Skip to main content Accessibility help
×
Hostname: page-component-76fb5796d-9pm4c Total loading time: 0 Render date: 2024-04-28T05:00:13.825Z Has data issue: false hasContentIssue false

Section 1 - Scientific Foundations of Male Infertility

Published online by Cambridge University Press:  08 July 2023

Larry I. Lipshultz
Affiliation:
Baylor College of Medicine, Texas
Stuart S. Howards
Affiliation:
University of Virginia
Craig S. Niederberger
Affiliation:
University of Illinois, Chicago
Dolores J. Lamb
Affiliation:
Weill Cornell Medical College, New York
Get access
Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2023

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Further Reading

Wein, AJ, Kavoussi, LR, Novick, AC, Partin, AW, Peters, CA (eds). Campbell-Walsh Urology, 10th ed. Saint Louis, MO: Elsevier, 2011.Google Scholar
Moore, KL, Persaud, TVN, Torchia, MG. The Developing Human: Clinically Oriented Embryology, 11th ed. Philadelphia, PA: Saunders, 2020.Google Scholar

References

Moore, KL, Persaud, TVN, Torchia, MG. The Developing Human: Clinically Oriented Embryology, 11th ed. Philadelphia, PA: Saunders, 2020.Google Scholar
Ochsner, MG, Brannan, W, Goodier, EH. Absent vas deferens associated with renal agenesis. JAMA 1972;222:1055–6.CrossRefGoogle ScholarPubMed
McCallum, T, Milunsky, J, Munarriz, R, Carson, R, Sadeghi-Nejad, H, Oates, R. Unilateral renal agenesis associated with congenital bilateral absence of the vas deferens: phenotypic findings and genetic considerations. Hum Reprod 2001;16:282–8.CrossRefGoogle ScholarPubMed
Holt, SA, Peterson, NE. Ectopia of seminal vesicle. Associated with agenesis of ipsilateral kidney. Urology 1974;4:322–4.Google Scholar
Wein, AJ, Kavoussi, LR, Novick, AC, Partin, AW, Peters, CA (eds). Campbell-Walsh Urology, 10th ed. Saint Louis, MO: Elsevier, 2011.Google Scholar
Nievelstein, RA, van der Werff, JF, Verbeek, FJ, Valk, J, Vermeij-Keers, C. Normal and abnormal embryonic development of the anorectum in human embryos. Teratology 1998;57:70–8.Google Scholar
Nebot-Cegarra, J, Fàbregas, PJ, Sánchez-Pérez, I. Cellular proliferation in the urorectal septation complex of the human embryo at Carnegie stages 13–18: a nuclear area-based morphometric analysis. J Anat 2005;207:353–64.Google Scholar
Liaw, A, Cunha, GR, Shen, J, et al. Development of the human bladder and ureterovesical junction. Differentiation 2018;103:6673.CrossRefGoogle ScholarPubMed
Viana, R, Batourina, E, Huang, H, et al. The development of the bladder trigone, the center of the anti-reflux mechanism. Development 2007;134:3763–9.CrossRefGoogle Scholar
Brewster, SF. The development and differentiation of human seminal vesicles. J Anat 1985;143:4555.Google Scholar
Thomson, AA, Marker, PC. Branching morphogenesis in the prostate gland and seminal vesicles. Differentiation 2006;74:382–92.Google Scholar
Owen, DH, Katz, DF. A review of the physical and chemical properties of human semen and the formulation of a semen simulant. J Androl 2005;26:459–69.Google Scholar
Huang, L, Pu, Y, Alam, S, Birch, L, Prins, GS. Estrogenic regulation of signaling pathways and homeobox genes during rat prostate development. J Androl 2004;25:330–7.CrossRefGoogle ScholarPubMed
Prins, GS, Tang, WY, Belmonte, J, Ho, SM. Perinatal exposure to oestradiol and bisphenol A alters the prostate epigenome and increases susceptibility to carcinogenesis. Basic Clin Pharmacol Toxicol 2008;102:134–8.Google Scholar
Rafailidis, V, Varelas, S, Apostolopoulou, F, Rafailidis, D. Nonobliteration of the processus vaginalis. J Ultrasound Med 2016;35:805–18.Google Scholar
Kolon, TF, Herndon, CD, Baker, LA, et al. Evaluation and treatment of cryptorchidism: AUA guideline. J Urol 2014;192:337–45.Google Scholar
Nachtigal, MW, Hirokawa, Y, Enyeart-VanHouten, DL, Flanagan, JN, Hammer, GD, Ingraham, HA. Wilms’ tumor 1 and Dax-1 modulate the orphan nuclear receptor SF-1 in sex-specific gene expression. Cell 1998;93:445–54.CrossRefGoogle ScholarPubMed
Sekido, R, Bar, I, Narváez, V, Penny, G, Lovell-Badge, R. SOX9 is up-regulated by the transient expression of SRY specifically in Sertoli cell precursors. Dev Biol 2004;274:271–9.CrossRefGoogle ScholarPubMed
Colvin, JS, Green, RP, Schmahl, J, Capel, B, Ornitz, DM. Male-to-female sex reversal in mice lacking fibroblast growth factor 9. Cell 2001;104:875–89.Google Scholar
Tevosian, SG, Albrecht, KH, Crispino, JD, Fujiwara, Y, Eicher, EM, Orkin, SH. Gonadal differentiation, sex determination and normal Sry expression in mice require direct interaction between transcription partners GATA4 and FOG2. Development 2002;129:4627–34.Google Scholar
Nef, S, Verma-Kurvari, S, Merenmies, J, et al. Testis determination requires insulin receptor family function in mice. Nature 2003;426:291–5.Google Scholar
Brennan, J, Tilmann, C, Capel, B. Pdgfr-α mediates testis cord organization and fetal Leydig cell development in the XY gonad. Genes Dev 2003;17:800–10.Google Scholar
Glenister, TW. The origin and fate of the urethral plate in man. J Anat 1954;88:413–25.Google Scholar
Hadidi, AT, Roessler, J, Coerdt, W. Development of the human male urethra: a histochemical study on human embryos. J Pediatr Surg 2014;49:1146–52.Google Scholar
Li, Y, Sinclair, A, Cao, M, et al. Canalization of the urethral plate precedes fusion of the urethral folds during male penile urethral development: the double zipper hypothesis. J Urol 2015;193:1353–9.CrossRefGoogle ScholarPubMed

