Skip to main content Accessibility help
×
Hostname: page-component-76fb5796d-x4r87 Total loading time: 0 Render date: 2024-04-28T07:02:30.129Z Has data issue: false hasContentIssue false

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
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

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.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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 Scholar
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.CrossRefGoogle ScholarPubMed
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.Google Scholar
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.Google Scholar
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 Scholar
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.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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.Google Scholar
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 Scholar
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.CrossRefGoogle ScholarPubMed
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.Google Scholar
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.Google Scholar
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.Google Scholar
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.CrossRefGoogle ScholarPubMed
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 Scholar
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.Google Scholar
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.Google Scholar
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.Google Scholar
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.Google Scholar
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.Google Scholar
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

References

World Health Organization. WHO Laboratory Manual for the Examination and Processing of Human Semen, 5th ed. Geneva: World Health Organization, 2010.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
Sokol, RZ, Sparkes, R. Demonstrated paternity in spite of severe idiopathic oligospermia. Fertil Steril 1987;47:356–8.Google Scholar
Silber, SJ. The relationship of abnormal semen parameters to male fertility. Hum Reprod 1989;4:947–53.Google Scholar
Puga Molina, LC, Luque, GM, Balestrini, PA, Marin-Briggiler, CI, Romarowski, A, Buffone, MG. Molecular sasis of human sperm capacitation. Front Cell Dev Biol 2018;6:72.Google Scholar
World Health Organization. WHO Laboratory Manual for the Examination of Human Semen and Sperm-Cervical Mucus Interaction, 4th ed. Geneva: World Health Organization and Cambridge: Cambridge University Press 1999.Google Scholar
Muratori, M, Marchiani, S, Tamburrino, L, Baldi, E. Sperm DNA fragmentation: mechanisms of origin. Adv Exp Med Biol 2019;1166:7585.Google Scholar
Muratori, M, Marchiani, S, Tamburrino, L, et al. DNA fragmentation in brighter sperm predicts male fertility independently from age and semen parameters. Fertil Steril 2015;104:58290 e4.Google Scholar
Robinson, L, Gallos, ID, Conner, SJ, et al. The effect of sperm DNA fragmentation on miscarriage rates: a systematic review and meta-analysis. Hum Reprod 2012;27:2908–17.Google Scholar
Cissen, M, Wely, MV, Scholten, I, et al. Measuring sperm DNA fragmentation and clinical outcomes of medically assisted reproduction: a systematic review and meta-analysis. PLoS ONE 2016;11:e0165125.Google Scholar
Simon, L, Zini, A, Dyachenko, A, Ciampi, A, Carrell, DT. A systematic review and meta-analysis to determine the effect of sperm DNA damage on in vitro fertilization and intracytoplasmic sperm injection outcome. Asian J Androl 2017;19:8090.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
Evgeni, E, Charalabopoulos, K, Asimakopoulos, B. Human sperm DNA fragmentation and its correlation with conventional semen parameters. J Reprod Infertil 2014;15:214.Google Scholar
Wang, BN, Nie, B, Tang, D, et al. Analysis of meiotic segregation patterns and interchromosomal effects in sperm from 13 Robertsonian translocations. Balkan J Med Genet 2017;20:4350.Google Scholar
Robbins, DJ, Coleman, MS. Initiator role of double stranded DNA in terminal transferase catalyzed polymerization reactions. Nucleic Acids Res 1988;16:2943–57.Google Scholar
