbilical cord mesenchymal stem cells into steroidogenic cells in comparison to bone marrow mesenchymal stem cells. Cell Prolif. 45, 101–110 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Hou, L. et al. Gonadotropins facilitate potential differentiation of human bone marrow mesenchymal stem cells into Leydig cells in vitro. Kaohsiung J. Med. Sci. 32, 1–9 (2016).

    Article  PubMed  Google Scholar 

  145. Xing, X. et al. Differentiation of human umbilical cord mesenchymal stem cells into steroidogenic cells in vitro. Exp. Ther. Med. 12, 3527–3534 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Yang, C. et al. Transplanted adipose-derived stem cells ameliorate testicular dysfunction in a d-galactose-induced aging rat model. J. Cell Physiol. 230, 2403–2414 (2015).

    Article  CAS  PubMed  Google Scholar 

  147. Yazawa, T. et al. PPAR-gamma coactivator-1alpha regulates progesterone production in ovarian granulosa cells with SF-1 and LRH-1. Mol. Endocrinol. 24, 485–496 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Zhang, Z. Y., Xing, X. Y., Ju, G. Q., Zhong, L. & Sun, J. Mesenchymal stem cells from human umbilical cord ameliorate testicular dysfunction in a male rat hypogonadism model. Asian J. Androl. 19, 543–547 (2017).

    Article  CAS  PubMed  Google Scholar 

  149. Gauthier-Fisher, A., Szaraz, P. & Librach, C. L. Pericytes in the umbilical cord. Adv. Exp. Med. Biol. 1122, 211–233 (2019).

    Article  CAS  PubMed  Google Scholar 

  150. de Witte, S. F. H. et al. Aging of bone marrow- and umbilical cord-derived mesenchymal stromal cells during expansion. Cytotherapy 19, 798–807 (2017).

    Article  PubMed  CAS  Google Scholar 

  151. De Witte, S. F. H. et al. Epigenetic changes in umbilical cord mesenchymal stromal cells upon stimulation and culture expansion. Cytotherapy 20, 919–929 (2018).

    Article  PubMed  CAS  Google Scholar 

  152. Wang, Y. et al. Long-term cultured mesenchymal stem cells frequently develop genomic mutations but do not undergo malignant transformation. Cell Death Dis. 4, e950 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Zhou, T. et al. Generation of human induced pluripotent stem cells from urine samples. Nat. Protoc. 7, 2080–2089 (2012).

    Article  CAS  PubMed  Google Scholar 

  154. Liu, L. P. & Zheng, Y. W. Predicting differentiation potential of human pluripotent stem cells: possibilities and challenges. World J. Stem Cell 11, 375–382 (2019).

    Article  CAS  Google Scholar 

  155. Bilic, J. & Izpisua Belmonte, J. C. Concise review: Induced pluripotent stem cells versus embryonic stem cells: close enough or yet too far apart? Stem Cell 30, 33–41 (2012).

    Article  CAS  Google Scholar 

  156. Crawford, P. A., Sadovsky, Y. & Milbrandt, J. Nuclear receptor steroidogenic factor 1 directs embryonic stem cells toward the steroidogenic lineage. Mol. Cell Biol. 17, 3997–4006 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Jadhav, U. & Jameson, J. L. Steroidogenic factor-1 (SF-1)-driven differentiation of murine embryonic stem (ES) cells into a gonadal lineage. Endocrinology 152, 2870–2882 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Lin, Y. & Chen, G. in StemBook (IOS Press, 2008).

  159. Yazawa, T. et al. Differentiation of mesenchymal stem cells and embryonic stem cells into steroidogenic cells using steroidogenic factor-1 and liver receptor homolog-1. Mol. Cell Endocrinol. 336, 127–132 (2011).

    Article  CAS  PubMed  Google Scholar 

  160. Yang, Y. et al. Directed mouse embryonic stem cells into leydig-like cells rescue testosterone-deficient male rats in vivo. Stem Cell Dev. 24, 459–470 (2015).

    Article  CAS  Google Scholar 

  161. Sonoyama, T. et al. Differentiation of human embryonic stem cells and human induced pluripotent stem cells into steroid-producing cells. Endocrinology 153, 4336–4345 (2012).

