Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-pftt2 Total loading time: 0 Render date: 2024-05-16T21:10:30.527Z Has data issue: false hasContentIssue false

Chapter 17 - Laboratory Evaluation of the Infertile Male

from Section 3 - Laboratory Diagnosis 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

Sperm are critical for fertility. Thus, a properly collected semen analysis is the keystone of the laboratory evaluation of the infertile male. Combined with the history and physical examination, semen analysis can suggest an etiology for infertility and direct the clinician towards further evaluation and treatment options. This chapter will discuss the components of the standard semen analysis and their interpretation, as well as commonly ordered adjunct semen, hormonal, and genetic testing.

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

World Health Organization. WHO Laboratory Manual for the Examination and Processing of Human Semen, 5th ed. Geneva: World Health Organization, 2010.Google Scholar
Zavos, PM, Goodpasture, JC. Clinical improvements of specific seminal deficiencies via intercourse with a seminal collection device versus masturbation. Fertil Steril 1989;51:190–3.Google Scholar
Sofikitis, NV, Miyagawa, I. Endocrinological, biophysical, and biochemical parameters of semen collected via masturbation versus sexual intercourse. J Androl 1993;14:366–73.Google Scholar
Elzanaty, S, Malm, J. Comparison of semen parameters in samples collected by masturbation at a clinic and at home. Fertil Steril 2008;89:1718–22.Google Scholar
Purvis, K, Magnus, O, Morkas, L, Abyholm, T, Rui, H. Ejaculate composition after masturbation and coitus in the human male. Int J Androl 1986;9:401–6.CrossRefGoogle ScholarPubMed
Yamamoto, Y, Sofikitis, N, Mio, Y, Miyagawa, I. Influence of sexual stimulation on sperm parameters in semen samples collected via masturbation from normozoospermic men or cryptozoospermic men participating in an assisted reproduction programme. Andrologia 2000;32:131–8.Google Scholar
Handelsman, DJ, Sivananathan, T, Andres, L, Bathur, F, Jayadev, V, Conway, AJ. Randomised controlled trial of whether erotic material is required for semen collection: impact of informed consent on outcome. Andrology 2013;1:943–7.Google Scholar
Guzick, DS, Overstreet, JW, Factor-Litvak, P, et al. Sperm morphology, motility, and concentration in fertile and infertile men. N Engl J Med 2001;345:1388–93.Google Scholar
Huang, XF. Reference limits: limited references in laboratories worldwide. Asian J Androl 2010;12:447–8.Google Scholar
Iwamoto, T, Nozawa, S, Yoshiike, M, et al. Semen quality of 324 fertile Japanese men. Hum Reprod 2006;21:760–5.Google Scholar
Jorgensen, N, Andersen, AG, Eustache, F, Irvine, DS, Suominen, J, Petersen, JH, et al. Regional differences in semen quality in Europe. Hum Reprod 2001;16:1012–9.Google Scholar
Redmon, JB, Thomas, W, Ma, W, et al. Semen parameters in fertile US men: the Study for Future Families. Andrology 2013;1:806–14.Google Scholar
Alvarez, C, Castilla, JA, Martinez, L, Ramirez, JP, Vergara, F, Gaforio, JJ. Biological variation of seminal parameters in healthy subjects. Hum Reprod 2003;18:2082–8.Google Scholar
Blickenstorfer, K, Voelkle, M, Xie, M, Frohlich, A, Imthurn, B, Leeners, B. Are WHO recommendations to perform 2 consecutive semen analyses for reliable diagnosis of male infertility still valid? J Urol 2019;201:783–91.Google Scholar
