The Regulation of Spermatogenesis by Androgens (2024)

1. Chang C, Lee SO, Wang RS, Yeh S, Chang TM. Androgen receptor (AR) physiological roles in male and female reproductive systems: lessons learned from AR-knockout mice lacking AR in selective cells. Biol Reprod. 2013;89:21. [PMC free article] [PubMed] [Google Scholar]

2. Walker WH. Non-classical actions of testosterone and spermatogenesis. Philos Trans R Soc Lond B Biol Sci. 2010;365:1557–69. [PMC free article] [PubMed] [Google Scholar]

3. Walker WH. Testosterone signaling and the regulation of spermatogenesis. Spermatogenesis. 2011;1:116–120. [PMC free article] [PubMed] [Google Scholar]

4. Verhoeven G, Willems A, Denolet E, Swinnen JV, De Gendt K. Androgens and spermatogenesis: lessons from transgenic mouse models. Philos Trans R Soc Lond B Biol Sci. 2010;365:1537–56. [PMC free article] [PubMed] [Google Scholar]

5. Alves MG, Rato L, Carvalho RA, Moreira PI, Socorro S, Oliveira PF. Hormonal control of Sertoli cell metabolism regulates spermatogenesis. Cell Mol Life Sci. 2013;70:777–93. [PMC free article] [PubMed] [Google Scholar]

6. Ruwanpura SM, McLachlan RI, Meachem SJ. Hormonal regulation of male germ cell development. J Endocrinol. 2010;205:117–31. [PubMed] [Google Scholar]

7. Richardson LL, Kleinman HK, Dym M. Basem*nt membrane gene expression by Sertoli and peritubular myoid cells in vitro in the rat. Biol Reprod. 1995;52:320–30. [PubMed] [Google Scholar]

8. Skinner MK, Tung PS, Fritz IB. Cooperativity between Sertoli cells and testicular peritubular cells in the production and deposition of extracellular matrix components. J Cell Biol. 1985;100:1941–7. [PMC free article] [PubMed] [Google Scholar]

9. Mruk DD, Cheng CY. Sertoli-Sertoli and Sertoli-germ cell interactions and their significance in germ cell movement in the seminiferous epithelium during spermatogenesis. Endocr Rev. 2004;25:747–806. [PubMed] [Google Scholar]

10. Skinner MK. Sertoli Cell Secreted Regulatory Factors. In: Skinner MK, Griswold MD, editors. Sertoli Cell Biology. Elsevier Science; San Diego: 2005. pp. 107–120. [Google Scholar]

11. Wang RS, Yeh S, Tzeng CR, Chang C. Androgen receptor roles in spermatogenesis and fertility: lessons from testicular cell-specific androgen receptor knockout mice. Endocr Rev. 2009;30:119–32. [PMC free article] [PubMed] [Google Scholar]

12. Lyon MF, Glenister PH, Lamoreux ML. Normal spermatozoa from androgen-resistant germ cells of chimaeric mice and the role of androgen in spermatogenesis. Nature. 1975;258:620–2. [PubMed] [Google Scholar]

13. Johnston DS, Russell LD, Friel PJ, Griswold MD. Murine germ cells do not require functional androgen receptors to complete spermatogenesis following spermatogonial stem cell transplantation. Endocrinology. 2001;142:2405–8. [PubMed] [Google Scholar]

14. Tsai MY, Yeh SD, Wang RS, Yeh S, Zhang C, Lin HY, et al. Differential effects of spermatogenesis and fertility in mice lacking androgen receptor in individual testis cells. Proc Natl Acad Sci U S A. 2006;103:18975–80. [PMC free article] [PubMed] [Google Scholar]

15. Comhaire FH, Vermeulen A. Testosterone concentration in the fluids of seminiferous tubules, the interstitium and the rete testis of the rat. J Endocrinol. 1976;70:229–235. [PubMed] [Google Scholar]

16. Turner TT, Jones CE, Howards SS, Ewing LL, Zegeye B, Gunsalus GL. On the androgen microenvironment of maturing spermatozoa. Endocrinology. 1984;115:1925–1932. [PubMed] [Google Scholar]

17. Awoniyi CA, Santulli R, Sprando RL, Ewing LL, Zirkin BR. Restoration of advanced spermatogenic cells in the experimentally regressed rat testis: quantitative relationship to testosterone concentration within the testis. Endocrinology. 1989;124:1217–23. [PubMed] [Google Scholar]

18. Maddocks S, Hargreave TB, Reddie K, Fraser HM, Kerr JB, Sharpe RM. Intratesticular hormone levels and the route of secretion of hormones from the testis of the rat, guinea pig, monkey and human. Int J Androl. 1993;16:272–8. [PubMed] [Google Scholar]