References

Hermo, L, Oliveira, RL, Smith, CE, Au, CE, Bergeron, JJM. Golgi apparatus regulation of differentiation: a case study for germ cells of the rat testis. In: Cheng, CY, ed. Spermatogenesis: Biology and Clinical Implications. London: CRC Press, 2019; pp. 139.Google Scholar
Hermo, L, Pelletier, RM, Cyr, DG, Smith, CE. Surfing the wave, cycle, life history, and genes/proteins expressed by testicular germ cells. Part 5: intercellular junctions and contacts between germ cells and Sertoli cells and their regulatory interactions, testicular cholesterol, and genes/proteins associated with more than one germ cell generation. Microsc Res Tech 2010;73:409–94.Google Scholar
Hermo, L, Pelletier, RM, Cyr, DG, Smith, CE. Surfing the wave, cycle, life history, and genes/proteins expressed by testicular germ cells. Part 1: background to spermatogenesis, spermatogonia, and spermatocytes. Microsc Res Tech 2010;73:241–78.Google Scholar
Hermo, L, Pelletier, RM, Cyr, DG, Smith, CE. Surfing the wave, cycle, life history, and genes/proteins expressed by testicular germ cells. Part 2: changes in spermatid organelles associated with development of spermatozoa. Microsc Res Tech 2010;73:279319.Google Scholar
de Kretser, DM, Kerr, JB. The cytology of the testis. In: Knobil, E, Neill, JB, Ewing, LL, Greenwald, GS, Markert, CL, Pfaff, DW, eds. The Physiology of Reproduction, Vol. 1. New York, NY: Raven Press, 1988; pp. 837932.Google Scholar
O’Donnell, L. Mechanisms of spermiogenesis and spermiation and how they are disturbed. Spermatogenesis 2014;4:e979623.CrossRefGoogle ScholarPubMed
O’Donnell, L, O’Bryan, MK. Microtubules and spermatogenesis. Semin Cell Dev Biol 2014;30:4554.Google Scholar
Hess, RA, de Franca, LR. Spermatogenesis and cycle of the seminiferous epithelium. Adv Exp Med Biol 2008;636:115.Google ScholarPubMed
Griswold, MD. 50 years of spermatogenesis: Sertoli cells and their interactions with germ cells. Biol Reprod 2018;99:87100.Google Scholar
Griswold, MD. Spermatogenesis: the commitment to meiosis. Physiol Rev 2016;96:117.Google Scholar
McCarrey, J. Development of the germ cell. In: Desjardins, C, Ewing, L, eds. Cell and Molecular Biology of the Testis. New York, NY: Oxford University Press, 1993; pp. 5889.Google Scholar
Amann, RP. The cycle of the seminiferous epithelium in humans: a need to revisit? J Androl 2008;29:469–87.Google Scholar
Muciaccia, B, Boitani, C, Berloco, BP, et al. Novel stage classification of human spermatogenesis based on acrosome development. Biol Reprod 2013;89:60.Google Scholar
Vogl, AW, Vaid, KS, Guttman, JA. The Sertoli cell cytoskeleton. Adv Exp Med Biol 2008;636:186211.Google Scholar
Russell, LD, Peterson, RN. Sertoli cell junctions: morphological and functional correlates. Int Rev Cytol 1985;94:177211.CrossRefGoogle ScholarPubMed
Wong, EWP, Mruk, DD, Cheng, CY. Biology and regulation of ectoplasmic specialization, an atypical adherens junction type, in the testis. Biochem Biophys Acta 2008;1778:692708.CrossRefGoogle ScholarPubMed
Mruk, DD, Cheng, CY. Cell–cell interactions at the ectoplasmic specialization in the testis. Trends Endocrinol Metab 2004;15:439–47.Google Scholar
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;25:747806.CrossRefGoogle ScholarPubMed
Wong, EWP, Cheng, CY. Polarity proteins and cell–cell interactions in the testis. Int Rev Cell Mol Biol 2009;278:309–53.Google Scholar
Assemat, E, Bazellieres, E, Pallesi-Pocachard, E, Le Bivic, A, Massey-Harroche, D. Polarity complex proteins. Biochim Biophys Acta 2008;1778:614–30.Google Scholar
Iden, S, Collard, JG. Crosstalk between small GTPases and polarity proteins in cell polarization. Nat Rev Mol Cell Biol 2008;9:846–59.Google Scholar
Wong, EWP, Mruk, DD, Lee, WM, Cheng, CY. Par3/Par6 polarity complex coordinates apical ectoplasmic specialization and blood–testis barrier restructuring during spermatogenesis. Proc Natl Acad Sci U S A 2008;105:9657–62.Google Scholar
Wong, EWP, Sun, S, Li, MWM, Lee, WM, Cheng, CY. 14-3-3 protein regulates cell adhesion in the seminiferous epithelium of rat testes. Endocrinology 2009;150:4713–23.Google Scholar
Gao, Y, Lui, WY, Lee, WM, Cheng, CY. Polarity protein Crumbs homolog-3 (CRB3) regulates ectoplasmic specialization dynamics through its action on F-actin organization in Sertoli cells. Sci Rep 2016;6:28589.Google Scholar
Su, WH, Wong, EWP, Mruk, DD, Cheng, CY. The Scribble/Lgl/Dlg polarity protein complex is a regulator of blood–testis barrier dynamics and spermatid polarity during spermatogenesis. Endocrinology 2012;153:6041–53.Google Scholar
Chen, H, Xiao, X, Lui, WY, Lee, WM, Cheng, CY. Vangl2 regulates spermatid planar cell polarity through microtubule (MT)-based cytoskeleton in the rat testis. Cell Death Dis 2018;9:340.CrossRefGoogle ScholarPubMed
Chen, H, Mruk, DD, Lee, WM, Cheng, CY. Regulation of spermatogenesis by a local functional axis in the testis: role of the basement membrane-derived noncollagenous 1 domain peptide. FASEB J 2017;31:3587–607.Google Scholar
Montcouquiol, M, Kelley, MW. Development and patterning of the cochlea: from convergent extension to planar polarity. Cold Spring Harb Perspect Med 2020;10:a033266.Google Scholar
Tarchini, B, Lu, X. New insights into regulation and function of planar polarity in the inner ear. Neurosci Lett 2019;709:134373.Google Scholar
Axelrod, JD. Planar cell polarity signaling in the development of left–right asymmetry. Curr Opin Cell Biol 2020;62:61–9.Google Scholar
Sutherland, A, Keller, R, Lesko, A. Convergent extension in mammalian morphogenesis. Semin Cell Dev Biol 2020;100:199211.CrossRefGoogle ScholarPubMed
Fisher, KH, Strutt, D. A theoretical framework for planar polarity establishment through interpretation of graded cues by molecular bridges. Development 2019;146:dev168955.CrossRefGoogle ScholarPubMed
Chen, H, Mruk, DD, Lee, WM, Cheng, CY. Planar cell polarity (PCP) protein Vangl2 regulates ectoplasmic specialization dynamics via its effects on actin microfilaments in the testes of male rats. Endocrinology 2016;157:2140–59.Google Scholar
Li, L, Mao, B, Yan, M, et al. Planar cell polarity protein Dishevelled 3 (Dvl3) regulates ectoplasmic specialization (ES) dynamics in the testis through changes in cytoskeletal organization. Cell Death Dis 2019;10:194.Google Scholar
Parvinen, M. Regulation of the seminiferous epithelium. Endocr Rev 1982;3:404–17.Google Scholar
Clermont, Y. Kinetics of spermatogenesis in mammals: seminiferous epithelium cycle and spermatogonial renewal. Physiol Rev 1972;52:198235.Google Scholar
Clermont, Y. The cycle of the seminiferous epithelium in man. Am J Anat 1963;112:3551.CrossRefGoogle ScholarPubMed
Leblond, CP, Clermont, Y. Spermiogenesis of rat, mouse, hamster, and guinea pig as revealed by the periodic acid-fuchin sulfurous acid technique. Am J Anat 1952;90:167210.Google Scholar
Hess, RA. Quantitative and qualitative characteristics of the stages and transitions in the cycle of the rat seminiferous epithelium: light microscopic observations of perfusion-fixed and plastic-embedded testes. Biol Reprod 1990;43:525–42.Google Scholar
Xiao, X, Mruk, DD, Wong, CKC, Cheng, CY. Germ cell transport across the seminiferous epithelium during spermatogenesis. Physiology 2014;29:286–98.CrossRefGoogle ScholarPubMed
Wong, V, Russell, LD. Three-dimensional reconstruction of a rat stage V Sertoli cell: I. Methods, basic configuration, and dimensions. Am J Anat 1983;167:143–61.CrossRefGoogle Scholar
Russell, LD, Tallon-Doran, M, Weber, JE, Wong, V, Peterson, RN. Three-dimensional reconstruction of a rat stage V Sertoli cell: III. A study of specific cellular relationships. Am J Anat 1983;167:181–92.Google Scholar
Weber, JE, Russell, LD, Wong, V, Peterson, RN. Three dimensional reconstruction of a rat stage V Sertoli cell: II. Morphometry of Sertoli–Sertoli and Sertoli–germ cell relationships. Am J Anat 1983;167:163–79.Google Scholar
Vaidžiulytė, K, Coppey, M, Schauer, K. Intracellular organization in cell polarity – placing organelles into the polarity loop. J Cell Sci 2019;132:jcs230995.Google Scholar
Riga, A, Castiglioni, VG, Boxem, M. New insights into apical-basal polarization in epithelia. Curr Opin Cell Biol 2020;62:18.Google Scholar
Bazellieres, E, Aksenova, V, Barthelemy-Requin, M, Massey-Harroche, D, Le Bivic, A. Role of the Crumbs proteins in ciliogenesis, cell migration and actin organization. Semin Cell Dev Biol 2018;81:1320.Google Scholar
Peglion, F, Goehring, NW. Switching states: dynamic remodelling of polarity complexes as a toolkit for cell polarization. Curr Opin Cell Biol 2019;60:121–30.Google Scholar
Montcouguiol, M, Kelley, MW. Development and patterning of the cochlea: From convergent extension to planar polarity. Cold Spring Harb Perspect Med 2020;10:a033266.CrossRefGoogle Scholar
Bonello, TT, Peifer, M. Scribble: a master scaffold in polarity, adhesion, synaptogenesis, and proliferation. J Cell Biol 2019;218:742–56.Google Scholar
Chen, H, Mruk, DD, Lui, WY, Wong, CKC, Lee, WM, Cheng, CY. Cell polarity and planar cell polarity (PCP) in spermatogenesis. Semin Cell Dev Biol 2018;81:71–7.CrossRefGoogle ScholarPubMed
Aw, WY, Heck, BW, Joyce, B, Devenport, D. Transient tissue-scale deformation coordinates alignment of planar cell polarity junctions in the mammalian skin. Curr Biol 2016;26:2090–100.CrossRefGoogle ScholarPubMed
Satir, P. Chirality of the cytoskeleton in the origins of cellular asymmetry. Philos Trans R Soc Lond B Biol Sci 2016;371:20150408.Google Scholar
Taylor, J, Abramova, N, Charlton, J, Adler, PN. Van Gogh: a new Drosophila tissue polarity gene. Genetics 1998;150:199210.CrossRefGoogle Scholar
Aw, WY, Devenport, D. Planar cell polarity: global inputs establishing cellular asymmetry. Curr Opin Cell Biol 2017;44:110–16.Google Scholar
Goodrich, LV, Strutt, D. Principles of planar polarity in animal development. Development 2011;138:1877–92.Google Scholar
Xie, Y, Miao, H, Blankenship, JT. Membrane trafficking in morphogenesis and planar polarity. Traffic 2018. doi: 10.1111/tra.12580.CrossRefGoogle Scholar
Li, L, Chen, H, Lian, QQ, Ge, RS, Cheng, CY. Does planar cell polarity matter during spermatogenesis? In: Cheng, CY, ed. Spermatogenesis: Biology and Clinical Implications. London: CRC Press/Taylor & Francis Group, 2018; pp. 211–19.Google Scholar
Park, TJ, Haigo, SL, Wallingford, JB. Ciliogenesis defects in embryos lacking inturned or fuzzy function are associated with failure of planar cell polarity and Hedgehog signaling. Nat Genet 2006;38:303–11.CrossRefGoogle ScholarPubMed
Sokol, SY. Spatial and temporal aspects of Wnt signaling and planar cell polarity during vertebrate embryonic development. Semin Cell Dev Biol 2015;42:7885.Google Scholar
Goffinet, AM, Tissir, F. Seven pass cadherins CELSR1–3. Semin Cell Dev Biol 2017;69:102–10.Google Scholar
May-Simera, H, Kelley, MW. Planar cell polarity in the inner ear. Curr Top Dev Biol 2012;101:111–40.Google Scholar
Humphries, AC, Mlodzik, M. From instruction to output: Wnt/PCP signaling in development and cancer. Curr Opin Cell Biol 2018;51:110–16.Google Scholar
Yang, Y, Mlodzik, M. Wnt-Frizzled/planar cell polarity signaling: cellular orientation by facing the wind (Wnt). Annu Rev Cell Dev Biol 2015;31:623–46.Google Scholar
Li, L, Chen, H, Lian, Q, Ge, RS, Cheng, CY. Does planar cell polarity matter during spermatogenesis? In: Cheng, CY, ed. Spermatogenesis: Biology and Clinical Implications. London: CRC Press/Taylor & Francis Group, 2018; pp. 211–19.Google Scholar
White, JJ, Mazzeu, JF, Hoischen, A, et al. DVL3 alleles resulting in a −1 frameshift of the last exon mediate autosomal-dominant Robinow syndrome. Am J Hum Genet 2016;98:553–61.Google Scholar
White, JJ, Mazzeu, JF, Coban-Akdemir, Z, et al. WNT signaling perturbations underlie the genetic heterogeneity of Robinow syndrome. Am J Hum Genet 2018;102:2743.Google Scholar
Horbelt, CV. Robinow syndrome, Cockayne syndrome, and Pfeiffer syndrome: an overview of physical, neurological, and oral characteristics. Gen Dent 2010;58:1417.Google Scholar
Lei, YP, Zhang, T, Li, H, Wu, BL, Jin, L, Wang, HY. VANGL2 mutations in human cranial neural-tube defects. N Engl J Med 2010;362:2232–5.CrossRefGoogle ScholarPubMed
Hermo, L, Pelletier, RM, Cyr, DG, Smith, CE. Surfing the wave, cycle, life history, and genes/proteins expressed by testicular germ cells. Part 4: intercellular bridges, mitochondria, nuclear envelope, apoptosis, ubiquitination, membrane/voltage-gated channels, methylation/acetylation, and transcription factors. Microsc Res Tech 2010;73:364408.Google Scholar
Dym, M. Basement membrane regulation of Sertoli cells. Endocr Rev 1994;15:102–15.Google Scholar
Siu, MKY, Cheng, CY. Dynamic cross-talk between cells and the extracellular matrix in the testis. Bioessays 2004;26:978–92.Google Scholar