    Article  CAS  PubMed  Google Scholar 

  162. Li, L. et al. Directing differentiation of human induced pluripotent stem cells toward androgen-producing Leydig cells rather than adrenal cells. Proc. Natl Acad. Sci. USA 116, 23274–23283 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Chen, X. et al. Differentiation of human induced pluripotent stem cells into Leydig-like cells with molecular compounds. Cell Death Dis. 10, 220 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  164. Matthews, K. R. & Morali, D. National human embryo and embryoid research policies: a survey of 22 top research-intensive countries. Regen. Med. 15, 1905–1917 (2020).

    Article  CAS  PubMed  Google Scholar 

  165. Robertson, J. A. Human embryonic stem cell research: ethical and legal issues. Nat. Rev. Genet. 2, 74–78 (2001).

    Article  CAS  PubMed  Google Scholar 

  166. Yoshihara, M. et al. Hotspots of de novo point mutations in induced pluripotent stem cells. Cell Rep. 21, 308–315 (2017).

    Article  CAS  PubMed  Google Scholar 

  167. Dakhore, S., Nayer, B. & Hasegawa, K. Human pluripotent stem cell culture: current status, challenges, and advancement. Stem Cell Int. 2018, 7396905 (2018).

    Google Scholar 

  168. Puri, M. C. & Nagy, A. Concise review: embryonic stem cells versus induced pluripotent stem cells: the game is on. Stem Cell 30, 10–14 (2012).

    Article  CAS  Google Scholar 

  169. Jopling, C., Boue, S. & Izpisua Belmonte, J. C. Dedifferentiation, transdifferentiation and reprogramming: three routes to regeneration. Nat. Rev. Mol. Cell Biol. 12, 79–89 (2011).

    Article  CAS  PubMed  Google Scholar 

  170. Yang, Y. et al. Direct reprogramming of mouse fibroblasts toward Leydig-like cells by defined factors. Stem Cell Rep. 8, 39–53 (2017).

    Article  CAS  Google Scholar 

  171. Yang, Y. et al. Conversion of fibroblast into functional leydig-like cell using defined small molecules. Stem Cell Rep. 15, 408–423 (2020).

    Article  CAS  Google Scholar 

  172. Ruiz-Babot, G. et al. Modeling congenital adrenal hyperplasia and testing interventions for adrenal insufficiency using donor-specific reprogrammed cells. Cell Rep. 22, 1236–1249 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Zhou, J. et al. Conversion of human fibroblasts into functional Leydig-like cells by small molecules and a single factor. Biochem. Biophys. Res. Commun. 516, 1–7 (2019).

    Article  CAS  PubMed  Google Scholar 

  174. Raab, S., Klingenstein, M., Liebau, S. & Linta, L. A comparative view on human somatic cell sources for iPSC generation. Stem Cell Int. 2014, 768391 (2014).

    Google Scholar 

  175. Chamberlain, S. J. Disease modelling using human iPSCs. Hum. Mol. Genet. 25, R173–R181 (2016).

    Article  CAS  PubMed  Google Scholar 

  176. Kumar, S., Blangero, J. & Curran, J. E. Induced pluripotent stem cells in disease modeling and gene identification. Methods Mol. Biol. 1706, 17–38 (2018).

    Article  PubMed  CAS  Google Scholar 

  177. Kondo, T. et al. Modeling Alzheimer’s disease with iPSCs reveals stress phenotypes associated with intracellular Abeta and differential drug responsiveness. Cell Stem Cell 12, 487–496 (2013).

    Article  CAS  PubMed  Google Scholar 

  178. Stark, Z. et al. Integrating genomics into healthcare: a global responsibility. Am. J. Hum. Genet. 104, 13–20 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Park, S. et al. A comprehensive, ethnically diverse library of sickle cell disease-specific induced pluripotent stem cells. Stem Cell Rep. 8, 1076–1085 (2017).

    Article  CAS  Google Scholar 

  180. Kotze, M. J. et al. Genomic medicine and risk prediction across the disease spectrum. Crit. Rev. Clin. Lab. Sci. 52, 120–137 (2015).

    Article  PubMed  Google Scholar 

  181. Hall, W. D., Morley, K. I. & Lucke, J. C. The prediction of disease risk in genomic medicine. EMBO Rep. 5 Spec No (Suppl 1), S22-6 (2004).