Amann, RP, Chapman, PL. Total sperm per ejaculate of men: obtaining a meaningful value or a mean value with appropriate precision. J Androl 2009;30:642–9.Google Scholar
Andrade-Rocha, FT. Physical analysis of ejaculate to evaluate the secretory activity of the seminal vesicles and prostate. Clin Chem Lab Med 2005;43:1203–10.Google ScholarPubMed
Robert, M, Gibbs, BF, Jacobson, E, Gagnon, C. Characterization of prostate-specific antigen proteolytic activity on its major physiological substrate, the sperm motility inhibitor precursor/semenogelin I. Biochemistry 1997;36):3811–19.Google Scholar
Mandal, A, Bhattacharyya, AK. Biochemical parameters of slowly liquefying human ejaculates. Arch Androl 1988;20:141–5.Google Scholar
Wilson, VB, Bunge, RG. Infertility and semen non-liquefaction. J Urol 1975;113:509–10.Google Scholar
Elzanaty, S, Malm, J, Giwercman, A. Visco-elasticity of seminal fluid in relation to the epididymal and accessory sex gland function and its impact on sperm motility. Int J Androl 2004;27:94100.Google Scholar
Esfandiari, N, Burjaq, H, Gotlieb, L, Casper, RF. Seminal hyperviscosity is associated with poor outcome of in vitro fertilization and embryo transfer: a prospective study. Fertil Steril 2008;90:1739–43.CrossRefGoogle ScholarPubMed
Esfandiari, N, de Lamirande, E, Gukturk, A, et al. Seminal hyperviscosity is not associated with semenogelin degradation or sperm deoxyribonucleic acid damage: a prospective study of infertile couples. Fertil Steril 2014;101:1599–603.CrossRefGoogle ScholarPubMed
Aydemir, B, Onaran, I, Kiziler, AR, Alici, B, Akyolcu, MC. The influence of oxidative damage on viscosity of seminal fluid in infertile men. J Androl 2008;29:41–6.Google Scholar
Layali, I, Tahmasbpour, E, Joulaei, M, Jorsaraei, SG, Farzanegi, P. Total antioxidant capacity and lipid peroxidation in semen of patient with hyperviscosity. Cell J 2015;16:554–9.Google ScholarPubMed
Gonzalez-Estrella, JA, Coney, P, Ostash, K, Karabinus, D. Dithiothreitol effects on the viscosity and quality of human semen. Fertil Steril 1994;62:1238–43.Google Scholar
Kussler, AP, Pimentel, AM, Alcoba, DD, et al. Mechanical processing of hyperviscous semen specimens can negatively affect sperm DNA fragmentation. Int Urol Nephrol 2014;46:737–42.Google Scholar
Nosi, E, Gritzapis, AD, Makarounis, K, et al. Improvement of sperm quality in hyperviscous semen following DNase I treatment. Int J Endocrinol 2019;2019:6325169.Google Scholar
Nikkanen, V. The effects of vasectomy on viscosity, pH and volume of semen in man. Andrologia 1979;11:123–5.Google Scholar
Welliver, C, Benson, AD, Frederick, L, et al. Analysis of semen parameters during 2 weeks of daily ejaculation: a first in humans study. Transl Androl Urol 2016;5:749–55.Google Scholar
Rui, H, Gerhardt, P, Mevag, B, Thomassen, Y, Purvis, K. Seminal plasma characteristics during frequent ejaculation. Int J Androl 1984;7:119–28.Google Scholar
Sigman, M, Boyle, K, Jarow, JP. Prevalence of sperm in the post-ejaculatory urine of fertile and subfertile men. Urology 2008;71:110–12.Google Scholar
Tash, JA, McGovern, JH, Schlegel, PN. Acquired hypogonadotropic hypogonadism presenting as decreased seminal volume. Urology 2000;56:669.Google Scholar
Tulandi, T, McInnes, RA. Induction of fertility in a man with hypogonadotropic hypogonadism with very low seminal volume. Fertil Steril 1986;46:730–3.Google Scholar
Paduch, DA, Polzer, PK, Ni, X, Basaria, S. Testosterone replacement in androgen-deficient men with ejaculatory dysfunction: a randomized controlled trial. J Clin Endocrinol Metab 2015;100:2956–62.Google Scholar