Koch, M, Olson, PF, Albus, A, et al. Characterization and expression of the laminin γ3 chain: a novel, non-basement membrane-associated, laminin chain. J Cell Biol 1999;145:605–18.Google Scholar
Yan, HHN, Cheng, CY. Laminin α3 forms a complex with β3 and γ3 chains that serves as the ligand for α6β1-integrin at the apical ectoplasmic specialization in adult rat testes. J Biol Chem 2006;281:17286–303.Google Scholar
Siu, MKY, Cheng, CY. Interactions of proteases, protease inhibitors, and the β1 integrin/laminin γ3 protein complex in the regulation of ectoplasmic specialization dynamics in the rat testis. Biol Reprod 2004;70:945–64.Google Scholar
Palombi, F, Salanova, M, Tarone, G, Farini, D, Stefanini, M. Distribution of β1 integrin subunit in rat seminiferous epithelium. Biol Reprod 1992;47:1173–82.Google Scholar
Salanova, M, Stefanini, M, De Curtis, I, Palombi, F. Integrin receptor α6β1 is localized at specific sites of cell-to-cell contact in rat seminiferous epithelium. Biol Reprod 1995;52:7987.Google Scholar
Salanova, M, Ricci, G, Boitani, C, Stefanini, M, De Grossi, S, Palombi, F. Junctional contacts between Sertoli cells in normal and aspermatogenic rat seminiferous epithelium contain α6β1 integrins, and their formation is controlled by follicle-stimulating hormone. Biol Reprod 1998;58:371–8.Google Scholar
Su, L, Mruk, DD, Lie, PPY, Silvestrini, B, Cheng, CY. A peptide derived from laminin-γ3 reversibly impairs spermatogenesis in rats. Nat Commun 2012;3:1185.Google Scholar
Gao, Y, Mruk, DD, Lui, WY, Lee, WM, Cheng, CY. F5-peptide induces aspermatogenesis by disrupting organization of actin- and microtubule-based cytoskeletons in the testis. Oncotarget 2016;7:64203–20.Google Scholar
Lie, PPY, Mruk, DD, Mok, KW, Su, L, Lee, WM, Cheng, CY. Focal adhesion kinase-Tyr407 and -Tyr397 exhibit antagonistic effects on blood–testis barrier dynamics in the rat. Proc Natl Acad Sci U S A 2012;109:12562–7.Google Scholar
Chen, H, Gao, Y, Mruk, DD, et al. Rescue of PFOS-induced human Sertoli cell injury by overexpressing a p-FAK-Y407E phosphomimetic mutant. Sci Rep 2017;7:15810.Google Scholar
Siu, MKY, Cheng, CY. Extracellular matrix: recent advances on its role in junction dynamics in the seminiferous epithelium during spermatogenesis. Biol Reprod 2004;71:375–91.Google Scholar
Timpl, R, Brown, J. Supramolecular assembly of basement membranes. Bioessays 1996;18:123–32.Google Scholar
Timpl, R, Wiedemann, H, van Delden, V, Furthmayr, H, Kuhn, K. A network model for the organization of type IV collagen molecules in basement membranes. Eur J Biochem 1989;120:203–11.Google Scholar
Hudson, BG, Reeders, ST, Tryggvason, K. Type IV collagen: structure, gene organization, and role in human diseases. Molecular basis of Goodpasture and Alport syndromes and diffuse leiomyomatosis. J Biol Chem 1993;268:26033–6.Google Scholar
Prockop, DJ, Kivirikko, KI. Collagens: molecular biology, diseases, and potentials for therapy. Annu Rev Biochem 1995;64:403–34.Google Scholar
Gatseva, A, Sin, YY, Brezzo, G, Van Agtmael, T. Basement membrane collagens and disease mechanisms. Essays Biochem 2019;63:297312.Google Scholar
Oh, SP, Warman, ML, Seldin, MF, et al. Cloning of cDNA and genomic DNA encoding human type XVIII collagen and localization of the α1 (XVIII) collagen gene to mouse chromosome 10 and human chromosome 21. Genomics 1994;19:494–9.Google Scholar
Davis, CM, Papadopoulos, V, Sommers, CL, Kleinman, HK, Dym, M. Differential expression of extracellular matrix components in rat Sertoli cells. Biol Reprod 1990;43:860–9.Google Scholar
Enders, GC, Kahsai, TZ, Lian, G, Funabiki, K, Killen, PD, Hudson, BG. Developmental changes in seminiferous tubule extracellular matrix components of the mouse testis: α3 (IV) collagen chain expressed at the initiation of spermatogenesis. Biol Reprod 1995;53:1489–99.Google Scholar
Fröjdman, K, Pelliniemi, LJ, Virtanen, I. Differential distribution of type IV collagen chains in the developing rat testis and ovary. Differentiation 1998;63:125–30.Google Scholar
Miner, JH, Sanes, RJ. Molecular and functional defects in kidneys of mice lacking collagen α3(IV): implications for Alport syndrome. J Cell Biol 1996;135:1402–13.Google Scholar
Kahsai, TZ, Enders, GC, Gunwar, S, et al. Seminiferous tubule basement membrane. Composition and organization of type IV collagen chains, and the linkage of α3(IV) and α5(IV) chains. J Biol Chem 1997;272:17023–32.Google Scholar
Gunwar, S, Ballester, F, Noelken, ME, Sado, Y, Ninomiya, Y, Hudson, BG. Glomerular basement membrane. Identification of a novel disulfide-cross-linked network of α3, α4, and α5 chains of type IV collagen and its implications for the pathogenesis of Alport syndrome. J Biol Chem 1998;273:8767–75.Google Scholar
Miner, JH, Sanes, RJ. A network model for the organization of type IV collagen molecules in basement membranes. J Cell Biol 1994;127:879–91.Google Scholar
Kalluri, R, Cosgrove, D. Assembly of type IV collagen. Insights from α3(IV) collagen-deficient mice. J Biol Chem 2000;275:12719–24.Google Scholar
Mok, KW, Mruk, DD, Lee, WM, Cheng, CY. A study to assess the assembly of a functional blood–testis barrier in developing rat testes. Spermatogenesis 2011;1:270–80.Google Scholar
Richardson, LL, Kleinman, HK, Dym, M. Basement membrane gene expression by Sertoli and peritubular myoid cells in vitro in the rat. Biol Reprod 1995;52:320–30.Google Scholar
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.Google Scholar
Siu, MKY, Lee, WM, Cheng, CY. The interplay of collagen IV, tumor necrosis factor-α, gelatinase B (matrix metalloprotease-9), and tissue inhibitor of metalloprotease-1 in the basal lamina regulates Sertoli cell-tight junction dynamics in the rat testis. Endocrinology 2003;144:371–87.Google Scholar
Lustig, L, Denduchis, B, Gonzalez, NN, Puig, RP. Experimental orchitis induced in rats by passive transfer of an antiserum to seminiferous tubule basement membrane. Arch Androl 1978;1:333–43.Google Scholar
Li, MWM, Xia, W, Mruk, DD, et al. TNFα reversibly disrupts the blood–testis barrier and impairs Sertoli–germ cell adhesion in the seminiferous epithelium of adult rat testes. J Endocrinol 2006;190:313–29.CrossRefGoogle ScholarPubMed
Wong, EW, Cheng, CY. NC1 domain of collagen alpha3(IV) derived from the basement membrane regulates Sertoli cell blood–testis barrier dynamics. Spermatogenesis 2013;3:e25465.Google Scholar
Su, WH, Cheng, CY. Cdc42 is involved in NC1-peptide regulated BTB dynamics through actin and microtubule cytoskeletal re-organization. FASEB J 2019;33:14461–78.Google Scholar
Gao, Y, Chen, H, Lui, WY, Lee, WM, Cheng, CY. Basement membrane laminin α2 regulation of BTB dynamics via its effects on F-actin and microtubule (MT) cytoskeletons is mediated through mTORC1 signaling. Endocrinology 2017;158:963–78.Google Scholar
Gao, Y, Mruk, D, Chen, H, Lui, WY, Lee, WM, Cheng, CY. Regulation of the blood–testis barrier by a local axis in the testis: role of laminin α2 in the basement membrane. FASEB J 2017;31:584–97.CrossRefGoogle ScholarPubMed
Meyuhas, O. Ribosomal protein S6 phosphorylation: four decades of research. Int Rev Cell Mol Biol 2015;320:4173.Google Scholar
Mok, KW, Mruk, DD, Silvestrini, B, Cheng, CY. rpS6 regulates blood–testis barrier dynamics by affecting F-actin organization and protein recruitment. Endocrinology 2012;153:5036–48.Google Scholar
Mok, KW, Mruk, DD, Cheng, CY. rpS6 regulates blood–testis barrier dynamics through Akt-mediated effects on MMP-9. J Cell Sci 2014;127:4870–82.Google Scholar
Mok, KW, Chen, H, Lee, WM, Cheng, CY. rpS6 regulates blood–testis barrier dynamics through Arp3-mediated actin microfilament organization in rat Sertoli cells. An in vitro study. Endocrinology 2015;156:1900–13.Google Scholar
Li, SYT, Yan, M, Chen, H, et al. mTORC1/rpS6 regulates blood–testis barrier (BTB) dynamics and spermatogenetic function in the testis in vivo. Am J Physiol Endocrinol Metab 2018;314:E174–90.Google Scholar
Yan, M, Li, L, Mao, BP, et al. mTORC1/rpS6 signaling complex modifies BTB transport function – an in vivo study using the adjudin model. Am J Physiol Endocrinol Metab 2019;317:E121–38.Google Scholar
Tung, KS, Harakal, J, Qiao, H, et al. Egress of sperm autoantigen from seminiferous tubules maintains systemic tolerance. J Clin Invest 2017;127:1046–60.Google Scholar
Russell, LD, Saxena, NK, Weber, JE. Intratesticular injection as a method to assess the potential toxicity of various agents to study mechanisms of normal spermatogenesis. Gamete Res 1987;17:4356.Google Scholar
Dunleavy, JEM, O’Bryan, MK, Stanton, PG, O’Donnell, L. The cytoskeleton in spermatogenesis. Reproduction 2019;157:R53–72.Google Scholar
Cheng, CY, Mruk, DD. Regulation of spermiogenesis, spermiation and blood–testis barrier dynamics: novel insights from studies on Eps8 and Arp3. Biochem J 2011;435:553–62.Google Scholar
Tang, EI, Mruk, DD, Cheng, CY. Regulation of microtubule (MT)-based cytoskeleton in the seminiferous epithelium during spermatogenesis. Semin Cell Dev Biol 2016;59:3545.Google Scholar
Ortega, N, Werb, Z. New functional roles for noncollagenous domains of basement membrane collagens. J Cell Sci 2002;115:4201–14.Google Scholar
Sudhakar, A, Boosani, CS. Signaling mechanisms of endogenous angiogenesis inhibitors derived from type IV collagen. Gene Regul Syst Biol 2007;1:217–26.Google Scholar
Assadian, S, Teodoro, JG. Regulation of collagen-derived antiangiogenic factors by p53. Expert Opin Biol Ther 2008;8:941–50.Google Scholar
Barczyk, M, Carracedo, S, Gullberg, D. Integrins. Cell Tissue Res 2010;339:269–80.Google Scholar
Moreno-Layseca, P, Icha, J, Hamidi, H, Ivaska, J. Integrin trafficking in cells and tissues. Nat Cell Biol 2019;21:122–32.Google Scholar
Bachmann, M, Kukkurainen, S, Hytönen, VP, Wehrle-Haller, B. Cell adhesion by integrins. Physiol Rev 2019;99:1655–99.Google Scholar
Dunn, HA, Orlandi, C, Martemyanov, KA. Beyond the ligand: extracellular and transcellular G protein-coupled receptor complexes in physiology and pharmacology. Pharmacol Rev 2019;71:503–19.Google Scholar
Leitinger, B. Discoidin domain receptor functions in physiological and pathological conditions. Int Rev Cell Mol Biol 2014;310:3987.Google Scholar
de Rooij, DG. The spermatogonial stem cell niche. Microsc Res Tech 2009;72:580–5.Google Scholar
Clermont, Y, Leblond, CP, Messier, B. [Durée du cycle de l’épithélium seminal du rat]. Arch Anat Microsc Morphol Exp 1959;48:3756.Google Scholar
Hilscher, W. [Beitrage zur Orthologie und Palhologie des “Spermatogoniogenes” der Ratte]. Beitr Pathol Anat 1964;130:69132.Google Scholar
Franca, LR, Ogawa, T, Avarbock, MR, Brinster, RL, Russell, LD. Germ cell genotype controls cell cycle during spermatogenesis in the rat. Biol Reprod 1998;59:1371–7.Google Scholar
Clermont, Y, Harvey, SC. Duration of the cycle of the seminiferous epithelium of normal, hypophysectomized and hypophysectomized-hormone treated albino rats. Endocrinology 1965;76:80–9.Google Scholar
Russell, LD, Brinster, RL. Ultrastructural observations of spermatogenesis following transplantation of rat testis cells into mouse seminiferous tubules. J Androl 1996;17:615–27.Google Scholar
Aslam, H, Rosiipen, G, Krishnamurthy, H, et al. The cycle duration of the seminiferous epithelium remains unaltered during GnRH antagonist-induced testicular involution in rats and monkeys. J Endocrinol 1999;161:281–8.Google Scholar
Leblond, C, Clermont, Y. Definition of the stages of the cycle of the seminiferous epithelium in the rat. Ann N Y Acad Sci 1952;55:548–73.Google Scholar
Oakberg, EF. Duration of spermatogenesis in the mouse and timing of stages of the cycle of the seminiferous epithelium. Am J Anat 1956;99:507–16.Google Scholar
Clermont, Y, Trott, M. Duration of the cycle of the seminiferous epithelium in the mouse and hamster determined by means of 3H-thymidine and radioautography. Fertil Sertil 1969;20:805–17.Google Scholar
Russell, LD, Franca, LR, Brinster, RL. Ultrastructural observations of spermatogenesis in mice resulting from transplantation of mouse spermatogonia. J Androl 1996;17:603–14.Google Scholar
de Rooij, DG, Russell, LD. All you wanted to know about spermatogonia but were afraid to ask. J Androl 2000;21:776–98.Google Scholar
Ehmcke, J, Schlatt, S. A revised model for spermatogonial expansion in man: lessons from non-human primates. Reproduction 2006;132:673–80.Google Scholar
Ehmcke, J, Wistuba, J, Schlatt, S. Spermatogonial stem cells: questions, models and perspectives. Hum Reprod Update 2006;12:275–82.Google Scholar
Heller, CG, Clermont, Y. Kinetics of the germinal epithelium in man. Recent Prog Horm Res 1964;20:545–75.Google Scholar
Heller, CG, Clermont, Y. Spermatogenesis in man: an estimate of its duration. Science 1963;140:184–6.Google Scholar
Wu, S, Yan, M, Li, L, et al. mTORC1/rpS6 and spermatogenic function in the testis – insights from the adjudin model. Reprod Toxicol 2019;89:5466.Google Scholar