  182. Yoshida, Y. & Yamanaka, S. Induced pluripotent stem cells 10 years later: for cardiac applications. Circ. Res. 120, 1958–1968 (2017).

    Article  CAS  PubMed  Google Scholar 

  183. Smith, R. C. & Tabar, V. Constructing and deconstructing cancers using human pluripotent stem cells and organoids. Cell Stem Cell 24, 12–24 (2019).

    Article  CAS  PubMed  Google Scholar 

  184. Duca, Y., Aversa, A., Condorelli, R. A., Calogero, A. E. & La Vignera, S. Substance abuse and male hypogonadism. J. Clin. Med. 8, 732 (2019).

    Article  CAS  PubMed Central  Google Scholar 

  185. Wang, Y., Chen, F., Ye, L., Zirkin, B. & Chen, H. Steroidogenesis in Leydig cells: effects of aging and environmental factors. Reproduction 154, R111–R122 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  186. Kojima, Y. et al. Role of transcription factors Ad4bp/SF-1 and DAX-1 in steroidogenesis and spermatogenesis in human testicular development and idiopathic azoospermia. Int. J. Urol. 13, 785–793 (2006).

    Article  CAS  PubMed  Google Scholar 

  187. Karpova, T. et al. Steroidogenic factor 1 differentially regulates fetal and adult leydig cell development in male mice. Biol. Reprod. 93, 83 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  188. Kilcoyne, K. R. et al. Fetal programming of adult Leydig cell function by androgenic effects on stem/progenitor cells. Proc. Natl Acad. Sci. USA 111, E1924–E1932 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Qin, J., Tsai, M. J. & Tsai, S. Y. Essential roles of COUP-TFII in Leydig cell differentiation and male fertility. PLoS One 3, e3285 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  190. Yamashita, S., Tai, P., Charron, J., Ko, C. & Ascoli, M. The Leydig cell MEK/ERK pathway is critical for maintaining a functional population of adult Leydig cells and for fertility. Mol. Endocrinol. 25, 1211–1222 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Sharma, R., Bose, D., Maminishkis, A. & Bharti, K. Retinal pigment epithelium replacement therapy for age-related macular degeneration: are we there yet? Annu. Rev. Pharmacol. Toxicol. 60, 553–572 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Badyra, B., Sulkowski, M., Milczarek, O. & Majka, M. Mesenchymal stem cells as a multimodal treatment for nervous system diseases. Stem Cell Transl. Med. 9, 1174–1189 (2020).

    Article  Google Scholar 

  193. Kogawa, R., Nakamura, K. & Mochizuki, Y. A new islet transplantation method combining mesenchymal stem cells with recombinant peptide pieces, microencapsulated islets, and mesh bags. Biomedicines 8, 299 (2020).

    Article  CAS  PubMed Central  Google Scholar 

  194. Baburski, A. Z., Andric, S. A. & Kostic, T. S. Luteinizing hormone signaling is involved in synchronization of Leydig cell’s clock and is crucial for rhythm robustness of testosterone production dagger. Biol. Reprod. 100, 1406–1415 (2019).

    Article  PubMed  Google Scholar 

  195. Rone, M. B. et al. Identification of a dynamic mitochondrial protein complex driving cholesterol import, trafficking, and metabolism to steroid hormones. Mol. Endocrinol. 26, 1868–1882 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Chung, J. Y. et al. Drug ligand-induced activation of translocator protein (TSPO) stimulates steroid production by aged brown Norway rat Leydig cells. Endocrinology 154, 2156–2165 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Musicki, B. et al. TSPO ligand FGIN-1-27 controls priapism in sickle cell mice via endogenous testosterone production. J. Cell Physiol. 236, 3073–3082 (2021).

    Article  CAS  PubMed  Google Scholar 

  198. Aghazadeh, Y., Martinez-Arguelles, D. B., Fan, J., Culty, M. & Papadopoulos, V. Induction of androgen formation in the male by a TAT-VDAC1 fusion peptide blocking 14-3-3varepsilon protein adaptor and mitochondrial VDAC1 interactions. Mol. Ther. 22, 1779–1791 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Ranga, A., Gjorevski, N. & Lutolf, M. P. Drug discovery through stem cell-based organoid models. Adv. Drug Deliv. Rev. 69–70, 19–28 (2014).