Amory, JK, Wang, C, Swerdloff, RS, et al. The effect of 5 alpha-reductase inhibition with dutasteride and finasteride on semen parameters and serum hormones in healthy men. J Clin Endocrinol Metab 2007;92:1659–65.Google Scholar
Overstreet, JW, Fuh, VL, Gould, J, et al. Chronic treatment with finasteride daily does not affect spermatogenesis or semen production in young men. J Urol 1999;162:1295–300.Google Scholar
Schlegel, PN, Sigman, M, Collura, B, et al. Diagnosis and treatment of infertility in men: AUA/ASRM Guideline Part I. J Urol 2021;205:3643.Google Scholar
Weiske, WH, Salzler, N, Schroeder-Printzen, I, Weidner, W. Clinical findings in congenital absence of the vasa deferentia. Andrologia 2000;32:1318.Google Scholar
Fox, CA, Meldrum, SJ, Watson, BW. Continuous measurement by radio-telemetry of vaginal pH during human coitus. J Reprod Fertil 1973;33:6975.Google Scholar
Harraway, C, Berger, NG, Dubin, NH. Semen pH in patients with normal versus abnormal sperm characteristics. Am J Obstet Gynecol 2000;182:1045–7.Google Scholar
Haugen, TB, Grotmol, T. pH of human semen. Int J Androl 1998;21:105–8.Google Scholar
Meares, EM Jr. Prostatitis syndromes: new perspectives about old woes. J Urol 1980;123:141–7.Google Scholar
Valsa, J, Skandhan, KP, Khan, PS, Sumangala, B, Gondalia, M. Split ejaculation study: semen parameters and calcium and magnesium in seminal plasma. Cent European J Urol 2012;65:216–18.Google Scholar
Van Assche, E, Bonduelle, M, Tournaye, H, et al. Cytogenetics of infertile men. Hum Reprod 1996;11 Suppl 4:124; discussion 5–6.Google Scholar
Cruger, DG, Agerholm, I, Byriel, L, Fedder, J, Bruun-Petersen, G. Genetic analysis of males from intracytoplasmic sperm injection couples. Clin Genet 2003;64:198203.CrossRefGoogle ScholarPubMed
Foresta, C, Garolla, A, Bartoloni, L, Bettella, A, Ferlin, A. Genetic abnormalities among severely oligospermic men who are candidates for intracytoplasmic sperm injection. J Clin Endocrinol Metab 2005;90:152–6.Google Scholar
Mascarenhas, M, Thomas, S, Kamath, MS, et al. Prevalence of chromosomal abnormalities and Y chromosome microdeletion among men with severe semen abnormalities and its correlation with successful sperm retrieval. J Hum Reprod Sci 2016;9:187–93.Google ScholarPubMed
Jaffe, TM, Kim, ED, Hoekstra, TH, Lipshultz, LI. Sperm pellet analysis: a technique to detect the presence of sperm in men considered to have azoospermia by routine semen analysis. J Urol 1998;159:1548–50.Google Scholar
Ron-El, R, Strassburger, D, Friedler, S, et al. Extended sperm preparation: an alternative to testicular sperm extraction in non-obstructive azoospermia. Hum Reprod 1997;12:1222–6.Google Scholar
Corea, M, Campagnone, J, Sigman, M. The diagnosis of azoospermia depends on the force of centrifugation. Fertil Steril 2005;83:920–2.Google Scholar
Schoor, RA, Elhanbly, S, Niederberger, CS, Ross, LS. The role of testicular biopsy in the modern management of male infertility. J Urol 2002;167:197200.CrossRefGoogle ScholarPubMed
Salonia, A, Bettochi, C, Carvalho, J, et al. EAU Guidelines on Sexual and Reproductive Health 2020. European Association of Urology Guidelines 2020 Edition. Presented at the European Association of Urology Annual Congress Amsterdam 2020. Arnhem: European Association of Urology Guidelines Office, 2020.Google Scholar