Further Reading

Cornwall, GA. New insights into epididymal biology and function. Hum Reprod Update 2009;15:213–27.Google Scholar
James, ER, Carrell, DT, Aston, KI, Jenkins, TG, Yeste, M, Salas-Huetos, A. The role of the epididymis and the contribution of epididymosomes to mammalian reproduction. Int J Mol Sci 2020;21:5377.Google Scholar
Stival, C, Puga Molina Ldel, C, Paudel, B, Buffone, MG, Visconti, PE, Krapf, D. Sperm capacitation and acrosome reaction in mammalian sperm. Adv Anat Embryol Cell Biol 2016;220:93106.Google Scholar
Wang, H, McGoldrick, LL, Chung, J-J. Sperm ion channels and transporters in male fertility and infertility. Nat Rev Urol 2021;18:4666.Google Scholar

References

James, ER, Carrell, DT, Aston, KI, Jenkins, TG, Yeste, M, Salas-Huetos, A. The role of the epididymis and the contribution of epididymosomes to mammalian reproduction. Int J Mol Sci 2020;21:5377.Google Scholar
Suede, SH, Malik, A, Sapra, A. Histology, Spermatogenesis. Treasure Island, FL: StatPearls Publishing, 2020.Google Scholar
Holstein, AF, Maekawa, M, Nagano, T, Davidoff, MS. Myofibroblasts in the lamina propria of human semi-niferous tubules are dynamic structures of heterogeneous phenotype. Arch Histol Cytol 1996;59:109–25.Google Scholar
Cooper, TG. Epididymal research: more warp than weft? Asian J Androl 2015;17:699703.Google Scholar
Jones, RC. Evolution of the vertebrate epididymis. J Reprod Fertil Suppl 1998;53:163–81.Google Scholar
Sullivan, R, Legare, C, Thabet, M, Thimon, V. Gene expression in the epididymis of normal and vasectomized men: what can we learn about human sperm maturation? J Androl 2011;32:686–97.Google Scholar
Rey, R, Picard, JY. Embryology and endocrinology of genital development. Baillieres Clin Endocrinol Metab 1998;12:1733.CrossRefGoogle ScholarPubMed
Yu, M, Wang, SM. Embryology, Wolffian Ducts. Treasure Island, FL: StatPearls Publishing, 2020.Google Scholar
Joseph, A, Yao, H, Hinton, BT. Development and morphogenesis of the Wolffian/epididymal duct, more twists and turns. Dev Biol 2009;325:614.Google Scholar
Hannema, SE, Hughes, IA. Regulation of Wolffian duct development. Horm Res 2007;67:142–51.Google Scholar
Dalati, MF, Oliveira, EST, Entezari, K. Review of the literature: organ of Giraldes epididymal appendage presenting as a painless scrotal mass in a 19-year-old male – a rare urologic entity. Case Rep Urol 2015;2015:748097.Google Scholar
Turner, TT. De Graaf’s thread: the human epididymis. J Androl 2008;29:237–50.Google Scholar
Kormano, M, Reijonen, K. Microvascular structure of the human epididymis. Am J Anat 1976;145:23–7.Google Scholar
Gaudin, J, Lefevre, C, Person, H, N’Guyen, H, Senecail, B. The venous hilum of the testis and epididymis: anatomic aspect. Surg Radiol Anat 1988;10:233–42.Google Scholar
Ricker, DD. The autonomic innervation of the epididymis: its effects on epididymal function and fertility. J Androl 1998;19:14.Google Scholar
Belleannee, C, Thimon, V, Sullivan, R. Region-specific gene expression in the epididymis. Cell Tissue Res 2012;349:717–31.Google Scholar
Domeniconi, RF, Souza, AC, Xu, B, Washington, AM, Hinton, BT. Is the epididymis a series of organs placed side by side? Biol Reprod 2016;95:10.Google Scholar
Leir, SH, Yin, S, Kerschner, JL, Cosme, W, Harris, A. An atlas of human proximal epididymis reveals cell-specific functions and distinct roles for CFTR. Life Sci Alliance 2020;3:e202000744.Google Scholar
Hess, RA. Small tubules, surprising discoveries: from efferent ductules in the turkey to the discovery that estrogen receptor alpha is essential for fertility in the male. Anim Reprod 2015;12:723.Google Scholar
Oliveira, CA, Carnes, K, Franca, LR, Hermo, L, Hess, RA. Aquaporin-1 and -9 are differentially regulated by oestrogen in the efferent ductule epithelium and initial segment of the epididymis. Biol Cell 2005;97:385–95.Google Scholar
Joseph, A, Shur, BD, Hess, RA. Estrogen, efferent ductules, and the epididymis. Biol Reprod 2011;84:207–17.Google Scholar
Corbo, RM, Ulizzi, L, Piombo, L, Martinez-Labarga, C, De Stefano, GF, Scacchi, R. Estrogen receptor alpha polymorphisms and fertility in populations with different reproductive patterns. Mol Hum Reprod 2007;13:537–40.Google Scholar
Hess, RA. The efferent ductules: structure and functions. In: Robaire, B and Hinton, BT, eds. The Epididymis: From Molecules to Clinical Practice: A Comprehensive Survey of the Efferent Ducts, the Epididymis and the Vas Deferens. New York, NY: Springer, 2002; pp. 4980.Google Scholar
Rosenfeld, CS. Male reproductive tract cilia beat to a different drummer. Proc Natl Acad Sci U S A 2019;116:3361–3.Google Scholar
Aire, TA. Active spermiophagy in the initial part of the proximal efferent duct of the epididymis of normal domestic fowl (Gallus domesticus). Res Vet Sci 2000;68:135–40.Google Scholar
Ilio, KY, Hess, RA. Structure and function of the ductuli efferentes: a review. Microsc Res Tech 1994;29:432–67.Google Scholar
Browne, JA, Yang, R, Leir, SH, Eggener, SE, Harris, A. Expression profiles of human epididymis epithelial cells reveal the functional diversity of caput, corpus and cauda regions. Mol Hum Reprod 2016;22:6982.Google Scholar
Cornwall, GA. New insights into epididymal biology and function. Hum Reprod Update 2009;15:213–27.Google Scholar
De Grava Kempinas, W, Klinefelter, GR. Interpreting histopathology in the epididymis. Spermatogenesis 2014;4:e979114.Google Scholar
Zhou, W, De Iuliis, GN, Dun, MD, Nixon, B. Characteristics of the epididymal luminal environment responsible for sperm maturation and storage. Front Endocrinol (Lausanne) 2018;9:59.Google Scholar
Hermo, L, Oko, R, Robaire, B. Epithelial cells of the epididymis show regional variations with respect to the secretion of endocytosis of immobilin as revealed by light and electron microscope immunocytochemistry. Anat Rec 1992;232:202–20.Google Scholar
Jervis, KM, Robaire, B. Changes in gene expression during aging in the Brown Norway rat epididymis. Exp Gerontol 2002;37:897906.Google Scholar
Zirkin, BR, Santulli, R, Strandberg, JD, Wright, WW, Ewing, LL. Testicular steroidogenesis in the aging brown Norway rat. J Androl 1993;14:118–23.Google Scholar
Serre, V, Robaire, B. Paternal age affects fertility and progeny outcome in the Brown Norway rat. Fertil Steril 1998;70:625–31.Google Scholar
Serre, V, Robaire, B. Segment-specific morphological changes in aging Brown Norway rat epididymis. Biol Reprod 1998;58:497513.Google Scholar
Gregory, M, Cyr, DG. The blood–epididymis barrier and inflammation. Spermatogenesis. 2014;4:e979619.Google Scholar
Elfgen, V, Mietens, A, Mewe, M, Hau, T, Middendorff, R. Contractility of the epididymal duct: function, regulation and potential drug effects. Reproduction 2018;156:R125–41.Google Scholar
Vadnais, ML, Aghajanian, HK, Lin, A, Gerton, GL. Signaling in sperm: toward a molecular understanding of the acquisition of sperm motility in the mouse epididymis. Biol Reprod 2013;89:127.Google Scholar
Esposito, G, Jaiswal, BS, Xie, F, et al. Mice deficient for soluble adenylyl cyclase are infertile because of a severe sperm-motility defect. Proc Natl Acad Sci U S A 2004;101:2993–8.Google Scholar
Gervasi, MG, Visconti, PE. Molecular changes and signaling events occurring in spermatozoa during epididymal maturation. Andrology 2017;5:204–18.Google Scholar
Cooper, TG. The epididymis, cytoplasmic droplets and male fertility. Asian J Androl 2011;13:130–8.Google Scholar
Michel, V, Pilatz, A, Hedger, MP, Meinhardt, A. Epididymitis: revelations at the convergence of clinical and basic sciences. Asian J Androl 2015;17:756–63.Google Scholar
Arrighi, S. Are the basal cells of the mammalian epididymis still an enigma? Reprod Fertil Dev 2014;26:1061–71.Google Scholar
Nashan, D, Malorny, U, Sorg, C, Cooper, T, Nieschlag, E. Immuno-competent cells in the murine epididymis. Int J Androl 1989;12:8594.Google Scholar
Ritchie, AW, Hargreave, TB, James, K, Chisholm, GD. Intra-epithelial lymphocytes in the normal epididymis. A mechanism for tolerance to sperm auto-antigens? Br J Urol 1984;56:7983.Google Scholar
Shum, WW, Da Silva, N, McKee, M, Smith, PJ, Brown, D, Breton, S. Transepithelial projections from basal cells are luminal sensors in pseudostratified epithelia. Cell 2008;135:1108–17.Google Scholar
Shum, WW, Ruan, YC, Da Silva, N, Breton, S. Establishment of cell–cell cross talk in the epididymis: control of luminal acidification. J Androl 2011;32:576–86.Google Scholar
Leung, GP, Cheung, KH, Leung, CT, Tsang, MW, Wong, PY. Regulation of epididymal principal cell functions by basal cells: role of transient receptor potential (Trp) proteins and cyclooxygenase-1 (COX-1). Mol Cell Endocrinol 2004;216:513.Google Scholar
Dube, E, Hermo, L, Chan, PT, Cyr, DG. Alterations in the human blood–epididymis barrier in obstructive azoospermia and the development of novel epididymal cell lines from infertile men. Biol Reprod 2010;83:584–96.Google Scholar
Itoh, M, Terayama, H, Naito, M, Ogawa, Y, Tainosho, S. Tissue microcircumstances for leukocytic infiltration into the testis and epididymis in mice. J Reprod Immunol 2005;67:5767.Google Scholar
Wijayarathna, R, Hedger, MP. Activins, follistatin and immunoregulation in the epididymis. Andrology 2019;7:703–11.Google Scholar
Ozbek, M, Hitit, M, Ergun, E, et al. Expression profile of Toll-like receptor 4 in rat testis and epididymis throughout postnatal development. Andrologia 2020;52:e13518.Google Scholar
Atlas, THP. The epididymis-specific proteome. Available from: www.proteinatlas.org/humanproteome/tissue/epididymis.Google Scholar
Sipila, P, Krutskikh, A, Pujianto, DA, Poutanen, M, Huhtaniemi, I. Regional expression of androgen receptor coregulators and androgen action in the mouse epididymis. J Androl 2011;32:711–17.Google Scholar
Robaire, B, Viger, RS. Regulation of epididymal epithelial cell functions. Biol Reprod 1995;52:226–36.Google Scholar
Rago, V, Romeo, F, Giordano, F, et al. Expression of oestrogen receptors (GPER, ESR1, ESR2) in human ductuli efferentes and proximal epididymis. Andrology 2018;6:192–8.Google Scholar
Nanjappa, MK, Hess, RA, Medrano, TI, Locker, SH, Levin, ER, Cooke, PS. Membrane-localized estrogen receptor 1 is required for normal male reproductive development and function in mice. Endocrinology 2016;157:2909–19.Google Scholar
Couse, JE, Mahato, D, Eddy, EM, Korach, KS. Molecular mechanism of estrogen action in the male: insights from the estrogen receptor null mice. Reprod Fertil Dev 2001;13:211–19.Google Scholar
Hess, RA, Gist, DH, Bunick, D, et al. Estrogen receptor (alpha and beta) expression in the excurrent ducts of the adult male rat reproductive tract. J Androl.1997;18:602–11.Google Scholar
Xu, B, Washington, AM, Hinton, BT. PTEN signaling through RAF1 proto-oncogene serine/threonine kinase (RAF1)/ERK in the epididymis is essential for male fertility. Proc Natl Acad Sci U S A 2014;111:18643–8.Google Scholar
Moskovtsev, SI, Jarvi, K, Legare, C, Sullivan, R, Mullen, JB. Epididymal P34H protein deficiency in men evaluated for infertility. Fertil Steril 2007;88:1455–7.Google Scholar
Wu, H, Gao, Y, Ma, C, et al. A novel hemizygous loss-of-function mutation in ADGRG2 causes male infertility with congenital bilateral absence of the vas deferens. J Assist Reprod Genet 2020;37:1421–9.Google Scholar
Patat, O, Pagin, A, Siegfried, A, et al. Truncating mutations in the adhesion G protein-coupled receptor G2 gene ADGRG2 cause an X-linked congenital bilateral absence of vas deferens. Am J Hum Genet 2016;99:437–42.Google Scholar
Hamil, KG, Sivashanmugam, P, Richardson, RT, et al. HE2beta and HE2gamma, new members of an epididymis-specific family of androgen-regulated proteins in the human. Endocrinology 2000;141:1245–53.Google Scholar
Yenugu, S, Hamil, KG, French, FS, Hall, SH. Antimicrobial actions of the human epididymis 2 (HE2) protein isoforms, HE2alpha, HE2beta1 and HE2beta2. Reprod Biol Endocrinol 2004;2:61.Google Scholar
Radhakrishnan, Y, Hamil, KG, Tan, JA, et al. Novel partners of SPAG11B isoform D in the human male reproductive tract. Biol Reprod 2009;81:647–56.Google Scholar
Pujianto, DA, Loanda, E, Sari, P, Midoen, YH, Soeharso, P. Sperm-associated antigen 11A is expressed exclusively in the principal cells of the mouse caput epididymis in an androgen-dependent manner. Reprod Biol Endocrinol 2013;11:59.Google Scholar
Plante, G, Therien, I, Lachance, C, Leclerc, P, Fan, J, Manjunath, P. Implication of the human Binder of SPerm Homolog 1 (BSPH1) protein in capacitation. Mol Hum Reprod 2014;20:409–21.Google Scholar
Vanier, MT, Millat, G. Structure and function of the NPC2 protein. Biochim Biophys Acta 2004;1685:1421.Google Scholar
Sullivan, R, Legare, C, Villeneuve, M, Foliguet, B, Bissonnette, F. Levels of P34H, a sperm protein of epididymal origin, as a predictor of conventional in vitro fertilization outcome. Fertil Steril 2006;85:1557–9.Google Scholar
Cornwall, GA, Hsia, N. ADAM7, a member of the ADAM (a disintegrin and metalloprotease) gene family is specifically expressed in the mouse anterior pituitary and epididymis. Endocrinology 1997;138:4262–72.Google Scholar
Oh, JS, Han, C, Cho, C. ADAM7 is associated with epididymosomes and integrated into sperm plasma membrane. Mol Cells 2009;28:441–6.Google Scholar
Cohen, DJ, Da Ros, VG, Busso, D, et al. Participation of epididymal cysteine-rich secretory proteins in sperm-egg fusion and their potential use for male fertility regulation. Asian J Androl 2007;9:528–32.Google Scholar
Roberts, KP, Johnston, DS, Nolan, MA, et al. Structure and function of epididymal protein cysteine-rich secretory protein-1. Asian J Androl 2007;9:508–14.Google Scholar
Dube, E, Hermo, L, Chan, PT, Cyr, DG. Alterations in gene expression in the caput epididymides of nonobstructive azoospermic men. Biol Reprod 2008;78:342–51.Google Scholar
Thimon, V, Calvo, E, Koukoui, O, Legare, C, Sullivan, R. Effects of vasectomy on gene expression profiling along the human epididymis. Biol Reprod 2008;79:262–73.Google Scholar
Gibbs, GM, O’Bryan, MK. Cysteine rich secretory proteins in reproduction and venom. Soc Reprod Fertil Suppl 2007;65:261–7.Google Scholar
Kirchhoff, C, Hale, G. Cell-to-cell transfer of glycosylphosphatidylinositol-anchored membrane proteins during sperm maturation. Mol Hum Reprod 1996;2:177–84.Google Scholar
Yamaguchi, R, Yamagata, K, Hasuwa, H, Inano, E, Ikawa, M, Okabe, M. Cd52, known as a major maturation-associated sperm membrane antigen secreted from the epididymis, is not required for fertilization in the mouse. Genes Cells 2008;13:851–61.Google Scholar
Vadnais, ML, Cao, W, Aghajanian, HK, et al. Adenine nucleotide metabolism and a role for AMP in modulating flagellar waveforms in mouse sperm. Biol Reprod 2014;90:128.Google Scholar
Patil, AA, Cai, Y, Sang, Y, Blecha, F, Zhang, G. Cross-species analysis of the mammalian beta-defensin gene family: presence of syntenic gene clusters and preferential expression in the male reproductive tract. Physiol Genomics 2005;23:517.Google Scholar
Tollner, TL, Venners, SA, Hollox, EJ, et al. A common mutation in the defensin DEFB126 causes impaired sperm function and subfertility. Sci Transl Med 2011;3:92ra65.Google Scholar
Rao, J, Herr, JC, Reddi, PP, et al. Cloning and characterization of a novel sperm-associated isoantigen (E-3) with defensin- and lectin-like motifs expressed in rat epididymis. Biol Reprod 2003;68:290301.Google Scholar
Suzuki, K, Yu, X, Chaurand, P, et al. Epididymis-specific lipocalin promoters. Asian J Androl 2007;9:515–21.Google Scholar
Thimon, V, Koukoui, O, Calvo, E, Sullivan, R. Region-specific gene expression profiling along the human epididymis. Mol Hum Reprod 2007;13:691704.Google Scholar
Belleannee, C, Calvo, E, Thimon, V, et al. Role of microRNAs in controlling gene expression in different segments of the human epididymis. PLoS ONE 2012;7:e34996.Google Scholar
Alwaal, A, Breyer, BN, Lue, TF. Normal male sexual function: emphasis on orgasm and ejaculation. Fertil Steril 2015;104:1051–60.Google Scholar
Revenig, L, Leung, A, Hsiao, W. Ejaculatory physiology and pathophysiology: assessment and treatment in male infertility. Transl Androl Urol 2014;3:41–9.Google Scholar
Suarez, SS, Pacey, AA. Sperm transport in the female reproductive tract. Hum Reprod Update 2006;12:2337.Google Scholar
Katz, DF, Slade, DA, Nakajima, ST. Analysis of pre-ovulatory changes in cervical mucus hydration and sperm penetrability. Adv Contracept 1997;13:143–51.Google Scholar