    Article  PubMed  CAS  Google Scholar 

  200. Musunuru, K. et al. Induced pluripotent stem cells for cardiovascular disease modeling and precision medicine: a scientific statement from the american heart association. Circ. Genom. Precis. Med. 11, e000043 (2018).

    PubMed  PubMed Central  Google Scholar 

  201. Chen, Q. et al. Endothelial cells are progenitors of cardiac pericytes and vascular smooth muscle cells. Nat. Commun. 7, 12422 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Silva, M. C. & Haggarty, S. J. Human pluripotent stem cell-derived models and drug screening in CNS precision medicine. Ann. N. Y. Acad. Sci. 1471, 18–56 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  203. Elitt, M. S., Barbar, L. & Tesar, P. J. Drug screening for human genetic diseases using iPSC models. Hum. Mol. Genet. 27, R89–R98 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Baert, Y., Dvorakova-Hortova, K., Margaryan, H. & Goossens, E. Mouse in vitro spermatogenesis on alginate-based 3D bioprinted scaffolds. Biofabrication 11, 035011 (2019).

    Article  CAS  PubMed  Google Scholar 

  205. Smith, J. F. et al. Testicular niche required for human spermatogonial stem cell expansion. Stem Cell Transl. Med. 3, 1043–1054 (2014).

    Article  CAS  Google Scholar 

  206. Fujisawa, M. Regulation of testicular function by cell-to-cell interaction. Reprod. Med. Biol. 5, 9–17 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  207. Rodriguez Gutierrez, D., Eid, W. & Biason-Lauber, A. A human gonadal cell model from induced pluripotent stem cells. Front. Genet. 9, 498 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  208. Piprek, R. P., Kolasa, M., Podkowa, D., Kloc, M. & Kubiak, J. Z. Transcriptional profiling validates involvement of extracellular matrix and proteinases genes in mouse gonad development. Mech. Dev. 149, 9–19 (2018).

    Article  CAS  PubMed  Google Scholar 

  209. Laronda, M. M. et al. A bioprosthetic ovary created using 3D printed microporous scaffolds restores ovarian function in sterilized mice. Nat. Commun. 8, 15261 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Stanley, E. et al. Identification, proliferation, and differentiation of adult Leydig stem cells. Endocrinology 153, 5002–5010 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Contributions

L.L. and V.P. researched data for the article, made substantial contributions to the discussion of its content, wrote the article, and reviewed and edited the manuscript before submission.

Corresponding author

Correspondence to Vassilios Papadopoulos.

Ethics declarations

Competing interests

V.P. is named co-inventor on patents reporting on new molecules inducing testosterone production by normal and hypofunctional Leydig cells issued and filed with U.S.P.T.O. and other international agencies. These patents were licensed by McGill University to IASO BioMed, Colorado, USA. V.P. has received stock from IASO BioMed. L.L. declares no competing interests.

Additional information

Peer review information

Nature Reviews Urology thanks R. Ramasamy, Stefan Schlatt and the other, anonymous reviewer for their contribution to the peer review of this work.

Publisher’s note

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

Related links

www.clinicaltrials.gov

Glossary

Ductal structures

Embryos contain two sets of ductal structures, the Wolffian and Müllerian ducts, which develop into the male and female reproductive tracts, respectively.

Total testosterone

All testosterone in the blood, including free testosterone and testosterone bound to albumin and sex hormone binding globulin (SHBG).

Free testosterone

Testosterone in the blood not bound to any proteins.

Blastocyst

Mammalian preimplantation embryos consisting of an inner cell mass (giving rise to embryos) and trophectoderm (giving rise to the placenta).

Stemness

The ability of a cell to proliferate, to differentiate and to keep a balance between proliferation, regeneration and quiescence.

Myoid cells

Smooth muscle cells surrounding the seminiferous tubule.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, L., Papadopoulos, V. Advances in stem cell research for the treatment of primary hypogonadism. Nat Rev Urol 18, 487–507 (2021). https://doi.org/10.1038/s41585-021-00480-2

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41585-021-00480-2

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