19. Jarow JP, Chen H, Rosner TW, Trentacoste S, Zirkint BR. Assessment of the androgen environment within the human testis: minimally invasive method to obtain intratesticular fluid. J Androl. 2001;22:640–5. [PubMed] [Google Scholar]

20. Zirkin BR, Santulli R, Awoniyi CA, Ewing LL. Maintenance of advanced spermatogenic cells in the adult rat testis: quantitative relationship to testosterone concentration within the testis. Endocrinology. 1989;124:3043–9. [PubMed] [Google Scholar]

21. Hammond GL, Ruokonen A, Kontturi M, Koskela E, Vihko R. The simultaneous radioimmunoassay of seven steroids in human spermatic and peripheral venous blood. J Clin Endocrinol Metab. 1977;45:16–24. [PubMed] [Google Scholar]

22. Jarow JP, Wright WW, Brown TR, Yan X, Zirkin BR. Bioactivity of androgens within the testes and serum of normal men. J Androl. 2005;26:343–8. [PubMed] [Google Scholar]

23. Tsai MJ, O’Malley BW. Molecular mechanisms of action of steroid/thyroid receptor superfamily members. Ann Rev Biochem. 1994;63:451–486. [PubMed] [Google Scholar]

24. Shapiro E, Huang H, Masch RJ, McFadden DE, Wu XR, Ostrer H. Immunolocalization of androgen receptor and estrogen receptors alpha and beta in human fetal testis and epididymis. J Urol. 2005;174:1695–8. [PubMed] [Google Scholar]

25. Berensztein EB, Baquedano MS, Gonzalez CR, Saraco NI, Rodriguez J, Ponzio R, et al. Expression of aromatase, estrogen receptor alpha and beta, androgen receptor, and cytochrome P-450scc in the human early prepubertal testis. Pediatr Res. 2006;60:740–4. [PubMed] [Google Scholar]

26. Chemes HE, Rey RA, Nistal M, Regadera J, Musse M, Gonzalez-Peramato P, et al. Physiological androgen insensitivity of the fetal, neonatal, and early infantile testis is explained by the ontogeny of the androgen receptor expression in Sertoli cells. J Clin Endocrinol Metab. 2008;93:4408–12. [PubMed] [Google Scholar]

27. Boukari K, Meduri G, Brailly-Tabard S, Guibourdenche J, Ciampi ML, Massin N, et al. Lack of androgen receptor expression in Sertoli cells accounts for the absence of anti-Mullerian hormone repression during early human testis development. J Clin Endocrinol Metab. 2009;94:1818–25. [PMC free article] [PubMed] [Google Scholar]

28. Rey RA, Musse M, Venara M, Chemes HE. Ontogeny of the androgen receptor expression in the fetal and postnatal testis: its relevance on Sertoli cell maturation and the onset of adult spermatogenesis. Microsc Res Tech. 2009;72:787–95. [PubMed] [Google Scholar]

29. You L, Sar M. Androgen receptor expression in the testes and epididymides of prenatal and postnatal Sprague-Dawley rats. Endocrine. 1998;9:253–61. [PubMed] [Google Scholar]

30. Majumdar SS, Sarda K, Bhattacharya I, Plant TM. Insufficient androgen and FSH signaling may be responsible for the azoospermia of the infantile primate testes despite exposure to an adult-like hormonal milieu. Hum Reprod. 2012;27:2515–25. [PMC free article] [PubMed] [Google Scholar]

31. Buzek SW, Sanborn BM. Increase in testicular androgen receptor during sexual maturation in the rat. Biol Reprod. 1988;39:39–49. [PubMed] [Google Scholar]

32. Bremner WJ, Millar MR, Sharpe RM, Saunders PTK. Immunohistochemical localization of androgen receptors in the rat testis: evidence for stage-dependent expression and regulation by androgens. Endocrinology. 1994;135:1227–1234. [PubMed] [Google Scholar]

33. Zhou X, Kudo A, Kawakami H, Hirano H. Immunohistochemical localization of androgen receptor in mouse testicular germ cells during fetal and postnatal development. Anat Rec. 1996;245:509–518. [PubMed] [Google Scholar]

34. Suarez-Quian CA, Martinez-Garcia F, Nistal M, Regadera J. Androgen receptor distribution in adult human testis. J Clin Endocrin Metab. 1999;84:350–358. [PubMed] [Google Scholar]