Fernandez, JL, Cajigal, D, Lopez-Fernandez, C, Gosalvez, J. Assessing sperm DNA fragmentation with the sperm chromatin dispersion test. Methods Mol Biol 2011;682:291301.Google Scholar
Fernandez, JL, Muriel, L, Goyanes, V, et al. Halosperm is an easy, available, and cost-effective alternative for determining sperm DNA fragmentation. Fertil Steril 2005;84:860.Google Scholar
Fernandez, JL, Muriel, L, Goyanes, V, et al. Simple determination of human sperm DNA fragmentation with an improved sperm chromatin dispersion test. Fertil Steril 2005;84:833–42.Google Scholar
Simon, L, Carrell, DT. Sperm DNA damage measured by comet assay. Methods Mol Biol 2013;927:137–46.Google Scholar
Collins, AR, Dobson, VL, Dusinska, M, Kennedy, G, Stetina, R. The comet assay: what can it really tell us? Mutat Res 1997;375:183–93.Google Scholar
Konca, K, Lankoff, A, Banasik, A, et al. A cross-platform public domain PC image-analysis program for the comet assay. Mutat Res 2003;534(1–2):1520.Google Scholar
Gonzalez, JE, Romero, I, Barquinero, JF, Garcia, O. Automatic analysis of silver-stained comets by CellProfiler software. Mutat Res 2012;748(1–2):60–4.Google Scholar
Evenson, DP. Sperm chromatin structure assay (SCSA(R)). Methods Mol Biol 2013;927:147–64.Google Scholar
Evenson, DP, Jost, LK, Marshall, D, et al. Utility of the sperm chromatin structure assay as a diagnostic and prognostic tool in the human fertility clinic. Hum Reprod 1999;14:1039–49.Google Scholar
Giwercman, A, Lindstedt, L, Larsson, M, et al. Sperm chromatin structure assay as an independent predictor of fertility in vivo: a case-control study. Int J Androl 2010;33:e221–7.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
Gekas, J, Thepot, F, Turleau, C, et al. Chromosomal factors of infertility in candidate couples for ICSI: an equal risk of constitutional aberrations in women and men. Hum Reprod 2001;16:8290.Google Scholar
Kurinczuk, JJ, Bhattacharya, S. Assisted reproductive technologies: impact on fetal and neonatal outcomes. Semin Fetal Neonatal Med 2014;19:221.Google Scholar
Godo, A, Blanco, J, Vidal, F, Anton, E. Accumulation of numerical and structural chromosome imbalances in spermatozoa from reciprocal translocation carriers. Hum Reprod 2013;28:840–9.Google Scholar
Godo, A, Blanco, J, Vidal, F, Sandalinas, M, Garcia-Guixe, E, Anton, E. Altered segregation pattern and numerical chromosome abnormalities interrelate in spermatozoa from Robertsonian translocation carriers. Reprod Biomed Online 2015;31:7988.Google Scholar
Morel, F, Douet-Guilbert, N, Le Bris, MJ, et al. Meiotic segregation of translocations during male gametogenesis. Int J Androl 2004;27:200–12.Google Scholar
Zhao, WW, Wu, M, Chen, F, et al. Robertsonian translocations: an overview of 872 Robertsonian translocations identified in a diagnostic laboratory in China. PLoS ONE 2015;10:e0122647.Google Scholar
Yatsenko, AN, Yatsenko, SA, Weedin, JW, et al. Comprehensive 5-year study of cytogenetic aberrations in 668 infertile men. J Urol 2010;183:1636–42.Google Scholar
Schlegel, PN, Sigman, M, Collura, B, et al. Diagnosis and treatment of infertility in men: AUA/ASRM guideline part I. Fertil Steril 2021;115:5461.Google Scholar
Schlegel, PN, Sigman, M, Collura, B, et al. Diagnosis and treatment of infertility in men: AUA/ASRM guideline part II. Fertil Steril 2021;115:62–9.Google Scholar
Schlegel, PN, Sigman, M, Collura, B, et al. Diagnosis and treatment of infertility in men: AUA/ASRM Guideline PART II. J Urol 2021;205:4451.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
Tiepolo, L, Zuffardi, O. Localization of factors controlling spermatogenesis in the nonfluorescent portion of the human Y chromosome long arm. Hum Genet 1976;34:119–24.Google Scholar
Saxena, R, Brown, LG, Hawkins, T, et al. The DAZ gene cluster on the human Y chromosome arose from an autosomal gene that was transposed, repeatedly amplified and pruned. Nat Genet 1996;14:292–9.Google Scholar
Reijo, R, Alagappan, RK, Patrizio, P, Page, DC. Severe oligozoospermia resulting from deletions of azoospermia factor gene on Y chromosome. Lancet 1996;347:1290–3.Google Scholar