D’Cruz, OJ, Wang, BL, Haas, GG Jr. Phagocytosis of immunoglobulin G and C3-bound human sperm by human polymorphonuclear leukocytes is not associated with the release of oxidative radicals. Biol Reprod 1992;46:721–32.Google Scholar
Williams, M, Hill, CJ, Scudamore, I, Dunphy, B, Cooke, ID, Barratt, CL. Sperm numbers and distribution within the human fallopian tube around ovulation. Hum Reprod 1993;8:2019–26.Google Scholar
Chang, MC. Fertilizing capacity of spermatozoa deposited into the fallopian tubes. Nature 1951;168:697–8.Google Scholar
Austin, CR. The capacitation of the mammalian sperm. Nature 1952;170:326.Google Scholar
Stival, C, Puga Molina, L del C, Paudel, B, Buffone, MG, Visconti, PE, Krapf, D. Sperm capacitation and acrosome reaction in mammalian sperm. Adv Anat Embryol Cell Biol 2016;220:93106.Google Scholar
Pastor-Soler, N, Piétrement, C, Breton, S. Role of acid/base transporters in the male reproductive tract and potential consequences of their malfunction. Physiology (Bethesda) 2005;20:417–28.Google Scholar
Kavanagh, JP. Sodium, potassium, calcium, magnesium, zinc, citrate and chloride content of human prostatic and seminal fluid. J Reprod Fertil 1985;75:3541.Google Scholar
Kleinboelting, S, Diaz, A, Moniot, S, et al. Crystal structures of human soluble adenylyl cyclase reveal mechanisms of catalysis and of its activation through bicarbonate. Proc Natl Acad Sci U S A 2014;111:3727–32.Google Scholar
Wang, H, McGoldrick, LL, Chung, J-J. Sperm ion channels and transporters in male fertility and infertility. Nat Rev Urol 2021;18:4666.Google Scholar
Hamamah, S, Gatti, JL. Role of the ionic environment and internal pH on sperm activity. Hum Reprod 1998;13:2030.Google Scholar
Kirichok, Y, Navarro, B, Clapham, DE. Whole-cell patch-clamp measurements of spermatozoa reveal an alkaline-activated Ca2+ channel. Nature 2006;439:737–40.Google Scholar
Sun, XH, Zhu, YY, Wang, L, et al. The Catsper channel and its roles in male fertility: a systematic review. Reprod Biol Endocrinol 2017;15:65.Google Scholar
Takeshita, K, Sakata, S, Yamashita, E, et al. X-ray crystal structure of voltage-gated proton channel. Nat Struct Mol Biol 2014;21:352–7.Google Scholar
Wang, D, Hu, J, Bobulescu, IA, et al. A sperm-specific Na+/H+ exchanger (sNHE) is critical for expression and in vivo bicarbonate regulation of the soluble adenylyl cyclase (sAC). Proc Natl Acad Sci U S A 2007;104:9325–30.Google Scholar
Chávez, JC, Hernández-González, EO, Wertheimer, E, Visconti, PE, Darszon, A, Treviño, CL. Participation of the Cl/HCO(3) exchangers SLC26A3 and SLC26A6, the Cl channel CFTR, and the regulatory factor SLC9A3R1 in mouse sperm capacitation. Biol Reprod 2012;86:114.Google Scholar
Santi, CM, Martínez-López, P, de la Vega-Beltrán, JL, et al. The SLO3 sperm-specific potassium channel plays a vital role in male fertility. FEBS Lett 2010;584:1041–6.Google Scholar
Wang, D, King, SM, Quill, TA, Doolittle, LK, Garbers, DL. A new sperm-specific Na+/H+ exchanger required for sperm motility and fertility. Nat Cell Biol 2003;5:1117–22.Google Scholar
Chen, SR, Chen, M, Deng, SL, Hao, XX, Wang, XX, Liu, YX. Sodium–hydrogen exchanger NHA1 and NHA2 control sperm motility and male fertility. Cell Death Dis 2016;7:e2152.Google Scholar
Miller, MR, Kenny, SJ, Mannowetz, N, et al. Asymmetrically positioned flagellar control units regulate human sperm rotation. Cell Rep 2018;24:2606–13.Google Scholar
Chen, H, Ruan, YC, Xu, WM, Chen, J, Chan, HC. Regulation of male fertility by CFTR and implications in male infertility. Hum Reprod Update 2012;18:703–13.Google Scholar
Qi, H, Moran, MM, Navarro, B, et al. All four CatSper ion channel proteins are required for male fertility and sperm cell hyperactivated motility. Proc Natl Acad Sci U S A. 2007;104:1219–23.Google Scholar
Liu, J, Xia, J, Cho, KH, Clapham, DE, Ren, D. CatSperbeta, a novel transmembrane protein in the CatSper channel complex. J Biol Chem 2007;282:18945–52.Google Scholar
Wang, H, Liu, J, Cho, KH, Ren, D. A novel, single, transmembrane protein CATSPERG is associated with CATSPER1 channel protein. Biol Reprod 2009;81:539–44.Google Scholar
Chung, JJ, Navarro, B, Krapivinsky, G, Krapivinsky, L, Clapham, DE. A novel gene required for male fertility and functional CATSPER channel formation in spermatozoa. Nat Commun 2011;2:153.Google Scholar
Ren, D, Navarro, B, Perez, G, et al. A sperm ion channel required for sperm motility and male fertility. Nature 2001;413:603–9.Google Scholar
Ren, D, Xia, J. Calcium signaling through CatSper channels in mammalian fertilization. Physiology (Bethesda) 2010;25:165–75.Google Scholar
Li, H, Ding, X, Guo, C, Guan, H, Xiong, C. Immunization of male mice with B-cell epitopes in transmembrane domains of CatSper1 inhibits fertility. Fertil Steril 2012;97:445–52.Google Scholar
Sanchez, G, Nguyen, AN, Timmerberg, B, Tash, JS, Blanco, G. The Na,K-ATPase alpha4 isoform from humans has distinct enzymatic properties and is important for sperm motility. Mol Hum Reprod 2006;12:565–76.Google Scholar
Brenker, C, Zhou, Y, Müller, A, et al. The Ca2+-activated K+ current of human sperm is mediated by Slo3. Elif 2014;3:e01438.Google Scholar
Yamamoto, M, Hibi, H, Miyake, K, et al. Does epididymal length in men with congenital bilateral absence of the vas deferens have a correlation with the fertilization rate of epididymal sperm retrieved by micropuncture technique? Nagoya J Med Sci 1996;59:31–5.Google Scholar
McCallum, TJ, Milunsky, JM, Cunningham, DL, Harris, DH, Maher, TA, Oates, RD. Fertility in men with cystic fibrosis: an update on current surgical practices and outcomes. Chest 2000;118:1059–62.Google Scholar
Kaplan, E, Shwachman, H, Perlmutter, AD, Rule, A, Khaw, KT, Holsclaw, DS. Reproductive failure in males with cystic fibrosis. N Engl J Med 1968;279:65–9.Google Scholar
Goldstein, M, Schlossberg, S. Men with congenital absence of the vas deferens often have seminal vesicles. J Urol 1988;140:85–6.Google Scholar
Yamamoto, M, Hibi, H, Miyake, K, Asada, Y, Suganuma, N, Tomoda, Y. Microsurgical epididymal sperm aspiration versus epididymal micropuncture with perivascular nerve stimulation for intracytoplasmic sperm injection to treat unreconstructable obstructive azoospermia. Arch Androl 1996;36:217–24.Google Scholar
Johansen, TE. Anatomy of the testis and epididymis in cryptorchidism. Andrologia 1987;19:565–9.Google Scholar
Brisson, P, Feins, N, Patel, H. Torsion of the epididymis. J Pediatr Surg 2005;40:1795–7.Google Scholar
Favorito, LA, Riberio Julio-Junior, H, Sampaio, FJ. Relationship between undescended testis position and prevalence of testicular appendices, epididymal anomalies, and patency of processus vaginalis. Biomed Res Int 2017;2017:5926370.Google Scholar
Marshall, FF, Shermeta, DW. Epididymal abnormalities associated with undescended testis. J Urol 1979;121:341–3.Google Scholar
Heath, AL, Man, DW, Eckstein, HB. Epididymal abnormalities associated with maldescent of the testis. J Pediatr Surg 1984;19:47–9.Google Scholar
Nicholson, A, Rait, G, Murray-Thomas, T, Hughes, G, Mercer, CH, Cassell, J. Management of epididymo-orchitis in primary care: results from a large UK primary care database. Br J Gen Pract 2010;60:e407–22.Google Scholar
Campbell, MF. Gonococcus epididymitis: observations in three thousand cases from the urological services of Bellevue Hospital. Ann Surg 1927;86:577–90.Google Scholar
Kaver, I, Matzkin, H, Braf, ZF. Epididymo-orchitis: a retrospective study of 121 patients. J Fam Pract 1990;30:548–52.Google Scholar
Slavis, SA, Kollin, J, Miller, JB. Pyocele of scrotum: consequence of spontaneous rupture of testicular abscess. Urology 1989;33:313–16.Google Scholar
Berger, RE. Acute epididymitis: etiology and therapy. Semin Urol 1991;9:2831.Google Scholar
Berger, RE, Alexander, ER, Harnisch, JP, et al. Etiology, manifestations and therapy of acute epididymitis: prospective study of 50 cases. J Urol 1979;121:750–4.Google Scholar
Hagley, M. Epididymo-orchitis and epididymitis: a review of causes and management of unusual forms. Int J STD AIDS 2003;14:372–7; quiz 8.Google Scholar
Gould, SW. Epididymo-orchitis: a rare, fatal, intra-abdominal cause. Ann R Coll Surg Engl 1996;78:230.Google Scholar
Likitnukul, S, McCracken, GH Jr, Nelson, JD, Votteler, TP. Epididymitis in children and adolescents. A 20-year retrospective study. Am J Dis Child 1987;141:41–4.Google Scholar
Merlini, E, Rotundi, F, Seymandi, PL, Canning, DA. Acute epididymitis and urinary tract anomalies in children. Scand J Urol Nephrol 1998;32:273–5.Google Scholar
Bukowski, TP, Lewis, AG, Reeves, D, Wacksman, J, Sheldon, CA. Epididymitis in older boys: dysfunctional voiding as an etiology. J Urol 1995;154(2 Pt 2):762–5.Google Scholar
Singh, R, Mostafid, H, Hindley, RG. Measles, mumps and rubella – the urologist’s perspective. Int J Clin Pract 2006;60:335–9.Google Scholar
Caldamone, AA, Valvo, JR, Altebarmakian, VK, Rabinowitz, R. Acute scrotal swelling in children. J Pediatr Surg 1984;19:581–4.Google Scholar
Tanaka, K, Ogasawara, Y, Nikai, K, Yamada, S, Fujiwara, K, Okazaki, T. Acute scrotum and testicular torsion in children: a retrospective study in a single institution. J Pediatr Urol 2020;16:5560.Google Scholar
Boettcher, M, Bergholz, R, Krebs, TF, et al. Differentiation of epididymitis and appendix testis torsion by clinical and ultrasound signs in children. Urology 2013;82:899904.Google Scholar
Docimo, SG, Rukstalis, DB, Rukstalis, MR, Kang, J, Cotton, D, DeWolf, WC. Candida epididymitis: newly recognized opportunistic epididymal infection. Urology 1993;41:280–2.Google Scholar
Hood, SV, Bell, D, McVey, R, Wilson, G, Wilkins, EG. Prostatitis and epididymo-orchitis due to Aspergillus fumigatus in a patient with AIDS. Clin Infect Dis 1998;26:229–31.Google Scholar
Seçil, M, Göktay, AY, Dicle, O, Yörükoğlu, K. Bilateral epididymal Candida abscesses: sonographic findings and sonographically guided fine-needle aspiration. J Clin Ultrasound 1998;26:413–15.Google Scholar
Singh, D, Fontanella, M, Voci, S. Tuberculous epididymitis. Ultrasound Q 2012;28:145–7.Google Scholar
O’Connell, HE, Russell, JM, Schultz, TC. Delayed epididymitis following intravesical bacillus Calmette–Guerin administration. Aust N Z J Surg 1993;63:70–2.Google Scholar
Gelfand, M, Ross, CM, Blair, DM, Castle, WM, Weber, MC. Schistosomiasis of the male pelvic organs. Severity of infection as determined by digestion of tissue and histologic methods in 300 cadavers. Am J Trop Med Hyg 1970;19:779–84.Google Scholar
Kollias, G, Kyriakopoulos, M, Tiniakos, G. Epididymitis from Enterobius vermicularis: case report. J Urol 1992;147:1114–16.Google Scholar
Shiadeh, MN, Niyyati, M, Fallahi, S, Rostami, A. Human parasitic protozoan infection to infertility: a systematic review. Parasitol Res 2016;115:469–77.Google Scholar
Carvalho, T, Trindade, S, Pimenta, S, Santos, AB, Rijo-Ferreira, F, Figueiredo, LM. Trypanosoma brucei triggers a marked immune response in male reproductive organs. PLoS Negl Trop Dis 2018;12:e0006690.Google Scholar
Williams, PB, Henderson, RJ, Sanusi, ID, Venable, DD. Ultrasound diagnosis of filarial funiculoepididymitis. Urology 1996;48:644–6.Google Scholar
Hutcheson, J, Peters, CA, Diamond, DA. Amiodarone induced epididymitis in children. J Urol 1998;160:515–17.Google Scholar
Shen, Y, Liu, H, Cheng, J, Bu, P. Amiodarone-induced epididymitis: a pathologically confirmed case report and review of the literature. Cardiology 2014;128:349–51.Google Scholar
Nikolaou, M, Ikonomidis, I, Lekakis, I, Tsiodras, S, Kremastinos, D. Amiodarone-induced epididymitis: a case report and review of the literature. Int J Cardiol 2007;121:e15–6.Google Scholar
Greene, HL, Graham, EL, Werner, JA, et al. Toxic and therapeutic effects of amiodarone in the treatment of cardiac arrhythmias. J Am Coll Cardiol 1983;2:1114–28.Google Scholar
Donzella, JG, Merrick, GS, Lindert, DJ, et al. Epididymitis after transrectal ultrasound-guided needle biopsy of prostate gland. Urology 2004;63:306–8.Google Scholar
Hoffelt, SC, Wallner, K, Merrick, G. Epididymitis after prostate brachytherapy. Urology 2004;63:293–6.Google Scholar
Perrouin-Verbe, B, Labat, JJ, Richard, I, Mauduyt de la Greve, I, Buzelin, JM, Mathe, JF. Clean intermittent catheterisation from the acute period in spinal cord injury patients. Long term evaluation of urethral and genital tolerance. Paraplegia 1995;33:619–24.Google Scholar
Moss, WM. A comparison of open-end versus closed-end vasectomies: a report on 6220 cases. Contraception 1992;46:521–5.Google Scholar
Gordon, LM, Stein, SM, Ralls, PW. Traumatic epididymitis: evaluation with color Doppler sonography. AJR Am J Roentgenol 1996;166:1323–5.Google Scholar
Sawyer, EK, Anderson, JR. Acute epididymitis: a work-related injury? J Natl Med Assoc 1996;88:385–7.Google Scholar
Yeung, CH, Wang, K, Cooper, TG. Why are epididymal tumours so rare? Asian J Androl 2012;14:465–75.Google Scholar
Elsasser, E. Tumors of the epididymis. Recent Results Cancer Res 1977;60:163–75.Google Scholar
Zou, ZJ, Xiao, YM, Liu, ZH, et al. Clinicopathological characteristics, treatment, and prognosis of rarely primary epididymal adenocarcinoma: a review and update. Biomed Res Int 2017;2017:4126740.Google Scholar
Cazorla, A, Algros, MP, Bedgedjian, I, Chabannes, E, Camparo, P, Valmary-Degano, S. Epididymal leiomyoadenomatoid tumor: a case report and review of literature. Curr Urol 2014;7:195–8.Google Scholar
Algaba, F, Santaularia, JM, Villavicencio, H. Metastatic tumor of the epididymis and spermatic cord. Eur Urol 1983;9:56–9.Google Scholar
Peng, J, Yuan, Y, Cui, W, et al. Causes of suspected epididymal obstruction in Chinese men. Urology 2012;80:1258–61.Google Scholar
Gupta, R, Singh, P, Kumar, R. Should men with idiopathic obstructive azoospermia be screened for genitourinary tuberculosis? J Hum Reprod Sci 2015;8:43–7.Google Scholar
Dohle, GR, Colpi, GM, Hargreave, TB, et al. EAU guidelines on male infertility. Eur Urol 2005;48:703–11.Google Scholar
Hopps, CV, Goldstein, M. Microsurgical reconstruction of iatrogenic injuries to the epididymis from hydrocelectomy. J Urol 2006;176:2077–9; discussion 80.Google Scholar
Breeland, E, Cohen, MS, Warner, RS, Leiter, E. Epididymal obstruction in azoospermic males. Infertility 1981;4:4966.Google Scholar
Ammar, T, Sidhu, PS, Wilkins, CJ. Male infertility: the role of imaging in diagnosis and management. Br J Radiol 2012;85:S59–68.Google Scholar
Berardinucci, D, Zini, A, Jarvi, K. Outcome of microsurgical reconstruction in men with suspected epididymal obstruction. J Urol 1998;159:831–4.Google Scholar
Coward, RM, Mills, JN. A step-by-step guide to office-based sperm retrieval for obstructive azoospermia. Transl Androl Urol 2017;6:730–44.Google Scholar
Meniru, GI, Gorgy, A, Batha, S, Clarke, RJ, Podsiadly, BT, Craft, IL. Studies of percutaneous epididymal sperm aspiration (PESA) and intracytoplasmic sperm injection. Hum Reprod Update 1998;4:5771.Google Scholar
Lin, YM, Hsu, CC, Kuo, TC, Lin, JS, Wang, ST, Huang, KE. Percutaneous epididymal sperm aspiration versus microsurgical epididymal sperm aspiration for irreparable obstructive azoospermia – experience with 100 cases. J Formos Med Assoc 2000;99:459–65.Google Scholar
Schlegel, PN, Palermo, GD, Alikani, M, et al. Micropuncture retrieval of epididymal sperm with in vitro fertilization: importance of in vitro micromanipulation techniques. Urology 1995;46:238–41.Google Scholar
Bernie, AM, Ramasamy, R, Stember, DS, Stahl, PJ. Microsurgical epididymal sperm aspiration: indications, techniques and outcomes. Asian J Androl 2013;15:40–3.Google Scholar
Esteves, SC, Miyaoka, R, Orosz, JE, Agarwal, A. An update on sperm retrieval techniques for azoospermic males. Clinics (Sao Paulo) 2013;68:99110.Google Scholar
Hammoud, I, Bailly, M, Bergere, M, et al. Testicular spermatozoa are of better quality than epididymal spermatozoa in patients with obstructive azoospermia. Urology 2017;103:106–11.Google Scholar
Farber, NJ, Flannigan, R, Srivastava, A, Wang, H, Goldstein, M. Vasovasostomy: kinetics and predictors of patency. Fertil Steril 2020;113:77480 e3.Google Scholar
Yoon, YE, Lee, HH, Park, SY, et al. The role of vasoepididymostomy for treatment of obstructive azoospermia in the era of in vitro fertilization: a systematic review and meta-analysis. Asian J Androl 2018;21:6773.Google Scholar
Chan, PT. The evolution and refinement of vasoepididymostomy techniques. Asian J Androl 2013;15:4955.Google Scholar
Hinton, BT, Cooper, TG. The epididymis as a target for male contraceptive development. Handb Exp Pharmacol 2010;198:117–37.Google Scholar