35. Vornberger W, Prins G, Musto NA, Suarez-Quian CA. Androgen receptor distribution in rat testis: new implications for androgen regulation of spermatogenesis. Endocrinology. 1994;134:2307–2316. [PubMed] [Google Scholar]

36. Shan L-X, Zhu K, L-J, Bardin CW, Hardy MP. Quantitative analysis of androgen receptor messenger ribonucleic acid in developing Leydig cells and Sertoli cells by in situ hybridization. Endocrinology. 1995;136:3856–3862. [PubMed] [Google Scholar]

37. Kerr JB, Millar M, Maddocks S, Sharpe RM. Stage-dependent changes in spermatogenesis and Sertoli cells in relation to the onset of spermatogenic failure following withdrawal of testosterone. Anat Rec. 1993;235:547–59. [PubMed] [Google Scholar]

38. Sharpe RM. Regulation of spermatogenesis. In: Knobil E, Neil JD, editors. The Physiology of Reproduction. Raven Press; New York: 1994. pp. 1363–1434. [Google Scholar]

39. Pelletier RM. The blood-testis barrier: the junctional permeability, the proteins and the lipids. Prog Histochem Cytochem. 2011;46:49–127. [PubMed] [Google Scholar]

40. Meng J, Holdcraft RW, Shima JE, Griswold MD, Braun RE. Androgens regulate the permeability of the blood-testis barrier. Proc Natl Acad Sci U S A. 2005;102:16696–700. [PMC free article] [PubMed] [Google Scholar]

41. Yan HH, Mruk DD, Lee WM, Cheng CY. Blood-testis barrier dynamics are regulated by testosterone and cytokines via their differential effects on the kinetics of protein endocytosis and recycling in Sertoli cells. Faseb J. 2008;22:1945–59. [PMC free article] [PubMed] [Google Scholar]

42. Su L, Mruk DD, Lee WM, Cheng CY. Differential effects of testosterone and TGF-beta3 on endocytic vesicle-mediated protein trafficking events at the blood-testis barrier. Exp Cell Res. 2010;316:2945–60. [PMC free article] [PubMed] [Google Scholar]

43. Yeh S, Tsai MY, Xu Q, Mu XM, Lardy H, Huang KE, et al. Generation and characterization of androgen receptor knockout (ARKO) mice: an in vivo model for the study of androgen functions in selective tissues. Proc Natl Acad Sci U S A. 2002;99:13498–503. [PMC free article] [PubMed] [Google Scholar]

44. De Gendt K, Swinnen JV, Saunders PT, Schoonjans L, Dewerchin M, Devos A, et al. A Sertoli cell-selective knockout of the androgen receptor causes spermatogenic arrest in meiosis. Proc Natl Acad Sci U S A. 2004;101:1327–32. [PMC free article] [PubMed] [Google Scholar]

45. Chang C, Chen YT, Yeh SD, Xu Q, Wang RS, Guillou F, et al. Infertility with defective spermatogenesis and hypotestosteronemia in male mice lacking the androgen receptor in Sertoli cells. Proc Natl Acad Sci U S A. 2004;101:6876–81. [PMC free article] [PubMed] [Google Scholar]

46. Stanton PG, Sluka P, Foo CF, Stephens AN, Smith AI, McLachlan RI, et al. Proteomic changes in rat spermatogenesis in response to in vivo androgen manipulation; impact on meiotic cells. PLoS One. 2012;7:e41718. [PMC free article] [PubMed] [Google Scholar]

47. O’Donnell L, McLachlan RI, Wreford NG, de Kretser DM, Robertson DM. Testosterone withdrawal promotes stage-specific detachment of round spermatids from the rat seminiferous epithelium. Biol Reprod. 1996;55:895–901. [PubMed] [Google Scholar]

48. Holdcraft RW, Braun RE. Androgen receptor function is required in Sertoli cells for the terminal differentiation of haploid spermatids. Development. 2004;131:459–67. [PubMed] [Google Scholar]

49. Kopera IA, Bilinska B, Cheng CY, Mruk DD. Sertoli-germ cell junctions in the testis: a review of recent data. Philos Trans R Soc Lond B Biol Sci. 2010;365:1593–605. [PMC free article] [PubMed] [Google Scholar]

50. Wong CH, Xia W, Lee NP, Mruk DD, Lee WM, Cheng CY. Regulation of ectoplasmic specialization dynamics in the seminiferous epithelium by focal adhesion-associated proteins in testosterone-suppressed rat testes. Endocrinology. 2005;146:1192–204. [PubMed] [Google Scholar]