Jorgez, CJ, Weedin, JW, Sahin, A, et al. Aberrations in pseudoautosomal regions (PARs) found in infertile men with Y-chromosome microdeletions. J Clin Endocrinol Metab 2011;96:E674–9.Google Scholar
Medina-Martinez, O, Haller, M, Rosenfeld, JA, O’Neill, MA, Lamb, DJ, Jamrich, M. The transcription factor Maz is essential for normal eye development. Dis Model Mech 2020;13:dmm044412.Google Scholar
Haller, M, Au, J, O’Neill, M, Lamb, DJ. 16p11.2 transcription factor MAZ is a dosage-sensitive regulator of genitourinary development. Proc Natl Acad Sci U S A 2018;115:E1849–58.Google Scholar
Haller, M, Mo, Q, Imamoto, A, Lamb, DJ. Murine model indicates 22q11.2 signaling adaptor CRKL is a dosage-sensitive regulator of genitourinary development. Proc Natl Acad Sci U S A 2017;114:4981–6.Google Scholar
Tannour-Louet, M, Han, S, Louet, JF, et al. Increased gene copy number of VAMP7 disrupts human male urogenital development through altered estrogen action. Nat Med 2014;20:715–24.Google Scholar
Yatsenko, AN, Georgiadis, AP, Ropke, A, et al. X-linked TEX11 mutations, meiotic arrest, and azoospermia in infertile men. N Engl J Med 2015;372:2097–107.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
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
Coutton, C, Escoffier, J, Martinez, G, Arnoult, C, Ray, PF. Teratozoospermia: spotlight on the main genetic actors in the human. Hum Reprod Update 2015;21:455–85.Google Scholar
Wang, WL, Tu, CF, Tan, YQ. Insight on multiple morphological abnormalities of sperm flagella in male infertility: what is new? Asian J Androl 2020;22:236–45.Google Scholar
Cooper, TG, Noonan, E, von Eckardstein, S, et al. World Health Organization reference values for human semen characteristics. Hum Reprod Update 2010;16:231–45.Google Scholar
Enciso, M, Alfarawati, S, Wells, D. Increased numbers of DNA-damaged spermatozoa in samples presenting an elevated rate of numerical chromosome abnormalities. Hum Reprod 2013;28:1707–15.Google Scholar
Ramasamy, R, Scovell, JM, Kovac, JR, Cook, PJ, Lamb, DJ, Lipshultz, LI. Fluorescence in situ hybridization detects increased sperm aneuploidy in men with recurrent pregnancy loss. Fertil Steril 2015;103:9069 e1.Google Scholar
Ramasamy, R, Besada, S, Lamb, DJ. Fluorescent in situ hybridization of human sperm: diagnostics, indications, and therapeutic implications. Fertil Steril 2014;102:1534–9.Google Scholar

References

MacLeod, J. The male factor in fertility and infertility; an analysis of ejaculate volume in 800 fertile men and in 600 men in infertile marriage. Fertil Steril 1950;1:347–61.Google Scholar
MacLeod, J, Gold, RZ. The male factor in fertility and infertility. II. Spermatozoon counts in 1000 men of known fertility and in 1000 cases of infertile marriage. J Urol 1951;66:436–49.Google Scholar
MacLeod, J, Gold, RZ. The male factor in fertility and infertility. III. An analysis of motile activity in the spermatozoa of 1000 fertile men and 1000 men in infertile marriage. Fertil Steril 1951;2:187204.Google Scholar
MacLeod, J, Gold, RZ. The male factor in fertility and infertility. IV. Sperm morphology in fertile and infertile marriage. Fertil Steril 1951;2:394414.Google Scholar
World Health Organization. WHO Laboratory Manual for the Examination of Human Semen and Sperm-Cervical Mucus Interaction, 1st ed. Singapore: Press Concern, 1980.Google Scholar
World Health Organization. WHO Laboratory Manual for the Examination of Human Semen and Sperm-Cervical Mucus Interaction. 2nd ed. Cambridge: Cambridge University Press, 1987.Google Scholar
World Health Organization. WHO Laboratory Manual for the Examination of Human Semen and Sperm-Cervical Mucus Interaction, 3rd ed. Cambridge: Cambridge University Press, 1992.Google Scholar
World Health Organization. WHO Laboratory Manual for the Examination of Human Semen and Sperm-Cervical Mucus Interaction, 4th ed. Cambridge: Cambridge University Press, 1999.Google Scholar
World Health Organization. WHO Laboratory Manual for the Examination and Processing of Human Semen, 5th ed. Geneva: World Health Organization, 2010.Google Scholar
Amann, RP. Can the fertility potential of a seminal sample be predicted accurately? J Androl 1989;10:89.Google Scholar