Further Reading

Campo, S, Ambao, V, Creus, S, et al. Carbohydrate complexity and proportions of serum FSH isoforms in the male: lectin-based studies. Mol Cell Endocrinol 2007;260–2:197204.Google Scholar
Das, N, Kumar, TR. Molecular regulation of follicle-stimulating hormone synthesis, secretion and action. J Mol Endocrinol 2018;60:R131–55.Google Scholar
Moore, AM, Coolen, LM, Porter, DT, Goodman, RL, Lehman, MN. KNDy cells revisited. Endocrinology 2018;159:3219–34.Google Scholar

References

Herbison, A. Physiology of the adult gonadotropin-releasing hormone neuronal network. In: Plant, TM, Zeleznik, AJ, eds. Knobil and Neill’s Physiology of Reproduction, 4th ed. Oxford: Academic Press, 2015; pp. 399467.Google Scholar
Krsmanovic, LZ, Hu, L, Leung, PK, Feng, H, Catt, KJ. Pulsatile GnRH secretion: roles of G protein-coupled receptors, second messengers and ion channels. Mol Cell Endocrinol 2010;314:158–63.Google Scholar
Krsmanovic, LZ, Stojilkovic, SS, Mertz, LM, Tomic, M, Catt, KJ. Expression of gonadotropin-releasing hormone receptors and autocrine regulation of neuropeptide release in immortalized hypothalamic neurons. Proc Natl Acad Sci U S A 1993;90:3908–12.Google Scholar
Xu, C, Xu, XZ, Nunemaker, CS, Moenter, SM. Dose-dependent switch in response of gonadotropin-releasing hormone (GnRH) neurons to GnRH mediated through the type I GnRH receptor. Endocrinology 2004;145:728–35.Google Scholar
Cho, S, Han, J, Sun, W, et al. Evidence for autocrine inhibition of gonadotropin-releasing hormone (GnRH) gene transcription by GnRH in hypothalamic GT1-1 neuronal cells. Brain Res Mol Brain Res 1997;50:51–8.Google Scholar
Krsmanovic, LZ, Hu, L, Leung, PK, Feng, H, Catt, KJ. Pulsatile GnRH secretion: roles of G protein-coupled receptors, second messengers and ion channels. Mol Cell Endocrinol 2010;314:158–63.Google Scholar
Clarkson, J, Han, SY, Piet, R, et al. Definition of the hypothalamic GnRH pulse generator in mice. Proc Natl Acad Sci U S A 2017;114:E10216–23.Google Scholar
Seminara, SB, Messager, S, Chatzidaki, EE, et al. The GPR54 gene as a regulator of puberty. N Engl J Med 2003;349:1614–27.Google Scholar
Lanfranco, F, Gromoll, J, von Eckardstein, S, Herding, EM, Nieschlag, E, Simoni, M. Role of sequence variations of the GnRH receptor and G protein-coupled receptor 54 gene in male idiopathic hypogonadotropic hypogonadism. Eur J Endocrinol 2005;153:845–52.Google Scholar
Han, SK, Gottsch, ML, Lee, KJ, et al. Activation of gonadotropin-releasing hormone neurons by kisspeptin as a neuroendocrine switch for the onset of puberty. J Neurosci 2005;25:11349–56.Google Scholar
Topaloglu, AK, Reimann, F, Guclu, M, et al. TAC3 and TACR3 mutations in familial hypogonadotropic hypogonadism reveal a key role for neurokinin B in the central control of reproduction. Nat Genet 2009;41:354–8.Google Scholar
Skorupskaite, K, George, JT, Anderson, RA. The kisspeptin–GnRH pathway in human reproductive health and disease. Hum Reprod Update 2014;20:485500.Google Scholar
Moore, AM, Coolen, LM, Porter, DT, Goodman, RL, Lehman, MN. KNDy cells revisited. Endocrinology 2018;159:3219–34.Google Scholar
Pitteloud, N, Dwyer, AA, DeCruz, S, et al. Inhibition of luteinizing hormone secretion by testosterone in men requires aromatization for its pituitary but not its hypothalamic effects: evidence from the tandem study of normal and gonadotropin-releasing hormone-deficient men. J Clin Endocrinol Metab 2008;93:784–91.Google Scholar
Hayes, FJ, Seminara, SB, DeCruz, S, Boepple, PA, Crowley, WF Jr. Aromatase inhibition in the human male reveals a hypothalamic site of estrogen feedback. J Clin Endocrinol Metab 2000;85:3027–35.Google Scholar
Smith, JT, Dungan, HM, Stoll, EA, et al. Differential regulation of KiSS-1 mRNA expression by sex steroids in the brain of the male mouse. Endocrinology 2005;146:2976–84.Google Scholar
Mayer, CM, Fick, LJ, Gingerich, S, Belsham, DD. Hypothalamic cell lines to investigate neuroendocrine control mechanisms. Front Neuroendocrinol 2009;30:405–23.Google Scholar
Hu, L, Gustofson, RL, Feng, H, et al. Converse regulatory functions of estrogen receptor-alpha and -beta subtypes expressed in hypothalamic gonadotropin-releasing hormone neurons. Mol Endocrinol 2008;22:2250–9.Google Scholar
Kaprara, A, Huhtaniemi, IT. The hypothalamus–pituitary–gonad axis: tales of mice and men. Metabolism 2018;86:317.Google Scholar
Campo, S, Andreone, L, Ambao, V, et al. Hormonal regulation of follicle-stimulating hormone glycosylation in males. Front Endocrinol 2019;10:17.Google Scholar
Wide, L, Naessen, T, Sundstrom-Poromaa, I, Eriksson, K. Sulfonation and sialylation of gonadotropins in women during the menstrual cycle, after menopause, and with polycystic ovarian syndrome and in men. J Clin Endocrinol Metab 2007;92:4410–17.Google Scholar
Das, N, Kumar, TR. Molecular regulation of follicle-stimulating hormone synthesis, secretion and action. J Mol Endocrinol 2018;60:R131–55.Google Scholar
Manna, PR, Joshi, L, Reinhold, VN, et al. Synthesis, purification and structural and functional characterization of recombinant form of a common genetic variant of human luteinizing hormone. Hum Mol Genet 2002;11:301–15.Google Scholar
Sairam, MR, Fleshner, P. Inhibition of hormone-induced cyclic AMP production and steroidogenesis in interstitial cells by deglycosylated lutropin. Endocrinology 1981;22:4154.Google Scholar
Simoni, M, Gromoll, J, Nieschlag, E. The follicle-stimulating hormone receptor: biochemistry; molecular biology, physiology, and pathophysiology. Endocr Rev 1997;18:739–73.Google Scholar
Tapanainen, JS, Aittomaki, K, Min, J, et al. Men homozygous for an inactivating mutation of the follicle-stimulating hormone (FSH) receptor gene present variable suppression of spermatogenesis and fertility. Nat Genet 1997;15:205–6.Google Scholar
Huhtaniemi, I, Alevizaki, M. Gonadotrophin resistance. Best Pract Res Clin Endocrinol Metab 2006;20:561–76.Google Scholar
Caroppo, E. Endocrine genetic defects. In: Skinner, MK, ed. Encyclopedia of Reproduction, 2nd ed, Vol. 4. Waltham, MA: Academic Press, 2018; pp. 252–7.Google Scholar
Thompson, IR, Kaiser, UB. GnRH pulse frequency-dependent differential regulation of LH and FSH gene expression. Mol Cell Endocrinol 2014;385:2835.Google Scholar
Fortin, J, Lamba, P, Wang, Y, Bernard, DJ. Conservation of mechanisms mediating gonadotrophin-releasing hormone 1 stimulation of human luteinizing hormone beta subunit transcription. Mol Hum Reprod 2009;15:7787.Google Scholar
Boepple, PA, Hayes, FJ, Dwyer, AA, et al. Relative roles of inhibin B and sex steroids in the negative feedback regulation of follicle-stimulating hormone in men across the full spectrum of seminiferous epithelium function. J Clin Endocrinol Metab 2008;93:1809–14.Google Scholar
Wijayarathna, R, de Kretser, DM. Activins in reproductive biology and beyond. Hum Reprod Update 2016;22:342–57.Google Scholar
Burger, LL, Haisenleder, DJ, Wotton, GM, Aylor, KW, Dalkin, AC, Marshall, JC. The regulation of FSHβ transcription by gonadal steroids: testosterone and estradiol modulation of the activin intracellular signaling pathway. Am J Physiol Endocrinol Metab 2007;293:E277–85.Google Scholar
Campo, S, Ambao, V, Creus, S, et al. Carbohydrate complexity and proportions of serum FSH isoforms in the male: lectin-based studies. Mol Cell Endocrinol 2007;260–2:197204.Google Scholar
Bagatell, CJ, Dahl, KD, Bremner, WJ. The direct pituitary effect of testosterone to inhibit gonadotropin secretion in men is partially mediated by aromatization to estradiol. J Androl 1994;15:1521.Google Scholar
Schnorr, JA, Bray, MJ, Veldhuis, JD. Aromatization mediates testosterone’s short-term feedback restraint of 24-hour endogenously driven and acute exogenous gonadotropin-releasing hormone-stimulated luteinizing hormone and follicle-stimulating hormone secretion in men. J Clin Endocrinol Metab 2001;86:2600–6.Google Scholar
Lindzey, J, Wetsel, WC, Couse, JF, Stoker, T, Cooper, R, Korach, KS. Effects of castration and chronic steroid treatments on hypothalamic gonadotropin-releasing hormone content and pituitary gonadotropins in male wild-type and estrogen receptor-α knockout mice. Endocrinology 1998;139:4092–101.Google Scholar
Veldhuis, JD, Takahashi, PY, Keenan, DM, Liu, PY, Mielke, KL, Weist, SM. Age disrupts androgen receptor-modulated negative feedback in the gonadal axis in healthy men. Am J Physiol Endocrinol Metab 2010;299:E675–82.Google Scholar
de Zegher, F, Devlieger, H, Veldhuis, JD. Pulsatile and sexually dimorphic secretion of luteinizing hormone in the human infant on the day of birth. Pediatr Res 1992;32:605–7.Google Scholar
Liu, PY, Beilin, J, Meier, C, et al. Age-related changes in serum testosterone and sex hormone binding globulin in Australian men: longitudinal analyses of two geographically separate regional cohorts. J Clin Endocrinol Metab 2007;92:3599–603.Google Scholar
Kaufman, JM, Vermeulen, A. The decline of androgen levels in elderly men and its clinical and therapeutic implications. Endocr Rev 2005;26:833–76.Google Scholar
Roelfsema, F, Yang, RJ, Liu, PY, Takahashi, PY, Veldhuis, JD. Feedback on LH in testosterone-clamped men depends on the mode of testosterone administration and body composition. J Endocr Soc 2018;3:235–49.Google Scholar
Liu, PY, Pincus, SM, Takahashi, PY, et al. Aging attenuates both the regularity and joint synchrony of LH and testosterone secretion in normal men: analyses via a model of graded GnRH receptor blockade. Am J Physiol Endocrinol Metab 2006;290:E34–41.Google Scholar
Veldhuis, JD, Liu, PY, Takahashi, PY, Keenan, DM. Dynamic testosterone responses to near-physiological LH pulses are determined by the time pattern of prior intravenous LH infusion. Am J Physiol Endocrinol Metab 2012;303:E720–8.Google Scholar
Ascoli, M, Fanelli, F, Segaloff, DL. The lutropin/choriogonadotropin receptor, a 2002 perspective. Endocr Rev 2002;23:141–74.Google Scholar
Lin, T, Calkins, JK, Morris, PL, Vale, W, Bardin, CW. Regulation of Leydig cell function in primary culture by inhibin and activin. Endocrinology 1989;125:2134–40.Google Scholar
Chen, YC, Cochrum, RK, Tseng, MT, et al. Effects of CDB-4022 on Leydig cell function in adult male rats. Biol Reprod 2007;77:1017–26.Google Scholar
Xiao, F, Lan, A, Lin, Z, et al. Impact of CAG repeat length in the androgen receptor gene on male infertility – a meta-analysis. Reprod Biomed Online 2016;33:3949.Google Scholar
Davey, RA, Grossman, M. Androgen receptor structure, function and biology: from bench to bedside. Clin Biochem Rev 2016;37:315.Google Scholar
Goldman, AL, Bhasin, S, Wu, FCW, Krishna, M, Matsumoto, AM, Jasuja, R. A reappraisal of testosterone’s binding in circulation: physiological and clinical implications. Endocr Rev 2017;38:302–24.Google Scholar
Hammes, A, Andreassen, TK, Spoelgen, R, et al. Role of endocytosis in cellular uptake of sex steroids. Cell 2005;122:751–62.Google Scholar
Keenan, DM, Takahashi, PY, Liu, PY, et al. An ensemble model of the male gonadal axis: illustrative application in aging men. Endocrinology 2006;147:2817–28.Google Scholar
Van Eenoo, P, Delbeke, FT. Metabolism and excretion of anabolic steroids in doping control – new steroids and new insights. J Steroid Biochem Mol Biol 2006;101:161–78.Google Scholar
Bulus, SE. Aromatase and estrogen receptor α deficiency. Fertil Steril 2014;101:323–9.Google Scholar
Anawalt, BD, Bebb, RA, Matsumoto, AM, et al. Serum inhibin B levels reflect Sertoli cell function in normal men and men with testicular dysfunction. J Clin Endocrinol Metab 1996;81:3341–5.Google Scholar
Marchetti, C, Hamdane, M, Mitchell, V, et al. Immunolocalization of inhibin and activin alpha and betaB subunits and expression of corresponding messenger RNAs in the human adult testis. Biol Reprod 2003;68:230–5.Google Scholar
Okuma, Y, O’Connor, AE, Hayashi, T, et al. Regulated production of activin A and inhibin B throughout the cycle of the seminiferous epithelium in the rat. J Endocrinol 2006;190:331–40.Google Scholar
Ivell, R, Heng, K, Anand-Ivell, R. Insulin-like factor 3 and the HPG axis in the male. Front Endocrinol (Lausanne) 2014;5:6.Google Scholar
Nef, S, Parada, LF. Cryptorchidism in mice mutant for Insl3. Nat Genet 1999;22:295–9.Google Scholar
Hutson, JM, Southwell, BR, Li, R, et al. The regulation of testicular descent and the effects of cryptorchidism. Endocr Rev 2013;34:725–52.Google Scholar
Bay, K, Virtanen, HE, Hartung, S, et al. Nordic Cryptorchidism Study Group, Toppari, J. Insulin-like factor 3 levels in cord blood and serum from children: effects of age, postnatal hypothalamic–pituitary–gonadal axis activation, and cryptorchidism. J Clin Endocrinol Metab 2007;92:4020–7.Google Scholar
Trabado, S, Maione, L, Bry-Gauillard, H, et al. Insulin-like peptide 3 (INSL3) in men with congenital hypogonadotropic hypogonadism/Kallmann syndrome and effects of different modalities of hormonal treatment: a single-center study of 281 patients. J Clin Endocrinol Metab 2014;99:E268–75.Google Scholar
Kawamura, K, Kumagai, J, Sudo, S, et al. Paracrine regulation of mammalian oocyte maturation and male germ cell survival. Proc Natl Acad Sci U S A 2004;101:7323-8J.Google Scholar
Amory, JK, Page, ST, Anawalt, BD, Coviello, AD, Matsumoto, AM, Bremner, WJ. Elevated end-of-treatment serum INSL3 is associated with failure to completely suppress spermatogenesis in men receiving male hormone contraception. J Androl 2007;28:548–54.Google Scholar