51. Zhang J, Mruk DD, Cheng CY. Myotubularin phosphoinositide phosphatases, protein phosphatases, and kinases: their roles in junction dynamics and spermatogenesis. J Cell Physiol. 2005;204:470–83. [PubMed] [Google Scholar]

52. Zhang J, Wong CH, Xia W, Mruk DD, Lee NP, Lee WM, et al. Regulation of Sertoli-germ cell adherens junction dynamics via changes in protein-protein interactions of the N-cadherin-beta-catenin protein complex which are possibly mediated by c-Src and myotubularin-related protein 2: an in vivo study using an androgen suppression model. Endocrinology. 2005;146:1268–84. [PubMed] [Google Scholar]

53. Wang RS, Yeh S, Chen LM, Lin HY, Zhang C, Ni J, et al. Androgen receptor in Sertoli cell is essential for germ cell nursery and junctional complex formation in mouse testes. Endocrinology. 2006;147:5624–33. [PubMed] [Google Scholar]

54. Roura S, Miravet S, Piedra J, Garcia de Herreros A, Dunach M. Regulation of E-cadherin/Catenin association by tyrosine phosphorylation. J Biol Chem. 1999;274:36734–40. [PubMed] [Google Scholar]

55. Xia W, Cheng CY. TGF-beta3 regulates anchoring junction dynamics in the seminiferous epithelium of the rat testis via the Ras/ERK signaling pathway: An in vivo study. Dev Biol. 2005;280:321–43. [PubMed] [Google Scholar]

56. Kinch MS, Clark GJ, Der CJ, Burridge K. Tyrosine phosphorylation regulates the adhesions of ras-transformed breast epithelia. J Cell Biol. 1995;130:461–71. [PMC free article] [PubMed] [Google Scholar]

57. O’Donnell L, Pratis K, Wagenfeld A, Gottwald U, Muller J, Leder G, et al. Transcriptional profiling of the hormone-responsive stages of spermatogenesis reveals cell-, stage-, and hormone-specific events. Endocrinology. 2009;150:5074–84. [PubMed] [Google Scholar]

58. Shang Y, Myers M, Brown M. Formation of the androgen receptor transcription complex. Mol Cell. 2002;9:601–610. [PubMed] [Google Scholar]

59. Schauwaers K, De Gendt K, Saunders PT, Atanassova N, Haelens A, Callewaert L, et al. Loss of androgen receptor binding to selective androgen response elements causes a reproductive phenotype in a knockin mouse model. Proc Natl Acad Sci U S A. 2007;104:4961–6. [PMC free article] [PubMed] [Google Scholar]

60. Fix C, Jordan C, Cano P, Walker WH. Testosterone activates mitogen-activated protein kinase and the cAMP response element binding protein transcription factor in Sertoli cells. Proc Natl Acad Sci U S A. 2004;101:10919–10924. [PMC free article] [PubMed] [Google Scholar]

61. Cheng J, Watkins SC, Walker WH. Testosterone Activates MAP Kinase Via Src Kinase and the EGF Receptor in Sertoli Cells. Endocrinology. 2007;148:2066–2074. [PubMed] [Google Scholar]

62. Shupe J, Cheng J, Puri P, Kostereva N, Walker WH. Regulation of Sertoli-Germ Cell Adhesion and Sperm Release by FSH and Nonclassical Testosterone Signaling. Mol Endocrinol. 2011;25:238–52. [PMC free article] [PubMed] [Google Scholar]

63. Wang W, WRN, Chapin RE. Rat testicular src: Normal distribution and involvement in ethylene glycol monomethyl ether-induced apoptosis. Toxicology and Applied Pharmacology. 2000;163:125–134. [PubMed] [Google Scholar]

64. Chapin RE, Wine RN, Harris MW, Borchers CH, Haseman JK. Structure and control of a cell-cell adhesion complex associated with spermiation in rat seminiferous epithelium. J Androl. 2001;22:1030–52. [PubMed] [Google Scholar]

65. McCabe MJ, Allan CM, Foo CF, Nicholls PK, McTavish KJ, Stanton PG. Androgen initiates Sertoli cell tight junction formation in the hypogonadal (hpg) mouse. Biol Reprod. 2012;87:38. [PubMed] [Google Scholar]

66. Li JC, Mruk D, Cheng CY. The inter-Sertoli tight junction permeability barrier is regulated by the interplay of protein phosphatases and kinases: an in vitro study. J Androl. 2001;22:847–56. [PubMed] [Google Scholar]

67. Wong CH, Cheng CY. Mitogen-activated protein kinases, adherens junction dynamics, and spermatogenesis: a review of recent data. Dev Biol. 2005;286:1–15. [PubMed] [Google Scholar]