Bonde, JP, Ernst, E, Jensen, TK, et al. Relation between semen quality and fertility: a population-based study of 430 first-pregnancy planners. Lancet 1998;352:1172–7.Google Scholar
Slama, R, Eustache, F, Ducot, B, et al. Time to pregnancy and semen parameters: a cross-sectional study among fertile couples from four European cities. Hum Reprod 2002;17:503–15.Google Scholar
Zhang, MH, Shi, ZD, Yu, JC, Zhang, YP, Wang, LG, Qiu, Y. Scrotal heat stress causes sperm chromatin damage and cysteinyl aspartate-spicific proteinases 3 changes in fertile men. J Assist Reprod Genet 2015;32:747–55.Google Scholar
Jones, R, Mann, T. Toxicity of exogenous fatty acid peroxides towards spermatozoa. J Reprod Fertil 1977;50:255–60.Google Scholar
Asafu-Adjei, D, Judge, C, Deibert, CM, Li, G, Stember, D, Stahl, PJ. Systematic Review of the impact of varicocele grade on response to surgical management. J Urol 2020;203:4856.CrossRefGoogle ScholarPubMed
Redmon, JB, Drobnis, EZ, Sparks, A, Wang, C, Swan, SH. Semen and reproductive hormone parameters in fertile men with and without varicocele. Andrologia 2019;51:e13407.CrossRefGoogle ScholarPubMed
Damsgaard, J, Joensen, UN, Carlsen, E, et al. Varicocele is associated with impaired semen quality and reproductive hormone levels: a study of 7035 healthy young men from six European countries. Eur Urol 2016;70:1019–29.Google Scholar
Mekhaimar, A, Goble, M, Brunckhorst, O, et al. A systematic review of transurethral resection of ejaculatory ducts for the management of ejaculatory duct obstruction. Turk J Urol 2020;46:335–47.Google Scholar
von Zumbusch, A, Fiedler, K, Mayerhofer, A, Jessberger, B, Ring, J, Vogt, HJ. Birth of healthy children after intracytoplasmic sperm injection in two couples with male Kartagener’s syndrome. Fertil Steril 1998;70:643–6.Google Scholar
Gatimel, N, Moreau, J, Parinaud, J, Leandri, RD. Sperm morphology: assessment, pathophysiology, clinical relevance, and state of the art in 2017. Andrology 2017;5:845–62.Google Scholar
Kruger, TF, Menkveld, R, Stander, FS, et al. Sperm morphologic features as a prognostic factor in in vitro fertilization. Fertil Steril 1986;46:1118–23.Google Scholar
Kruger, TF, Acosta, AA, Simmons, KF, Swanson, RJ, Matta, JF, Oehninger, S. Predictive value of abnormal sperm morphology in in vitro fertilization. Fertil Steril 1988;49:112–17.Google Scholar
Kovac, JR, Smith, RP, Cajipe, M, Lamb, DJ, Lipshultz, LI. Men with a complete absence of normal sperm morphology exhibit high rates of success without assisted reproduction. Asian J Androl 2017;19:3942.Google ScholarPubMed
Hotaling, JM, Smith, JF, Rosen, M, Muller, CH, Walsh, TJ. The relationship between isolated teratozoospermia and clinical pregnancy after in vitro fertilization with or without intracytoplasmic sperm injection: a systematic review and meta-analysis. Fertil Steril 2011;95:1141–5.Google Scholar
Kohn, TP, Kohn, JR, Ramasamy, R. Effect of sperm morphology on pregnancy success via intrauterine insemination: a systematic review and meta-analysis. J Urol 2018;199:812–22.CrossRefGoogle ScholarPubMed
Fan, W, Li, SW, Li, L, et al. Outcome of conventional IVF and ICSI on sibling oocytes in the case of isolated teratozoospermia. J Assist Reprod Genet 2012;29:905–10.Google Scholar
Chansel-Debordeaux, L, Dandieu, S, Bechoua, S, Jimenez, C. Reproductive outcome in globozoospermic men: update and prospects. Andrology 2015;3:1022–34.CrossRefGoogle ScholarPubMed