Further Reading

Andersson, KE. Mechanisms of penile erection and basis for pharmacological treatment of erectile dysfunction. Pharmacol Rev 2011;63:811–59.Google Scholar
Lue, TF. Physiology of penile erection and pathophysiology of erectile dysfunction. In: Wein, AJ, Kavoussi, LR, Partin, AW, Peters, CA, eds. Campbell-Walsh Urology, 11th ed. Philadelphia, PA: Elsevier, 2016; pp. 612–42.Google Scholar

References

Herbenick, D, Reece, M, Schick, V, Sanders, SA. Erect penile length and circumference dimensions of 1,661 sexually active men in the United States. J Sex Med 2014;11:93101.Google Scholar
Gontero, P, Oderda, M, Filippini, C, et al. Does kidney transplantation onto the external iliac artery affect the haemodynamic parameters of the cavernosal arteries? Asian J Androl 2012;14:621–5.Google Scholar
Parthiban, S, Hotaling, JM, Kathrins, M, Baftiri, AP, Freels, S, Niederberger, CS. A novel method to determine perineal artery occlusion among male bicyclists. Peer J 2015;3:e1477.Google Scholar
Grossman, JAI, Caldamone, A, Khouri, R, Kenna, DM. Cutaneous blood supply of the penis. Plast Reconstr Surg 1989;83:213–16.Google Scholar
Hsieh, C-H, Huang, Y-P, Tsai, M-H, et al. Tunical outer layer plays an essential role in penile veno-occlusive mechanism evidenced from electrocautery effects to the corpora cavernosa in defrosted human cadavers. Urology 2015;86:1129–36.Google Scholar
Dean, RC, Lue, TF. Physiology of penile erection and pathophysiology of erectile dysfunction. Urol Clin North Am 2005;32:379–95.Google Scholar
Brow, SL, Seftel, AD, Strohl, KP, Herbener, TE. Vasculogenic impotence and cavernosal oxygen tension. Int J Impot Res 2000;12:1922.Google Scholar
Golijanin, D, Singer, E, Davis, R, Bhatt, S, Seftel, A, Dogra, V. Doppler evaluation of erectile dysfunction – Part 2. Int J Impot Res 2007;19:43–8.Google Scholar
Arnow, BA, Desmond, JE, Banner, LL, et al. Brain activation and sexual arousal in healthy, heterosexual males. Brain 2002;125:1014–23.Google Scholar
Simonsen, U, Comerma-Steffensen, S, Andersson, K-E.. Modulation of dopaminergic pathways to treat erectile dysfunction. Basic Clin Pharmacol Toxicol 2016;119:6374.Google Scholar
Bala, A, Minh, H, Nguyen, T, Hellstrom, WJG. Post-SSRI sexual dysfunction: a literature review. Sex Med Rev 2018;6:2934.Google Scholar
Buvat, J. Hyperprolactinemia and sexual function in men: a short review. Int J Impot Res 2003;15:373–7.Google Scholar
Eggleston, JC, Walsh, PC. Radical prostatectomy with preservation of sexual function: pathological findings in the first 100 cases. J Urol 1985;134:1146–8.Google Scholar
Lue, TF, Schmidt, RA, Tanagho, EA. Electrostimulation and penile erection. Urol Int 1985;40:60–4.Google Scholar
Baradaran, N, Awad, M, Gaither, TW, et al. The association of bicycle-related genital numbness and Sexual Health Inventory for Men (SHIM) score: results from a large, multinational, cross-sectional study. BJU Int 2019;124:336–41.Google Scholar
Krassioukov, A, Elliott, S. Neural control and physiology of sexual function: effect of spinal cord injury. Top Spinal Cord Inj Rehabil 2017;23:110.Google Scholar
Traish, A, Kim, NN, Moreland, RB, Goldstein I. Role of alpha adrenergic receptors in erectile function. Int J Impot Res 2000;12 Suppl 1:S48–63.Google Scholar
Martínez-Salamanca, JI, La Fuente, JM, Martínez-Salamanca, E, et al. α 1A-adrenergic receptor antagonism improves erectile and cavernosal responses in rats with cavernous nerve injury and enhances neurogenic responses in human corpus cavernosum from patients with erectile dysfunction secondary to radical prostatectomy. J Sex Med 2016;13:1844–57.Google Scholar
Toda, N, Ayajiki, K, Okamura, T. Nitric oxide and penile erectile function. Pharmacol Ther 2005;106:233–66.Google Scholar
Lin, C-S, Lau, A, Tu, R, Lue, TF. Expression of three isoforms of cGMP-binding cGMP-specific phosphodiesterase (PDE5) in human penile cavernosum. Biochem Biophys Res Commun 2000;268:628–35.Google Scholar
Chitaley, K, Webb, RC, Mills, TM. RhoA/Rho-kinase : a novel player in the regulation of penile erection. Int J Impot Res 2001;13:6772.Google Scholar
Burnett, AL, Nehra, A, Breau, RH, et al. Erectile dysfunction – AUA Guideline – unabridged. Am Urol Assoc Guideline 2018;April:136.Google Scholar
Rosen, RC, Riley, A, Wagner, G, Osterloh, IH, Kirkpatrick, J, Mishra, A. The international index of erectile function (IIEF): a multidimensional scale for assessment of erectile dysfunction. Urology 1997;49:822–30.Google Scholar
Cappelleri, JC, Rosen, RC. The Sexual Health Inventory for Men (SHIM): a 5-year review of research and clinical experience. Int J Impot Res 2005;17:307–19.Google Scholar
Derby, C, Araujo, A, Johannes, C, Feldman, H, McKinlay, J. Measurement of erectile dysfunction in population-based studies: the use of a single question self-assessment in the Massachusetts Male Aging Study. Int J Impot Res 2000;12:197204.Google Scholar
Sweet, G, Shindel, AW. Erectile dysfunction in younger man. AUA Updat Ser 2015;34:18.Google Scholar
Shindel, AW, Nelson, CJ, Naughton, CK, Ohebshalom, M, Mulhall, JP. Sexual function and quality of life in the male partner of infertile couples: prevalence and correlates of dysfunction. J Urol 2008;179:1056–9.Google Scholar
Satkunasivam, R, Ordon, M, Hu, B, et al. Hormone abnormalities are not related to the erectile dysfunction and decreased libido found in many men with infertility. Fertil Steril 2014;101:1594–8.Google Scholar
O’Brien, JH, Lazarou, S, Deane, L, Jarvi, K, Zini, A. Erectile dysfunction and andropause symptoms in infertile men. J Urol 2005;174:1932–4; discussion 1934.Google Scholar
Nehra, A, Jackson, G, Miner, M, et al. The Princeton III Consensus recommendations for the management of erectile dysfunction and cardiovascular disease. Mayo Clin Proc 2012;87:766–78.Google Scholar
Korenman, SG. Epidemiology of erectile dysfunction. Int J Impot Res 2003;15:6371.Google Scholar
Snyder, PJ, Bhasin, S, Cunningham, GR, et al. Effects of testosterone treatment in older men. N Engl J Med 2016;374:611–24.Google Scholar
Silva, AB, Sousa, N, Azevedo, LF, Martins, C. Physical activity and exercise for erectile dysfunction: systematic review and meta-analysis. Br J Sports Med 2017;51:1419–24.Google Scholar
Jorgenson, E, Matharu, N, Palmer, MR, et al. Genetic variation in the SIM1 locus is associated with erectile dysfunction. Proc Natl Acad Sci U S A 2018;115:11018–23.Google Scholar
Melnik, T, Soares, BGO, Nasselo, AG. Psychosocial interventions for erectile dysfunction. Cochrane Database Syst Rev 2007;3:CD004825.Google Scholar
Kim, N, Azadzoi, KM, Goldstein, I, Saenz de Tejada, I. A nitric oxide-like factor mediates nonadrenergic-noncholinergic neurogenic relaxation of penile corpus cavernosum smooth muscle. J Clin Invest 1991;88:112–18.Google Scholar
Burnett, AL, Lowenstein, CJ, Bredt, DS, Chang, TSK, Snyder, SH. Nitric oxide: a physiologic mediator of penile erection. Science 1992;257:401–3.Google Scholar
Goldstein, I, Burnett, AL, Rosen, RC, Park, PW, Stecher, VJ. The serendipitous story of sildenafil: an unexpected oral therapy for erectile dysfunction. Sex Med Rev 2019;7:115–28.Google Scholar
Boolell, M, Gepi-Attee, S, Gingell, JC, Allen, MJ. Sildenafil, a novel effective oral therapy for male erectile dysfunction. Br J Urol 1996;78:257–61.Google Scholar
Bella, AJ, Brant, WO, Lue, TF, Brock, GB. Non-arteritic anterior ischemic optic neuropathy (NAION) and phosphodiesterase type-5 inhibitors. Can J Urol 2006;13:3233–8.Google Scholar
Zucchi, A, Costantini, E, Scroppo, FI, et al. The first‐generation phosphodiesterase 5 inhibitors and their pharmacokinetic issue. Andrology 2019;7:804–17.Google Scholar
Hellstrom, WJG, Kaminetsky, J, Belkoff, LH, et al. Efficacy of avanafil 15 minutes after dosing in men with erectile dysfunction: a randomized, double-blind, placebo controlled study. J Urol 2015;194:485–92.Google Scholar
Goldstein, I. The hour lecture that changed sexual medicine – the Giles Brindley injection story. J Sex Med 2012;9:337–42.Google Scholar
Brindley, GS. Cavernosal alpha-blockade: a new technique for investigating and treating erectile impotence. Br J Psychiatry 1983;143:332–7.Google Scholar
Seidmon, EJ, Samaha, AM. The pH analysis of papaverine-phentolamine and prostaglandin E1 for pharmacologic erection. J Urol 1989;141:1458–9.Google Scholar
Glina, S, Virag, R, Luis Rhoden, E, Sharlip, ID. Intracavernous injection of papaverine for erectile failure. J Sex Med 2010;7:1331–5.Google Scholar
Shinn-Nan Lin, J, Lin, Y-M, Jou, Y-C, Cheng, J-T. Role of cyclic adenosine monophosphate in prostaglandin E(1)-induced penile erection in rabbits. Eur Urol 1995;28:259–65.Google Scholar
Linet, OI, Ogrinc, FG. Efficacy and safety of intracavernosal alprostadil in men with erectile dysfunction. N Engl J Med 1996;334:873–7.Google Scholar
Baltaci, S, Aydos, K, Kosar, A, Anafarta, K. Treating erectile dysfunction with a vacuum tumescence device: a retrospective analysis of acceptance and satisfaction. Br J Urol 1995;76:757–60.Google Scholar
Chen, J, Mabjeesh, NJ, Greenstein, A. Sildenafil versus the vacuum erection device: patient preference. J Urol 2001;166:1779–81.Google Scholar
Barton, GJ, Carlos, EC, Lentz, AC. Sexual quality of life and satisfaction with penile prostheses. Sex Med Rev 2019;7:178–88.Google Scholar
Gruenwald, I, Kitrey, ND, Appel, B, Vardi, Y. Low‐intensity extracorporeal shock wave therapy in vascular disease and erectile dysfunction: theory and outcomes. Sex Med Rev 2013;1:8390.Google Scholar
Capogrosso, P, Frey, A, Jensen, CFS, et al. Low-intensity shock wave therapy in sexual medicine – clinical recommendations from the European Society of Sexual Medicine (ESSM). J Sex Med 2019;16:1490–505.Google Scholar
Shin, DH, Spitz, A. The evaluation and treatment of delayed ejaculation. Sex Med Rev 2014;2(3–4):121–33.Google Scholar
Giuliano, F, Clement, P. Neuroanatomy and physiology of ejaculation. Annu Rev Sex Res 2005;16:190.Google Scholar
McMahon, CG, Abdo, C, Incrocci, L, et al. Disorders of orgasm and ejaculation in men. J Sex Med 2004;1:5865.Google Scholar
Levin, R. Revisiting post-ejaculation refractory time – what we know and what we do not know in males and in females. J Sex Med 2009;6:2376–89.Google Scholar
Kinsey, AC, Pomeroy, WR, Martin, CE. Sexual behavior in the human male. Am J Public Health 2003;93:894–8.Google Scholar
Holstege, G, Georgiadis, JR, Paans, AM, Meiners, LC, van der Graaf, FH, Reinders, AS. Brain activation during human ejaculation. J Neurosci 2003;23:9185–93.Google Scholar
Dunn, ME, Trost, JE. Male multiple orgasms: a descriptive study. Arch Sex Behav 1989;18:377–87.Google Scholar
Turley, KR, Rowland, DL. Evolving ideas about the male refractory period. BJU Int 2013;112:442–52.Google Scholar
Georgiadis, JR, Reinders, AA, Van der Graaf, FH, Paans, AM, Kortekaas, R. Brain activation during human male ejaculation revisited. Neuroreport 2007;18:553.Google Scholar
Cooper, TG, Weidner, W, Nieschlag, E. The influence of inflammation of the human genital tract on secretion of the seminal markers alpha glucosidase, glyceryl-phosphocholine, carnitine, fructose and citric acid. Int J Androl 1990;13:329–36.Google Scholar
Wolff, H, Bezold, G, Zebhauser, M, Meurer, M. Impact of clinically silent inflammation on male genital tract organs as reflected by biochemical markers in semen. J Androl 1991;12:331–4.Google Scholar
Yokoyama, H. [Gamma-glutamyl transpeptidase (gamma-GTP) in the era of metabolic syndrome]. Nihon Arukoru Yakubutsu Igakkai Zasshi 2007;42:110–24.Google Scholar
Pero, ME, Lombardi, P, Longobardi, V, et al. Influence of gamma glutamyl transferase and alkaline phosphatase 55. activity on in vitro fertilization of bovine frozen/thawed semen. Ital J Anim Sci 2017;16:390–2.Google Scholar
Edström, AM, Malm, J, Frohm, B, et al. The major bactericidal activity of human seminal plasma is zinc-dependent and derived from fragmentation of the semenogelins. J Immunol 2008;181:3413–21.Google Scholar
Sheu, G, Revenig, LM, Hsiao, W. Physiology of ejaculation. In: Mulhall, JP, Hsiao, W, eds. Men’s Sexual Health and Fertility. New York, NY: Springer Science, 2014; pp. 1329.Google Scholar
Master, VA, Turek, PJ. Ejaculatory physiology and dysfunction. Urol Clin North Am 2001;28:363–75.Google Scholar
Shafik, A, El-Sibai, A. Mechanism of ejection during ejaculation: identification of a urethrocavernosus reflex. Arch Androl 2000;44:7783.Google Scholar
Shafik, A. Pelvic floor muscles and sphincters during erection and ejaculation: experimental study. Arch Androl 1997;39:71–8.Google Scholar
Müller, A, Mulhall, JP (n.d.). Erection, emission, and ejaculation: mechanisms of control. Infert Male, pp. 132–52.Google Scholar
Giuliano, F. Neurophysiology of erection and ejaculation. J Sex Med 2011;8(Suppl. 4):310.Google Scholar
Gerstenberg, TC, Levin, RJ, Wagner, G. Erection and ejaculation in man – assessment of the electromyographic activity of the bulbocavernosus and ischiocavernosus muscles. Br J Urol 1990;65:395402.Google Scholar
McKenna, KE, Chung, SK, McVary, KT. A model for the study of sexual function in anesthetized male and female rats. Am J Physiol 1991;261:R1276–85.Google Scholar
McKenna, KE, Chung, SK, McVary, KT. A model for the study of sexual function in anesthetized male and female rats. Am J Physiol. 1991;261:R1276–85.Google Scholar
Larsson, K, van Dis, H. Seminal discharge following intracranial electrical stimulation. Brain Res 1970;23:381–6.Google Scholar
Alwaal, A, Breyer, BN, Lue, TF. Normal male sexual function: emphasis on orgasm and ejaculation. Fertil Steril 2015;104:1051–60.Google Scholar
Bancila, M, Verge, D, Rampin, O, et al. 5-Hydroxytryptamine 2C receptors on spinal neurons controlling penile erection in the rat. Neuroscience 1999;92:1523–37.Google Scholar
Giuliano, F, Clement, P. Physiology of ejaculation: emphasis on serotonergic control. Eur Urol 2005;48:408–17.Google Scholar
Chehensse, C, Bahrami, S, Denys, P, Clement, P, Bernabe, J, Giuliano, F. The spinal control of ejaculation revisited: a systematic review and meta-analysis of anejaculation in spinal cord injured patients. Hum Reprod Update 2013;19:507–26.Google Scholar
Heeb, MM, Yahr, P. Anatomical and functional connections among cell groups in the gerbil brain that are activated with ejaculation. J Comp Neurol 2001;439:248–58.Google Scholar
Heeb, MM, Yahr, P. Anatomical and functional connections among cell groups in the gerbil brain that are activated with ejaculation. J Comp Neurol 2001;439:248–58.Google Scholar
Coolen, LM, Peters, HJ, Veening, JG. Anatomical interrelationships of the medial preoptic area and other brain regions activated following male sexual behavior: a combined fos and tract-tracing study. J Comp Neurol 1998;397:421–35.Google Scholar
Marson, L, McKenna, KE. Stimulation of the hypothalamus initiates the urethrogenital reflex in male rats. Brain Res 1994;638:103–8.Google Scholar
Arendash, GW, Gorski, RA. Effects of discrete lesions of the sexually dimorphic nucleus of the preoptic area or other medial preoptic regions on the sexual behavior of male rats. Brain Res Bull 1983;10:147–54.Google Scholar
Simerly, RB, Swanson, LW. Projections of the medial preoptic nucleus: a Phaseolus vulgaris leucoagglutinin anterograde tract-tracing study in the rat. J Comp Neurol 1988;270:209–42.Google Scholar
Rizvi, TA, Ennis, M, Shipley, MT. Reciprocal connections between the medial preoptic area and the midbrain periaqueductal gray in rat: a WGA-HRP and PHA-L study. J Comp Neurol 1992;315:115.Google Scholar
Marson, L, McKenna, KE. A role for 5-hydroxytryptamine in descending inhibition of spinal sexual reflexes. Exp Brain Res 1990;88:313–20.Google Scholar
Marson, L, McKenna, KE. The identification of a brainstem site controlling spinal sexual reflexes in male rats. Brain Res 1990;515:303–8.Google Scholar
Marson, L. Lesions of the periaqueductal gray block the medial preoptic area-induced activation of the urethrogenital reflex in male rats. Neurosci Lett 2004;367:278–82.Google Scholar
Saper, CB, Loewy, AD, Swanson, LW, Cowan, WM. Direct hypothalamo-autonomic connections. Brain Res 1976;117:305–12.Google Scholar
Luiten, PG, Ter Horst, GJ, Karst, H, Steffens, AB. The course of paraventricular hypothalamic efferents to autonomic structures in medulla and spinal cord. Brain Res 1985;329:374–8.Google Scholar
Truitt, WA, Coolen, LM. Identification of a potential ejaculation generator in the spinal cord. Science 2002;297:1566–9.Google Scholar
Brindley, GS, Sauerwein, D, Hendry, WF. Hypogastric plexus stimulators for obtaining semen from paraplegic men. Br J Urol 1989;64:72–7.Google Scholar
Coolen, LM, Veening, JG, Wells, AB, Shipley, MT. Afferent connections of the parvocellular subparafascicular thalamic nucleus in the rat: evidence for functional subdivisions. J Comp Neurol 2003;463:132–56.Google Scholar
Borgdorff, AJ, Bernabé, J, Denys, P, Alexandre, L, Giuliano, F. Ejaculation elicited by microstimulation of lumbar spinothalamic neurons. Eur Urol 2008;54:449–56.Google Scholar
Clement, P, Giuliano, F. Physiology and pharmacology of ejaculation. Basic Clin Pharmacol Toxicol 2016;119:1825.Google Scholar
Nadelhaft, I, McKenna, KE. Sexual dimorphism in sympathetic preganglionic neurons of the rat hypogastric nerve. J Comp Neurol 1987;256:308–15.Google Scholar
Owman, C, Stjernquist, M. The peripheral nervous system. In: Bjorklund, A, Hokfelt, T, Owman, C, eds. Handbook of Chemical Neuroanatomy. Amsterdam: Elsevier Science, 1988; pp. 445544.Google Scholar
Nadelhaft, I, Booth, AM. The location and morphology of preganglionic neurons and the distribution of visceral afferents from the rat pelvic nerve: a horseradish peroxidase study. J Comp Neurol 1984;226:238–45.Google Scholar
Schroder, HD. Anatomical and pathoanatomical studies on the spinal efferent systems innervating pelvic structures. 1. Organization of spinal nuclei in animals. 2. The nucleus X-pelvic motor system in man. J Auton Nerv Syst 1985;14:2348.Google Scholar
Halata, Z, Munger, BL. The neuroanatomical basis for the protopathic sensibility of the human glans penis. Brain Res 1986;371:205–30.Google Scholar
Baron, R, Janig, W. Afferent and sympathetic neurons projecting into lumbar visceral nerves of the male rat. J Comp Neurol 1991;314:429–36.Google Scholar
McKenna, KE, Nadelhaft, I. The organization of the pudendal nerve in the male and female rat. J Comp Neurol 1986;248:532–49.Google Scholar
Comarr, A. Sexual function among patients with spinal cord injury. Urol Int 1970;25:134–68.Google Scholar
Chapple, CR, Aubry, ML, James, S, et al. Characterization of human prostatic adrenoceptors using pharmacology receptor binding and localization. Br J Urol 1989;63:487–96.Google Scholar
Adrian, TE, Guj, Allen JM, et al. Neuropeptide Y in the human male genital tract. Life Sci 1984;35:2643–8.Google Scholar
Lincoln, J, Burnstocl, G. Autonomic innervation of the urinary bladder and urethra. In: Maggi, CA, ed. Nervous Control of the Urogenital System. Chur, Switzerland: Harwood Academic, 1993; pp. 3368.Google Scholar
Morrison, J, Steers, WD, Brading, A, et al. Neurophysiology and neuropharmacology. In: Abrams, P, Cardozo, L, Khoury, S, Wein, A, eds. Incontinence. Plymouth: Health Publication, 2005: pp. 85164.Google Scholar
Bates, JN, Kohn, TP, Pastuszak, AW. Effect of thyroid hormone derangements on sexual function in men and women. Sex Med Rev 2020;8:217–30.Google Scholar
Abu El-Hamd, M, Farah, A. Possible role of serum testosterone, gonadotropins and prolactin in patients with premature ejaculation. Andrologia 2018;50(1).Google Scholar
Corona, G, Jannini, EA, Vignozzi, L, Rastrelli, G, Maggi, M. The hormonal control of ejaculation. Nat Rev Urol 2012;9:508–19.Google Scholar
Schulster, M, Bernie, AM, Ramasamy, R. The role of estradiol in male reproductive function. Asian J Androl 2016;18:435–40.Google Scholar
Taylor, D, Paton, C, Kerwin, R (eds). The Maudsley Prescribing Guidelines. London: Informa Healthcare, 2007.Google Scholar
Higgins, A, Nash, M, Lynch, AM. Antidepressant-associated sexual dysfunction: impact, effects, and treatment. Drug Healthc Patient Saf 2010;2:141–50.Google Scholar
Clément, P, Pozzato, C, Heidbreder, C, Alexandre, L, Giuliano, F, Melotto, S. Delay of ejaculation induced by SB-277011, a selective dopamine D3 receptor antagonist, in the rat. J Sex Med 2009;6:98108.Google Scholar
Clément, P, Bernabé, J, Kia, HK, Alexandre, L, Giuliano, F. D2-like receptors mediate the expulsion phase of ejaculation elicited by 8-hydroxy-2-(di-N-propylamino) tetralin in rats. J Pharmacol Exp Ther 2006;316:830–4.Google Scholar
Clément, P, Bernabé, J, Gengo, P, et al. Supraspinal site of action for the inhibition of ejaculatory reflex by dapoxetine. Eur Urol 2007;51:825–32.Google Scholar
Stafford, SA, Bowery, NG, Tang, K, Coote, JH. Activation by p-chloroamphetamine of the spinal ejaculatory pattern generator in anaesthetized male rats. Neuroscience 2006;140:1031–40.Google Scholar
Giuliano, F, Clement, P. Serotonin and premature ejaculation: from physiology to patient management. Eur Urol 2006;50:454–66.Google Scholar
Martin, E, Berka, V, Tsai, AL, Murad, F. Soluble guanylyl cyclase: the nitric oxide receptor. Methods Enzymol 2005;396:478–92.Google Scholar
Hull, EM, Lumley, LA, Matuszewich, L, Dominguez, J, Moses, J, Lorrain, DS. The roles of nitric oxide in sexual function of male rats. Neuropharmacology 1994;33:1499–504.Google Scholar
Dixon, JS, Jen, PY. Development of nerves containing nitric oxide synthase in the human male urogenital organs. Br J Urol 1995;76:719–25.Google Scholar
Calzo, P, et al. Erectile dysfunction in a sample of sexually active young adult men from a U.S. cohort: demographic, metabolic and mental health correlates. J Urol 2021 Feb; 2015 (2):539544. PMID: 32935616Google Scholar

Further Reading

Ben Maamar, M, Sadler-Riggleman, I, Skinner, MK. In: Skinner, MK, ed. Encyclopedia of Reproduction, 2nd ed, Vol. 5. Oxford: Academic Press, 2018; pp. 117–23.Google Scholar
Kubsad, D, Ben Maamar, M, Skinner, MK. Developmental epigenetic analysis of sperm. In: Skinner, MK, ed. Encyclopedia of Reproduction, 2nd ed, Vol. 5. Oxford: Academic Press, 2018; pp. 239–44.Google Scholar
Nilsson, EE, Sadler-Riggleman, I, Skinner, MK. Environmentally induced epigenetic transgenerational inheritance of disease. Environ Epigenet 2018;4:dvy016.Google Scholar