68. Siu MK, Wong CH, Lee WM, Cheng CY. Sertoli-germ cell anchoring junction dynamics in the testis are regulated by an interplay of lipid and protein kinases. J Biol Chem. 2005;280:25029–47. [PubMed] [Google Scholar]

69. Sen A, De Castro I, Defranco DB, Deng FM, Melamed J, Kapur P, et al. Paxillin mediates extranuclear and intranuclear signaling in prostate cancer proliferation. J Clin Invest. 2012;122:2469–81. [PMC free article] [PubMed] [Google Scholar]

70. Sen A, O’Malley K, Wang Z, Raj GV, Defranco DB, Hammes SR. Paxillin regulates androgen- and epidermal growth factor-induced MAPK signaling and cell proliferation in prostate cancer cells. J Biol Chem. 2010;285:28787–95. [PMC free article] [PubMed] [Google Scholar]

71. Rahman F, Christian HC. Non-classical actions of testosterone: an update. Trends Endocrinol Metab. 2007;18:371–8. [PubMed] [Google Scholar]

72. Lim P, Robson M, Spaliviero J, McTavish KJ, Jimenez M, Zajac JD, et al. Sertoli cell androgen receptor DNA binding domain is essential for the completion of spermatogenesis. Endocrinology. 2009;150:4755–65. [PubMed] [Google Scholar]

73. De Gendt K, Verhoeven G. Tissue- and cell-specific functions of the androgen receptor revealed through conditional knockout models in mice. Mol Cell Endocrinol. 2012;352:13–25. [PubMed] [Google Scholar]

74. Zhou Q, Nie R, Prins GS, Saunders PT, Katzenellenbogen BS, Hess RA. Localization of androgen and estrogen receptors in adult male mouse reproductive tract. J Androl. 2002;23:870–81. [PubMed] [Google Scholar]

75. Franca LR, Leal MC, Sasso-Cerri E, Vasconcelos A, Debeljuk L, Russell LD. Cimetidine (Tagamet) is a reproductive toxicant in male rats affecting peritubular cells. Biol Reprod. 2000;63:1403–12. [PubMed] [Google Scholar]

76. Denolet E, De Gendt K, Allemeersch J, Engelen K, Marchal K, Van Hummelen P, et al. The effect of a Sertoli cell-selective knockout of the androgen receptor on testicular gene expression in prepubertal mice. Mol Endocrinol. 2006;20:321–34. [PubMed] [Google Scholar]

77. Welsh M, Saunders PT, Atanassova N, Sharpe RM, Smith LB. Androgen action via testicular peritubular myoid cells is essential for male fertility. FASEB J. 2009;23:4218–30. [PMC free article] [PubMed] [Google Scholar]

78. Welsh M, Moffat L, Belling K, de Franca LR, Segatelli TM, Saunders PT, et al. Androgen receptor signalling in peritubular myoid cells is essential for normal differentiation and function of adult Leydig cells. Int J Androl. 2012;35:25–40. [PubMed] [Google Scholar]

79. Xu Q, Lin HY, Yeh SD, Yu IC, Wang RS, Chen YT, et al. Infertility with defective spermatogenesis and steroidogenesis in male mice lacking androgen receptor in Leydig cells. Endocrine. 2007;32:96–106. [PubMed] [Google Scholar]

80. Smith L. Good planning and serendipity: exploiting the Cre/Lox system in the testis. Reproduction. 2011;141:151–61. [PubMed] [Google Scholar]

81. Jamin SP, Arango NA, Mishina Y, Hanks MC, Behringer RR. Requirement of Bmpr1a for Mullerian duct regression during male sexual development. Nat Genet. 2002;32:408–10. [PubMed] [Google Scholar]

82. O’Hara L, Smith LB. Androgen receptor signalling in Vascular Endothelial cells is dispensable for spermatogenesis and male fertility. BMC Res Notes. 2012;5:16. [PMC free article] [PubMed] [Google Scholar]

83. Welsh M, Sharpe RM, Moffat L, Atanassova N, Saunders PT, Kilter S, et al. Androgen action via testicular arteriole smooth muscle cells is important for Leydig cell function, vasomotion and testicular fluid dynamics. PLoS One. 2010;5:e13632. [PMC free article] [PubMed] [Google Scholar]

84. Collin O, Bergh A, Damber JE, Widmark A. Control of testicular vasomotion by testosterone and tubular factors in rats. J Reprod Fertil. 1993;97:115–21. [PubMed] [Google Scholar]