Perrin, A, Morel, F, Moy, L, Colleu, D, Amice, V, De Braekeleer, M. Study of aneuploidy in large-headed, multiple-tailed spermatozoa: case report and review of the literature. Fertil Steril 2008;90:1201 e13–17.Google Scholar
Belker, AM, Sherins, RJ, Dennison-Lagos, L, Thorsell, LP, Schulman, JD. Percutaneous testicular sperm aspiration: a convenient and effective office procedure to retrieve sperm for in vitro fertilization with intracytoplasmic sperm injection. J Urol 1998;160(6 Pt 1):2058–62.Google Scholar
Johanisson, E, Campana, A, Luthi, R, de Agostini, A. Evaluation of ‘round cells’ in semen analysis: a comparative study. Hum Reprod Update 2000;6:404–12.Google Scholar
Brunner, RJ, Demeter, JH, Sindhwani, P. Review of guidelines for the evaluation and treatment of leukocytospermia in male infertility. World J Mens Health 2019;37:128–37.Google Scholar
Mazumdar, S, Levine, AS. Antisperm antibodies: etiology, pathogenesis, diagnosis, and treatment. Fertil Steril 1998;70:799810.CrossRefGoogle ScholarPubMed
Zini, A, Fahmy, N, Belzile, E, Ciampi, A, Al-Hathal, N, Kotb, A. Antisperm antibodies are not associated with pregnancy rates after IVF and ICSI: systematic review and meta-analysis. Hum Reprod 2011;26:1288–95.Google Scholar
Smith, TB, Dun, MD, Smith, ND, Curry, BJ, Connaughton, HS, Aitken, RJ. The presence of a truncated base excision repair pathway in human spermatozoa that is mediated by OGG1. J Cell Sci 2013;126(Pt 6):1488–97.Google Scholar
Garcia-Rodriguez, A, Gosalvez, J, Agarwal, A, Roy, R, Johnston, S. DNA damage and repair in human reproductive cells. Int J Mol Sci 2018;20:31.CrossRefGoogle ScholarPubMed
Colasante, A, Minasi, MG, Scarselli, F, et al. The aging male: relationship between male age, sperm quality and sperm DNA damage in an unselected population of 3124 men attending the fertility centre for the first time. Arch Ital Urol Androl 2019;90:254–9.Google Scholar
Agarwal, A, Gupta, S, Du Plessis, S, et al. Abstinence time and its impact on basic and advanced semen parameters. Urology 2016;94:102–10.Google Scholar
Moskovtsev, SI, Jarvi, K, Mullen, JB, Cadesky, KI, Hannam, T, Lo, KC. Testicular spermatozoa have statistically significantly lower DNA damage compared with ejaculated spermatozoa in patients with unsuccessful oral antioxidant treatment. Fertil Steril 2010;93:1142–6.Google Scholar
Tan, J, Taskin, O, Albert, A, Bedaiwy, MA. Association between sperm DNA fragmentation and idiopathic recurrent pregnancy loss: a systematic review and meta-analysis. Reprod Biomed Online 2019;38:951–60.Google Scholar
Ahmadi, A, Ng, SC. Fertilizing ability of DNA-damaged spermatozoa. J Exp Zool 1999;284:696704.Google Scholar
Genesca, A, Caballin, MR, Miro, R, Benet, J, Germa, JR, Egozcue, J. Repair of human sperm chromosome aberrations in the hamster egg. Hum Genet 1992;89:181–6.Google Scholar
Ribas-Maynou, J, Benet, J. Single and double strand sperm DNA damage: different reproductive effects on male fertility. Genes (Basel) 2019;10:105.Google Scholar
Aitken, RJ. Reactive oxygen species as mediators of sperm capacitation and pathological damage. Mol Reprod Dev 2017;84:1039–52.Google Scholar
Koppers, AJ, De Iuliis, GN, Finnie, JM, McLaughlin, EA, Aitken, RJ. Significance of mitochondrial reactive oxygen species in the generation of oxidative stress in spermatozoa. J Clin Endocrinol Metab 2008;93:3199–207.Google Scholar
Aitken, RJ, West, KM. Analysis of the relationship between reactive oxygen species production and leucocyte infiltration in fractions of human semen separated on Percoll gradients. Int J Androl 1990;13:433–51.Google Scholar