References

Sharlip, ID, Jarow, JP, Belker, AM, et al. Best practice policies for male infertility. Fertil Steril 2002;77:873–82.Google Scholar
Kitamura, A, Miyauchi, N, Hamada, H, et al. Epigenetic alterations in sperm associated with male infertility. Congenit Anom (Kyoto) 2015;55:133–44.Google Scholar
Jodar, M, Selvaraju, S, Sendler, E, Diamond, MP, Krawetz, SA. The presence, role and clinical use of spermatozoal RNAs. Hum Reprod Update 2013;19:604–24.Google Scholar
Minocherhomji, S, Athalye, AS, Madon, PF, Kulkarni, D,Uttamchandani, SA, Parikh, FR. A case-control study identifying chromosomal polymorphic variations as forms of epigenetic alterations associated with the infertility phenotype. Fertil Steril 2009;92:8895.Google Scholar
Rull, K, Nagirnaja, L, Laan, M. Genetics of recurrent miscarriage: challenges, current knowledge, future directions. Front Genet 2012;3:34.Google Scholar
Jenkins, TG, Aston, KI, James, ER, Carrell, DT. Sperm epigenetics in the study of male fertility, offspring health, and potential clinical applications. Syst Biol Reprod Med 2017;63:6976.Google Scholar
Holliday, R, Pugh, JE. DNA modification mechanisms and gene activity during development. Science 1975;187:226–32.Google Scholar
Singer, J, Roberts-Ems, J, Riggs, AD. Methylation of mouse liver DNA studied by means of the restriction enzymes msp I and hpa II. Science 1979;203:1019–21.Google Scholar
Liu, MY, DeNizio, JE, Schutsky, EK, Kohli, RM. The expanding scope and impact of epigenetic cytosine modifications. Curr Opin Chem Biol 2016;33:6773.Google Scholar
Loidl, P. Histone acetylation: facts and questions. Chromosoma 1994;103:441–9.Google Scholar
Davie, JR. Covalent modifications of histones: expression from chromatin templates. Curr Opin Genet Dev 1998;8:173–8.Google Scholar
Kornfeld, JW, Bruning, JC. Regulation of metabolism by long, non-coding RNAs. Front Genet 2014;5:57.Google Scholar
Yaniv, M. Chromatin remodeling: from transcription to cancer. Cancer Genet 2014;207:352–7.Google Scholar
Reik, W, Surani, MA. Germline and pluripotent stem cells. Cold Spring Harb Perspect Biol 2015;7:a019422.Google Scholar
Nilsson, E, Sadler-Riggleman, I, Skinner, MK. Environmentally induced epigenetic transgenerational inheritance of disease. Environ Epigenet 2018;4:dvy016.Google Scholar
Guerrero-Bosagna, C, Skinner, J. Environmentally induced epigenetic transgenerational inheritance of male infertility. Curr Opin Genet Dev 2014;26:7988.Google Scholar
Anway, MD, Cupp, AS, Uzumcu, M, Skinner, MK. Epigenetic transgenerational actions of endocrine disruptors and male fertility. Science 2005;308:1466–9.Google Scholar
Oliva, R, Castillo, J. Proteomics and the genetics of sperm chromatin condensation. Asian J Androl 2011;13:2430.Google Scholar
Murphy, KE, Murphy, PJ, Carrell, JT. Epigenetic changes in the paternal germline. In: Tollefsbol, T, ed. Transgenerational Epigenetics. London: Academic Press, 2014; pp. 4355.Google Scholar
Gunes, S, Kulac, T. The role of epigenetics in spermatogenesis. Turk J Urol 2013;39:181–7.Google Scholar
Portela, A, Esteller, M. Epigenetic modifications and human disease. Nat Biotechnol 2010;28:1057–68.Google Scholar
Kim, YZ. Altered histone modifications in gliomas. Brain Tumor Res Treat 2014;2:721.Google Scholar
Campos, E, Reinberg, D. Histones: annotating chromatin. Annu Rev Genet 2009;43:559–99.Google Scholar
Godde, JS, Ura, K. Dynamic alterations of linker histone variants during development. Int J Dev Biol 2009;53:215–24.Google Scholar
Govin, J, Caron, C, Lestrat, C, Rousseaux, S, Khochbin, S. The role of histones in chromatin remodelling during mammalian spermiogenesis. Eur J Biochem 2004;271:3459–69.Google Scholar
Montellier, E, Boussouar, F, Rousseaux, S, et al. Chromatin-to-nucleoprotamine transition is controlled by the histone H2B variant TH2B. Genes Dev 2013;27:1680–92.Google Scholar
Samson, M, Jow, MM, Wong, CC, et al. The specification and global reprogramming of histone epigenetic marks during gamete formation and early embryo development in C. elegans. PLoS Genet 2014;10:e1004588.Google Scholar
Skinner, MK. Endocrine disruptor induction of epigenetic transgenerational inheritance of disease. Mol Cell Endocrinol 2014;398:412.Google Scholar
Skinner, MK. Endocrine disruptors in 2015: epigenetic transgenerational inheritance. Nat Rev Endocrinol 2016;12:6870.Google Scholar
Layman, LC. The genetic basis of female reproductive disorders: etiology and clinical testing. Mol Cell Endocrinol 2013;370:138–48.Google Scholar
Houshdaran, S, Cortessis, VK, Siegmund, K, Yang, A, Laird, PW, Sokol, RZ. Widespread epigenetic abnormalities suggest a broad DNA methylation erasure defect in abnormal human sperm. PLoS ONE 2007;2:e1289.Google Scholar
Hartmann, S, Bergmann, M, Bohle, RM, Weidner, W, Steger, K. Genetic imprinting during impaired spermatogenesis. Mol Hum Reprod 2006;12:407–11.Google Scholar
Montjean, D, Ravel, C, Benkhalifa, M, et al. Methylation changes in mature sperm deoxyribonucleic acid from oligozoospermic men: assessment of genetic variants and assisted reproductive technology outcome. Fertil Steril 2013;100:1241–7.Google Scholar
Manipalviratn, S, DeCherney, A, Segars, J. Imprinting disorders and assisted reproductive technology. Fertil Steril 2009;91:305–15.Google Scholar
DeAngelis, AM, Martini, AE, Owen, CM. Assisted reproductive technology and epigenetics. Semin Reprod Med 2018;36: 221–32.Google Scholar
Das, L, Parbin, S, Pradhan, N, Kausar, C, Patra, SK. Epigenetics of reproductive infertility. Front Biosci (Schol Ed) 2017;9:509–35.Google Scholar
Marques, CJ, Costa, P, Vaz, B, et al. Abnormal methylation of imprinted genes in human sperm is associated with oligozoospermia. Mol Hum Reprod 2008; 14:6774.Google Scholar
Li, B, Li, JB, Xiao, XF, et al. Altered DNA methylation patterns of the H19 differentially methylated region and the DAZL gene promoter are associated with defective human sperm. PLoS ONE 2013;8:e71215.Google Scholar
Stuppia, L, Franzago, M, Ballerini, P, Gatta, V, Antonucci, I. Epigenetics and male reproduction: the consequences of paternal lifestyle on fertility, embryo development, and children lifetime health. Clin Epigenetics 2015;7:120.Google Scholar
Aston, KI, Uren, PJ, Jenkins, TG, et al. Aberrant sperm DNA methylation predicts male fertility status and embryo quality. Fertil Steril 2015;104:1388–97.Google Scholar
Bannister, AJ, Kouzarides, T. Regulation of chromatin by histone modifications. Cell Res 2011;21:381–95.Google Scholar
Hazzouri, M, Pivot-Pajot, C, Faure, AK, et al. Regulated hyperacetylation of core histones during mouse spermatogenesis: involvement of histone deacetylases. Eur J Cell Biol 2000;79:950–60.Google Scholar
Steilmann, C, Cavalcanti, MC, Bartkuhn, M, et al. The interaction of modified histones with the bromodomain testis-specific (BRDT) gene and its mRNA level in sperm of fertile donors and subfertile men. Reproduction 2010;140:435–43.Google Scholar
Okada, Y, Scott, G, Ray, MK, Mishina, Y, Zhang, Y. Histone demethylase JHDM2A is critical for Tnp1 and Prm1 transcription and spermatogenesis. Nature 2007;450:119–23.Google Scholar
Sonnack, V, Failing, K, Bergmann, M, Steger, K. Expression of hyperacetylated histone H4 during normal and impaired human spermatogenesis. Andrologia 2002;34:384–90.Google Scholar
Paradowska, AS, Miller, D, Spiess, AN, et al. Genome wide identification of promoter binding sites for H4K12ac in human sperm and its relevance for early embryonic development. Epigenetics 2012;7:1057–70.Google Scholar
Lee, MG, Wynder, C, Cooch, N, Shiekhattar, R. An essential role for CoREST in nucleosomal histone 3 lysine 4 demethylation. Nature 2005;437:432–5.Google Scholar
Bao, J, Bedford, MT. Epigenetic regulation of the histone-to-protamine transition during spermiogenesis. Reproduction 2016;151:R55–70.Google Scholar
Shi, Y, Lan, F, Matson, C, et al. Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell 2004;119:941–53.Google Scholar
Waterland, RA, Michels, KB. Epigenetic epidemiology of the developmental origins hypothesis. Annu Rev Nutr 2007;27:363–88.Google Scholar
Liu, Z, Zhou, S, Liao, L, Chen, X, Meistrich, M, Xu, J. Jmjd1a demethylase-regulated histone modification is essential for cAMP-response element modulator-regulated gene expression and spermatogenesis. J Biol Chem 2010;285:2758–70.Google Scholar
Gomes, AQ, Nolasco, S, Soares, H. Non-coding RNAs: multi-tasking molecules in the cell. Int J Mol Sci 2013;14:16010–39.Google Scholar
Saxe, JP, Lin, H. Small noncoding RNAs in the germline. Cold Spring Harb Perspect Biol 2011;3:a002717.Google Scholar
Gou, LT, Dai, P, Liu, MF. Small noncoding RNAs and male infertility. Wiley Interdiscip Rev RNA 2014;5:733-45.Google Scholar
Khazaie, Y, Nasr Esfahani, MH. MicroRNA and male infertility: a potential for diagnosis. Int J Fertil Steril 2014;8:113–18.Google Scholar
Luk, AC, Chan, WY, Rennert, OM, Lee, TL. Long noncoding RNAs in spermatogenesis: insights from recent high-throughput transcriptome studies. Reproduction 2014;147:R131–41.Google Scholar
Sharma, R, Biedenharn, KR, Fedor, JM, Agarwal, A. Lifestyle factors and reproductive health: taking control of your fertility. Reprod Biol Endocrinol 2013;11:66.Google Scholar
Cloonan, YK, Holt, VL, Goldberg, J. Male factor infertility: a twin study. Paediatr Perinat Epidemiol 2007;21:229–34.Google Scholar
Cortessis, VK, Thomas, DC, Levine, AJ, et al. Environmental epigenetics: prospects for studying epigenetic mediation of exposure-response relationships. Hum Genet 2012;131:1565–89.Google Scholar
Mirbahai, L, Chipman, JK. Epigenetic memory of environmental organisms: a reflection of lifetime stressor exposures. Mutat Res Genet Toxicol Environ Mutagen 2014;764–5:1017.Google Scholar
Wei, Y, Schatten, H, Sun, QY. Environmental epigenetic inheritance through gametes and implications for human reproduction. Hum Reprod Update 2015;21:194208.Google Scholar
Baccarelli, A, Bollati, V. Epigenetics and environmental chemicals. Curr Opin Pediatr 2009;21:243–51.Google Scholar
Wilhelm-Benartzi, CS, Houseman, EA, Maccani, MA, et al. In utero exposures, infant growth, and DNA methylation of repetitive elements and developmentally related genes in human placenta. Environ Health Perspect 2012;120:296302.Google Scholar
Martinez-Arguelles, DB, Campioli, E, Culty, M, Zirkin, BR, Papadopoulos, V. Fetal origin of endocrine dysfunction in the adult: the phthalate model. J Steroid Biochem Mol Biol 2013;137:517.Google Scholar
Skinner, MK, Manikkam, M, Guerrero-Bosagna, C. Epigenetic transgenerational actions of environmental factors in disease etiology. Trends Endocrinol Metab 2010;21:214–22.Google Scholar
Uzumcu, M, Suzuki, H, Skinner, MK. Effect of the anti-androgenic endocrine disruptor vinclozolin on embryonic testis cord formation and postnatal testis development and function. Reprod Toxicol 2004;18:765–74.Google Scholar
LaRocca, J, Boyajian, A, Brown, C, Smith, SD, Hixon, M. Effects of in utero exposure to bisphenol A or diethylstilbestrol on the adult male reproductive system. Birth Defects Res B Dev Reprod Toxicol 2011;92:526–33.Google Scholar
Park, CJ, Nah, WH, Lee, JE, Oh, YS, Gye, MC. Butyl paraben-induced changes in DNA methylation in rat epididymal spermatozoa. Andrologia 2012;44 Suppl 1:187–93.Google Scholar
Stouder, C, Paoloni-Giacobino, A. Specific transgenerational imprinting effects of the endocrine disruptor methoxychlor on male gametes. Reproduction 2011;141:207–16.Google Scholar
Skinner, MK, Ben Maamar, M, Sadler-Riggleman, I, et al. Alterations in sperm DNA methylation, non-coding RNA and histone retention associate with DDT-induced epigenetic transgenerational inheritance of disease. Epigenetics Chromatin 2018;11:8.Google Scholar
Ben Maamar, M, Sadler-Riggleman, I, Beck, D, et al. Alterations in sperm DNA methylation, non-coding RNA expression, and histone retention mediate vinclozolin-induced epigenetic transgenerational inheritance of disease. Environ Epigenet 2018;4:dvy010.Google Scholar
Sadler-Riggleman, I, Klukovich, R, Nilsson, E, et al. Epigenetic Transgenerational inheritance of testis pathology and Sertoli cell epimutations: generational origins of male infertility. Environ Epigenet 2019;5:dvz013.Google Scholar
Skakkebaek, NE, Rajpert-De Meyts, E, Jorgensen, N, et al. Testicular cancer trends as ‘whistle blowers’ of testicular developmental problems in populations. Int J Androl 2007;30:198204; discussion 204–5.Google Scholar
Singh, KP, Kumari, R, Pevey, C, Jackson, D, DuMond, JW. Long duration exposure to cadmium leads to increased cell survival, decreased DNA repair capacity, and genomic instability in mouse testicular Leydig cells. Cancer Lett 2009;279:8492.Google Scholar
Dubrova, YE. Radiation-induced transgenerational instability. Oncogene 2003;22:7087–93.Google Scholar
Merrifield, M, Kovalchuk, O. Epigenetics in radiation biology: a new research frontier. Front Genet 2013;4:40.Google Scholar
Skinner, MK. Environmental epigenetic transgenerational inheritance and somatic epigenetic mitotic stability. Epigenetics 2011;6:838–42.Google Scholar
Skinner, MK. What is an epigenetic transgenerational phenotype? F3 or F2. Reprod Toxicol 2008;25:26.Google Scholar
Guerrero-Bosagna, C, Savenkova, M, Haque, MM, Nilsson, E, Skinner, MK. Environmentally induced epigenetic transgenerational inheritance of altered Sertoli cell transcriptome and epigenome: molecular etiology of male infertility. PLoS ONE 2013;8:e59922.Google Scholar
Anway, MD, Memon, MA, Uzumcu, M, Skinner, MK. Transgenerational effect of the endocrine disruptor vinclozolin on male spermatogenesis. J Androl 2006;27:868–79.Google Scholar
Manikkam, M, Guerrero-Bosagna, C, Tracey, R, Haque, MM, Skinner, MK. Transgenerational actions of environmental compounds on reproductive disease and identification of epigenetic biomarkers of ancestral exposures. PLoS ONE 2012;7:e31901.Google Scholar
Pastuszak, AW, Lamb, DJ. The genetics of male fertility – from basic science to clinical evaluation. J Androl 2012;33:1075–84.Google Scholar
Matzuk, MM, Lamb, DJ. The biology of infertility: research advances and clinical challenges. Nat Med 2008;14:1197–213.Google Scholar
Sanabria, M, Cucielo, MS, Guerra, MT, et al. Sperm quality and fertility in rats after prenatal exposure to low doses of TCDD: a three-generation study. Reprod Toxicol 2016;65:2938.Google Scholar
Skakkebaek, NE, Rajpert-De Meyts, E, Buck Louis, GM, et al. Male reproductive disorders and fertility trends: influences of environment and genetic susceptibility. Physiol Rev 2016;96:5597.Google Scholar
Jensen, TK, Jacobsen, R, Christensen, K, Nielsen, NC, Bostofte, E. Good semen quality and life expectancy: a cohort study of 43,277 men. Am J Epidemiol 2009;170:559–65.Google Scholar
Eisenberg, ML, Li, S, Cullen, MR, Baker, LC. Increased risk of incident chronic medical conditions in infertile men: analysis of United States claims data. Fertil Steril 2016;105:629–36.Google Scholar
Levine, H, Jorgensen, N, Martino-Andrade, A, et al. Temporal trends in sperm count: a systematic review and meta-regression analysis. Hum Reprod Update 2017;23:646–59.Google Scholar
Marieb, EN, Hoehn, K. The reproductive system. In: Marieb, EN, Hoehn, K, eds. Human Anatomy and Physiology, 9th ed. Pearson, 2012; p. 1264.Google Scholar
Jirtle, RL, Skinner, MK. Environmental epigenomics and disease susceptibility. Nat Rev Genet 2007;8:253–62.Google Scholar
Tortora, GJ, Derrickson, BH. The cellular level of organization. In: Tortora, GJ, Derrickson, BH, eds. Principles of Anatomy and Physiology, 14th ed. Hoboken, NJ: John Wiley & Sons, 2014; pp. 59105.Google Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×