85. Damber JE, Maddocks S, Widmark A, Bergh A. Testicular blood flow and vasomotion can be maintained by testosterone in Leydig cell-depleted rats. Int J Androl. 1992;15:385–93. [PubMed] [Google Scholar]

86. Franca LR, Parreira GG, Gates RJ, Russell LD. Hormonal regulation of spermatogenesis in the hypophysectomized rat: quantitation of germ-cell population and effect of elimination of residual testosterone after long-term hypophysectomy. J Androl. 1998;19:335–40. [PubMed] [Google Scholar]

87. Willems A, Batlouni SR, Esnal A, Swinnen JV, Saunders PT, Sharpe RM, et al. Selective ablation of the androgen receptor in mouse sertoli cells affects sertoli cell maturation, barrier formation and cytoskeletal development. PLoS One. 2010;5:e14168. [PMC free article] [PubMed] [Google Scholar]

88. Willems A, De Gendt K, Deboel L, Swinnen JV, Verhoeven G. The development of an inducible androgen receptor knockout model in mouse to study the postmeiotic effects of androgens on germ cell development. Spermatogenesis. 2011;1:341–353. [PMC free article] [PubMed] [Google Scholar]

89. Hazra R, Corcoran L, Robson M, McTavish KJ, Upton D, Handelsman DJ, et al. Temporal role of Sertoli cell androgen receptor expression in spermatogenic development. Mol Endocrinol. 2013;27:12–24. [PMC free article] [PubMed] [Google Scholar]

90. Tan KA, De Gendt K, Atanassova N, Walker M, Sharpe RM, Saunders PT, et al. The role of androgens in Sertoli cell proliferation and functional maturation: studies in mice with total or Sertoli cell-selective ablation of the androgen receptor. Endocrinology. 2005;146:2674–83. [PubMed] [Google Scholar]

91. Hazra R, Jimenez M, Desai R, Handelsman DJ, Allan CM. Sertoli cell androgen receptor expression regulates temporal fetal and adult Leydig cell differentiation, function, and population size. Endocrinology. 2013;154:3410–22. [PubMed] [Google Scholar]

92. Zhou Q, Shima JE, Nie R, Friel PJ, Griswold MD. Androgen-regulated transcripts in the neonatal mouse testis as determined through microarray analysis. Biol Reprod. 2005;72:1010–9. [PubMed] [Google Scholar]

93. Sadate-Ngatchou PI, Pouchnik DJ, Griswold MD. Identification of testosterone-regulated genes in testes of hypogonadal mice using oligonucleotide microarray. Mol Endocrinol. 2004;18:422–33. [PubMed] [Google Scholar]

94. O’Shaughnessy PJ, Abel M, Charlton HM, Hu B, Johnston H, Baker PJ. Altered expression of genes involved in regulation of vitamin A metabolism, solute transportation, and cytoskeletal function in the androgen-insensitive tfm mouse testis. Endocrinology. 2007;148:2914–24. [PubMed] [Google Scholar]

95. Zhou W, Wang G, Small CL, Liu Z, Weng CC, Yang L, et al. Gene expression alterations by conditional knockout of androgen receptor in adult Sertoli cells of Utp14b jsd/jsd (jsd) mice. Biol Reprod. 2011;84:400–8. [PMC free article] [PubMed] [Google Scholar]

96. Eacker SM, Shima JE, Connolly CM, Sharma M, Holdcraft RW, Griswold MD, et al. Transcriptional profiling of androgen receptor (AR) mutants suggests instructive and permissive roles of AR signaling in germ cell development. Mol Endocrinol. 2007;21:895–907. [PubMed] [Google Scholar]

97. De Gendt K, Verhoeven G, Amieux PS, Wilkinson MF. The AR-regulated Translome in Sertoli Cells Defined Using the RiboTag Approach. Mol Endocrinol. 2014 In Press. [PMC free article] [PubMed] [Google Scholar]

98. Morales C, Griswold MD. Retinol-induced stage synchronization in seminiferous tubules of the rat. Endocrinology. 1987;121:432–4. [PubMed] [Google Scholar]

99. Dann CT, Alvarado AL, Molyneux LA, Denard BS, Garbers DL, Porteus MH. Spermatogonial stem cell self-renewal requires OCT4, a factor downregulated during retinoic acid-induced differentiation. Stem Cells. 2008;26:2928–37. [PubMed] [Google Scholar]

100. Sugimoto R, Nabeshima Y, Yoshida S. Retinoic acid metabolism links the periodical differentiation of germ cells with the cycle of Sertoli cells in mouse seminiferous epithelium. Mech Dev. 2012;128:610–24. [PubMed] [Google Scholar]