Aitken, RJ, Clarkson, JS. Cellular basis of defective sperm function and its association with the genesis of reactive oxygen species by human spermatozoa. J Reprod Fertil 1987;81:459–69.Google Scholar
Sigman, M, Jarow, JP. Endocrine evaluation of infertile men. Urology 1997;50:659–64.Google Scholar
Esteves, SC, Miyaoka, R, Agarwal, A. An update on the clinical assessment of the infertile male. [corrected]. Clinics (Sao Paulo) 2011;66:691700.Google Scholar
Ventimiglia, E, Capogrosso, P, Boeri, L, et al. Validation of the American Society for Reproductive Medicine guidelines/recommendations in white European men presenting for couple’s infertility. Fertil Steril 2016;106:107682 e1.Google Scholar
Trussell, JC, Coward, RM, Santoro, N, et al. Association between testosterone, semen parameters, and live birth in men with unexplained infertility in an intrauterine insemination population. Fertil Steril 2019;111:1129–34.Google Scholar
Jarow, JP, Chen, H, Rosner, TW, Trentacoste, S, Zirkin, BR. Assessment of the androgen environment within the human testis: minimally invasive method to obtain intratesticular fluid. J Androl 2001;22:640–5.Google Scholar
McLachlan, RI, O’Donnell, L, Stanton, PG, et al. Effects of testosterone plus medroxyprogesterone acetate on semen quality, reproductive hormones, and germ cell populations in normal young men. J Clin Endocrinol Metab 2002;87:546–56.Google Scholar
Coviello, AD, Bremner, WJ, Matsumoto, AM, et al. Intratesticular testosterone concentrations comparable with serum levels are not sufficient to maintain normal sperm production in men receiving a hormonal contraceptive regimen. J Androl 2004;25:931–8.Google Scholar
Lazarou, S, Reyes-Vallejo, L, Morgentaler, A. Wide variability in laboratory reference values for serum testosterone. J Sex Med 2006;3:1085–9.Google Scholar
Piro, C, Fraioli, F, Sciarra, F, Conti, C. Circadian rhythm of plasma testosterone, cortisol and gonadotropins in normal male subjects. J Steroid Biochem 1973;4:321–9.Google Scholar
Gagliano-Juca, T, Li, Z, Pencina, KM, et al. Oral glucose load and mixed meal feeding lowers testosterone levels in healthy eugonadal men. Endocrine 2019;63:149–56.Google Scholar
Brambilla, DJ, O’Donnell, AB, Matsumoto, AM, McKinlay, JB. Intraindividual variation in levels of serum testosterone and other reproductive and adrenal hormones in men. Clin Endocrinol (Oxf) 2007;67:853–62.Google Scholar
Morley, JE, Patrick, P, Perry, HM 3rd. Evaluation of assays available to measure free testosterone. Metabolism 2002;51:554–9.Google Scholar
Gordetsky, J, van Wijngaarden, E, O’Brien, J. Redefining abnormal follicle-stimulating hormone in the male infertility population. BJU Int 2012;110:568–72.Google Scholar
Martin-du-Pan, RC, Bischof, P. Increased follicle stimulating hormone in infertile men. Is increased plasma FSH always due to damaged germinal epithelium? Hum Reprod 1995;10:1940–5.Google Scholar
Kumanov, P, Nandipati, K, Tomova, A, Agarwal, A. Inhibin B is a better marker of spermatogenesis than other hormones in the evaluation of male factor infertility. Fertil Steril 2006;86:332–8.Google Scholar
Brodie, A, Inkster, S, Yue, W. Aromatase expression in the human male. Mol Cell Endocrinol 2001;178(1–2):23–8.Google Scholar
Robertson, KM, O’Donnell, L, Jones, ME, et al. Impairment of spermatogenesis in mice lacking a functional aromatase (cyp 19) gene. Proc Natl Acad Sci U S A 1999;96:7986–91.Google Scholar
