Skip to main content Accessibility help
×
Hostname: page-component-76fb5796d-x4r87 Total loading time: 0 Render date: 2024-04-29T12:36:12.430Z Has data issue: false hasContentIssue false

Chapter 3 - Maturation and Function of Sperm

from 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

Summary

Human sperm are created in the testis via a highly regulated and complex hormonal pathway arising from the hypothalamic–pituitary–testicular (HPT) axis under complex paracrine control by growth factors and cytokines. Following their creation, sperm must travel through the seminiferous tubules, epididymis, vas deferens, ejaculatory ducts, and urethra until they are expelled through ejaculation into the female genital tract. Fertilization capabilities of the sperm require morphologic and molecular changes that are acquired during transit of sperm through the male and female reproductive tracts.

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

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.CrossRefGoogle ScholarPubMed
Jones, RC. Evolution of the vertebrate epididymis. J Reprod Fertil Suppl 1998;53:163–81.Google ScholarPubMed
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.Google Scholar
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.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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

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
×