101. O’Rand GM, Widgren EE, Sivashanmugam P, Richardson RT, Hall SH, French FS, et al. Reversible immunocontraception in male monkeys immunized with eppin. Science. 2004;306:1189–90. [PubMed] [Google Scholar]

102. Uhrin P, Dewerchin M, Hilpert M, Chrenek P, Schofer C, Zechmeister-Machhart M, et al. Disruption of the protein C inhibitor gene results in impaired spermatogenesis and male infertility. J Clin Invest. 2000;106:1531–9. [PMC free article] [PubMed] [Google Scholar]

103. Takamiya K, Yamamoto A, Furukawa K, Zhao J, f*ckumoto S, Yamashiro S, et al. Complex gangliosides are essential in spermatogenesis of mice: possible roles in the transport of testosterone. Proc Natl Acad Sci U S A. 1998;95:12147–52. [PMC free article] [PubMed] [Google Scholar]

104. Sandhoff R, Geyer R, Jennemann R, Paret C, Kiss E, Yamash*ta T, et al. Novel class of glycosphingolipids involved in male fertility. J Biol Chem. 2005;280:27310–8. [PubMed] [Google Scholar]

105. Rabionet M, van der Spoel AC, Chuang CC, von Tumpling-Radosta B, Litjens M, Bouwmeester D, et al. Male germ cells require polyenoic sphingolipids with complex glycosylation for completion of meiosis: a link to ceramide synthase-3. J Biol Chem. 2008;283:13357–69. [PMC free article] [PubMed] [Google Scholar]

106. Andersen OM, Yeung CH, Vorum H, Wellner M, Andreassen TK, Erdmann B, et al. Essential role of the apolipoprotein E receptor-2 in sperm development. J Biol Chem. 2003;278:23989–95. [PubMed] [Google Scholar]

107. Maclean JA, 2nd, Chen MA, Wayne CM, Bruce SR, Rao M, Meistrich ML, et al. Rhox: a new homeobox gene cluster. Cell. 2005;120:369–82. [PubMed] [Google Scholar]

108. Maclean JA, 2nd, Hu Z, Welborn JP, Song HW, Rao MK, Wayne CM, et al. The RHOX Homeodomain Proteins Regulate the Expression of Insulin and Other Metabolic Regulators in the Testis. J Biol Chem. 2013;288:34809–25. [PMC free article] [PubMed] [Google Scholar]

109. Sanz E, Yang L, Su T, Morris DR, McKnight GS, Amieux PS. Cell-type-specific isolation of ribosome-associated mRNA from complex tissues. Proc Natl Acad Sci U S A. 2009;106:13939–44. [PMC free article] [PubMed] [Google Scholar]

110. Sanz E, Evanoff R, Quintana A, Evans E, Miller JA, Ko C, et al. RiboTag analysis of actively translated mRNAs in Sertoli and Leydig cells in vivo. PLoS One. 2013;8:e66179. [PMC free article] [PubMed] [Google Scholar]

111. Panneerdoss S, Chang YF, Buddavarapu KC, Chen HI, Shetty G, Wang H, et al. Androgen-responsive microRNAs in mouse Sertoli cells. PLoS One. 2012;7:e41146. [PMC free article] [PubMed] [Google Scholar]

The Regulation of Spermatogenesis by Androgens (2024)

FAQs

The Regulation of Spermatogenesis by Androgens? ›

Testosterone is the major androgen in the testis that regulates spermatogenesis. Testosterone is produced by the Leydig cell

Leydig cell
Leydig cells are the testicular cells responsible for the biosynthesis and secretion of androgens, which are critical for the development of the reproductive tract and for reproductive function in the male.
https://www.sciencedirect.com › topics › leydig-cell
in response to stimulation with luteinizing hormone (LH) and acts as a paracrine factor that diffuses into the seminiferous tubules
seminiferous tubules
The seminiferous tubule is the functional unit in the testis that produces spermatozoa (haploid, 1n) from spermatogonia (diploid, 2n) during spermatogenesis. From: Molecular and Cellular Endocrinology, 2010.
https://www.sciencedirect.com › topics › seminiferous-tubule
.

What hormone regulates spermatogenesis? ›

Sertoli cells have receptors for follicle stimulating hormone (FSH) and testosterone which are the main hormonal regulators of spermatogenesis. Hormones such as testosterone, FSH and luteinizing hormone (LH) are known to influence the germ cell fate.

What are the stages of spermatogenesis and its regulation? ›

Spermatogenesis starts at puberty, when the Leydig cells in the testes start to produce androgens under the influence of the Follicle-Stimulating Hormone (FSH) and the Luteinizing Hormone (LH), which are in turn controlled by the Gonadotrophin-Releasing Hormone (GnRH) produced by the hypothalamus.