Haverfield, JT, Ham, S, Brown, KA, Simpson, ER, Meachem, SJ. Teasing out the role of aromatase in the healthy and diseased testis. Spermatogenesis 2011;1:240–9.Google Scholar
Santen, RJ. Feedback control of luteinizing hormone and follicle-stimulating hormone secretion by testosterone and estradiol in men: physiological and clinical implications. Clin Biochem 1981;14:243–51.Google Scholar
Pavlovich, CP, King, P, Goldstein, M, Schlegel, PN. Evidence of a treatable endocrinopathy in infertile men. J Urol 2001;165:837–41.Google Scholar
Salama, N, Blgozah, S. Serum estradiol levels in infertile men with non-obstructive azoospermia. Ther Adv Reprod Health 2020;14:2633494120928342.Google Scholar
Del Giudice, F, Busetto, GM, De Berardinis, E, et al. A systematic review and meta-analysis of clinical trials implementing aromatase inhibitors to treat male infertility. Asian J Androl 2020;22:360–7.Google Scholar
Johnson, MD. Genetic risks of intracytoplasmic sperm injection in the treatment of male infertility: recommendations for genetic counseling and screening. Fertil Steril 1998;70:397411.Google Scholar
Vincent, MC, Daudin, M, De, MP, et al. Cytogenetic investigations of infertile men with low sperm counts: a 25-year experience. J Androl 2002;23:1822; discussion 44–5.Google Scholar
Ventimiglia, E, Capogrosso, P, Boeri, L, et al. When to perform karyotype analysis in infertile men? Validation of the European Association of Urology Guidelines with the proposal of a new predictive model. Eur Urol 2016;70:920–3.Google Scholar
Olesen, IA, Andersson, AM, Aksglaede, L, et al. Clinical, genetic, biochemical, and testicular biopsy findings among 1,213 men evaluated for infertility. Fertil Steril 2017;107:7482 e7.Google Scholar
Krausz, C, Casamonti, E. Spermatogenic failure and the Y chromosome. Hum Genet 2017;136:637–55.Google Scholar
Colaco, S, Modi, D. Genetics of the human Y chromosome and its association with male infertility. Reprod Biol Endocrinol 2018;16:14.Google Scholar
Yuen, RK, Merkoulovitch, A, MacDonald, JR, et al. Development of a high-resolution Y-chromosome microarray for improved male infertility diagnosis. Fertil Steril 2014;101:107985 e3.Google Scholar
Hopps, CV, Mielnik, A, Goldstein, M, Palermo, GD, Rosenwaks, Z, Schlegel, PN. Detection of sperm in men with Y chromosome microdeletions of the AZFa, AZFb and AZFc regions. Hum Reprod 2003;18:1660–5.Google Scholar
Kleiman, SE, Almog, R, Yogev, L, et al. Screening for partial AZFa microdeletions in the Y chromosome of infertile men: is it of clinical relevance? Fertil Steril 2012;98:43–7.Google Scholar
Brandell, RA, Mielnik, A, Liotta, D, et al. AZFb deletions predict the absence of spermatozoa with testicular sperm extraction: preliminary report of a prognostic genetic test. Hum Reprod 1998;13:2812–15.Google Scholar
Thornhill, AR, Guenther, AJ, Barbarotto, GM, et al. False-positive Y-microdeletion result for a fertile male caused by an alteration under a PCR primer. Int J Androl 2002;25:352–7.Google Scholar
Wu, Q, Chen, GW, Yan, TF, et al. Prevalent false positives of azoospermia factor a (AZFa) microdeletions caused by single-nucleotide polymorphism rs72609647 in the sY84 screening of male infertility. Asian J Androl 2011;13:877–80.Google Scholar
Rolf, C, Gromoll, J, Simoni, M, Nieschlag, E. Natural transmission of a partial AZFb deletion of the Y chromosome over three generations: case report. Hum Reprod 2002;17:2267–71.Google Scholar
Samli, H, Murat Samli, M, Solak, M. Natural transmission of AZFb Y-chromosomal microdeletion from father to his three sons. Arch Androl 2006;52:423–6.Google Scholar
Mirfakhraie, R, Mirzajani, F, Kalantar, SM, et al. High prevalence of AZFb microdeletion in Iranian patients with idiopathic non-obstructive azoospermia. Indian J Med Res 2010;132:265–70.Google Scholar
Kohn, TP, Kohn, JR, Owen, RC, Coward, RM. The prevalence of Y-chromosome microdeletions in oligozoospermic men: a systematic review and meta-analysis of European and North American studies. Eur Urol 2019;76:626–36.Google Scholar
Johnson, M, Raheem, A, De Luca, F, et al. An analysis of the frequency of Y-chromosome microdeletions and the determination of a threshold sperm concentration for genetic testing in infertile men. BJU Int 2019;123:367–72.Google Scholar
Rozen, SG, Marszalek, JD, Irenze, K, et al. AZFc deletions and spermatogenic failure: a population-based survey of 20,000 Y chromosomes. Am J Hum Genet 2012;91:890–6.Google Scholar
Mohammadpour Lashkari, F, Totonchi, M, Zamanian, MR, et al. 46,XX males: a case series based on clinical and genetics evaluation. Andrologia 2017;49(7).Google Scholar
Vorona, E, Zitzmann, M, Gromoll, J, Schuring, AN, Nieschlag, E. Clinical, endocrinological, and epigenetic features of the 46,XX male syndrome, compared with 47,XXY Klinefelter patients. J Clin Endocrinol Metab 2007;92:3458–65.Google Scholar
Anguiano, A, Oates, RD, Amos, JA, et al. Congenital bilateral absence of the vas deferens. A primarily genital form of cystic fibrosis. JAMA 1992;267:1794–7.Google Scholar
Yu, J, Chen, Z, Ni, Y, Li, Z. CFTR mutations in men with congenital bilateral absence of the vas deferens (CBAVD): a systemic review and meta-analysis. Hum Reprod 2012;27:2535.Google Scholar
Schwarzer, JU, Schwarz, M. Significance of CFTR gene mutations in patients with congenital aplasia of vas deferens with special regard to renal aplasia. Andrologia 2012;44:305–7.CrossRefGoogle ScholarPubMed
American College of Gynecology. ACOG Committee Opinion No. 762: prepregnancy counseling. Obstet Gynecol 2019;133:e78–89.Google Scholar
Cystic Fibrosis Mutation Database. CFMDB statistics. Available from: www.genet.sickkids.on.ca/StatisticsPage.html.Google Scholar
Lu, S, Cui, Y, Li, X, et al. Association of cystic fibrosis transmembrane-conductance regulator gene mutation with negative outcome of intracytoplasmic sperm injection pregnancy in cases of congenital bilateral absence of vas deferens. Fertil Steril 2014;101:1255–60.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
Pagin, A, Bergougnoux, A, Girodon, E, et al. Novel ADGRG2 truncating variants in patients with X-linked congenital absence of vas deferens. Andrology 2020;8:618–24.Google Scholar
Yang, B, Wang, J, Zhang, W, et al. Pathogenic role of ADGRG2 in CBAVD patients replicated in Chinese population. Andrology 2017;5:954–7.Google Scholar
Lee, CH, Wu, CC, Wu, YN, Chiang, HS. Gene copy number variations in Asian patients with congenital bilateral absence of the vas deferens. Hum Reprod 2009;24:748–55.Google Scholar
Wu, YN, Chen, KC, Wu, CC, Lin, YH, Chiang, HS. SLC9A3 affects vas deferens development and associates with Taiwanese congenital bilateral absence of the vas deferens. Biomed Res Int 2019;2019:3562719.Google Scholar
Thirumavalavan, N, Gabrielsen, JS, Lamb, DJ. Where are we going with gene screening for male infertility? Fertil Steril 2019;111:842–50.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
×