What does androgen stimulate the process of? ›

The hormone which acts on Sertoli cells and stimulate the process of spermiogenesis is. Androgen.

How does spermatogenesis work? ›

As mentioned above, spermatogenesis is the process by which sperm cell production occurs; the germ cells give rise to the haploid spermatozoa. Sperm production takes place inside the seminiferous tubules, which is a convoluted cluster of tubes located inside the testes.

What cell regulates spermatogenesis? ›

Normal spermatogenesis relies on Sertoli cells, which preserve cell junctions while providing nutrients for mitosis and meiosis of male germ cells. Several genes regulate normal spermatogenesis, some of which are not exclusively expressed in the testis and control multiple physiological processes in an organism.

Which hormone suppresses spermatogenesis? ›

the follicle-stimulating hormone FSH receptor gene present variable suppression of spermatogenesis and fertility.

Do androgens stimulate the process of spermatogenesis? ›

Testosterone is the major androgen in the testis that regulates spermatogenesis. Testosterone is produced by the Leydig cell in response to stimulation with luteinizing hormone (LH) and acts as a paracrine factor that diffuses into the seminiferous tubules.

What is the role of androgens in males? ›

Androgens are crucial for male sexual and reproductive function. They are also responsible for the development of secondary sexual characteristics in men, including facial and body hair growth and voice change. Androgens also affect bone and muscle development and metabolism.

How to regulate androgen hormones? ›

Anti-androgen medication may be prescribed for certain conditions to block the effects of androgens. Meanwhile, androgen therapy, also called testosterone replacement therapy (TRT), may help treat low androgen levels.

What are the factors controlling spermatogenesis? ›

For instance, testosterone production by the hypothalamus, Leydig cells, and pituitary gland is crucial for this process. Spermatogenesis is also sensitive to temperature fluctuations, dietary deficiencies, alcohol consumption, drug exposure, and disease presence, all of which can adversely affect sperm production.

What is spermatogenesis in very short answer? ›

Spermatogenesis is the process of sperm cell development. Rounded immature sperm cells undergo successive mitotic and meiotic divisions (spermatocytogenesis) and a metamorphic change (spermiogenesis) to produce spermatozoa.

What is the end result of spermatogenesis? ›

Spermatogenesis produces mature male gametes, commonly called sperm but more specifically known as spermatozoa, which are able to fertilize the counterpart female gamete, the oocyte, during conception to produce a single-celled individual known as a zygote.

What is the hormone that promotes spermatogenesis along? ›

Follicle-Stimulating Hormone (FSH)

Follicle-stimulating hormone (FSH) is a glycoprotein regulating development and reproduction. In both adult fertile males and females, FSH mediates spermatogenesis and folliculogenesis, acting through its G-protein coupled receptor (FSHR).

Does estrogen regulate spermatogenesis? ›

In the testes, spermatogenesis is modulated at every level by estrogen, starting with the hypothalamus-pituitary-gonadal axis, followed by the Leydig, Sertoli, and germ cells, and finishing with the ductal epithelium, epididymis, and mature sperm.

Which hormones facilitate spermatogenesis and testosterone production? ›

Male Hormones

FSH enters the testes and stimulates the Sertoli cells to begin facilitating spermatogenesis using negative feedback, as illustrated in Figure 1. LH also enters the testes and stimulates the interstitial cells of Leydig to make and release testosterone into the testes and the blood.

What hormones are released in spermatogenesis? ›

Hormones and the Endocrine System
Where the hormone is producedHormone(s) secreted
Testes (testicl*s)Testosterone
Pineal glandMelatonin
HypothalamusGrowth hormone releasing hormone (GHRH)
HypothalamusThyrotropin releasing hormone (TRH)
22 more rows

Top Articles
Latest Posts
Article information

Author: Foster Heidenreich CPA

Last Updated:

Views: 5820

Rating: 4.6 / 5 (56 voted)

Reviews: 95% of readers found this page helpful

Author information

Name: Foster Heidenreich CPA

Birthday: 1995-01-14

Address: 55021 Usha Garden, North Larisa, DE 19209

Phone: +6812240846623

Job: Corporate Healthcare Strategist

Hobby: Singing, Listening to music, Rafting, LARPing, Gardening, Quilting, Rappelling

Introduction: My name is Foster Heidenreich CPA, I am a delightful, quaint, glorious, quaint, faithful, enchanting, fine person who loves writing and wants to share my knowledge and understanding with you.