Smith, KD, Rodriguez-Rigau, LJ, Steinberger, E. Relation between indices of semen analysis and pregnancy rate in infertile couples. Fertil Steril 1977;28:1314–19.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
Niederberger, CS. Semen and the curse of cutoffs. J Urol 2011;185:381–2.Google Scholar
Patel, AS, Leong, JY, Ramasamy, R. Prediction of male infertility by the World Health Organization laboratory manual for assessment of semen analysis: a systematic review. Arab J Urol 2018;16:96102.Google Scholar
Blasco, L. Clinical tests of sperm fertilizing ability. Fertil Steril 1984;41:177–92.Google Scholar
Liu, DY, Baker, HWG. Tests of human sperm function and fertilization in vitro. Fertil Steril 1992;58:465–83.Google Scholar
De Jonge, C. Attributes of fertile spermatozoa: an update. J Androl 1999;20:463–73.Google Scholar
Muller, CH. Rationale, interpretation, validation, and uses of sperm function tests. J Androl 2000;21:1030.Google Scholar
Aitken, RJ. Sperm function tests and fertility. Int J Andrology 2006;29:6975.Google Scholar
Lamb, DJ. Semen analysis in 21st century medicine: the need for sperm function testing. Asian J Androl 2010;12:6470.Google Scholar
De Jonge, C. Semen analysis: looking for an upgrade in class. Fertil Steril 2012;97:260–6.Google Scholar
Oehninger, S, Franken, DR, Ombelet, W. Sperm functional tests. Fertil Steril 2014;102:1528–33.Google Scholar
Mortimer, D. The functional anatomy of the human spermatozoon: relating ultrastructure and function. Mol Human Reprod 2018;24:567–92.Google Scholar
Kricka, LJ, Wilding, P. Evolution of sperm testing – a bright future? Clin Chem 2019;65:212–14.Google Scholar
Mortimer, ST. A critical review of the physiological importance and analysis of sperm movement in mammals. Hum Reprod Update 1997;3:403–39.Google Scholar
De Jonge, C. Biological basis for human capacitation. Hum Reprod Update 2005;11:205–14.Google Scholar
Austin, CR. Observations on the penetration of sperm into the mammalian egg. Aust J Sci Res 1951;4(series B):581–96.Google Scholar
Austin, CR. The “capacitation” of mammalian sperm. Nature 1952;170:326.Google Scholar
Chang, MC. Fertilizing capacity of spermatozoa deposited into the fallopian tubes. Nature 1951;168:697–8.Google Scholar
De Jonge, C. Biological basis for human capacitation – revisted. Hum Reprod Update 2017;23:289–99.Google Scholar
Gervasi, MG, Visconti, PE. Chang’s meaning of capacitation: a molecular perspective. Mol Reprod Dev 2016;83:860–74.Google Scholar
Jin, SK, Yand, WX. Factors and pathways involved in capacitation: how are they regulated? Oncotarget 2017;8:3600–27.Google Scholar
Puga Molina, LC, Luque, GM, Balestrini, PA, Marín-Briggiler, CI, Romarowski, A, Buffone, MG. Molecular basis of human sperm capacitation. Front Cell Dev Biol 2018;6:72.Google Scholar
Aitken, RJ. Reactive oxygen species as mediators of sperm capacitation and pathological damage. Mol Reprod Dev 2017;84:1039–52.Google Scholar
Henkel, R, Samanta, L, Agarwal, A (eds). Oxidants, Antioxidants, and Impact of the Oxidative Status in Male Reproduction. London: Elsevier, 2019.Google Scholar
Sobrero, AJ, McLeod, J. The immediate postcoital test. Fertil Steril 1962;13:184–9.Google Scholar
Rubenstein, BB, Strauss, H, Lazarus, ML, Hankin, H. Sperm survival in women: motile sperm in fundus and tubes of surgical cases. Fertil Steril 1951;2:1519.Google Scholar
Settlage, DS, Motoshima, M, Tredway, DR. Sperm transport from the external cervical os to the fallopian tubes in women: a time and quantitation study. Fertil Steril 1973;24:655–61.Google Scholar
Ahlgren, M. Sperm transport to and survival in the human fallopian tube. Gynecol Invest 1975;6:206–14.Google Scholar
Yanagimachi, R. The movement of golden hamster spermatozoa before and after capacitation. J Reprod Fertil 1970;23:193–6.Google Scholar
Suarez, SS. Control of hyperactivation in sperm. Hum Reprod Update 2008;14:647–57.Google Scholar
Moghissi, KS. The function of the cervix in fertility. Fertil Steril 1972;23:295306.Google Scholar
Oei, SG. The postcoital test: a controversial investigation. Eur J Obstet Gynecol 1998;77:123–4.Google Scholar
Ola, B, Afnan, M, Papaioannou, S, Sharif, K, Bjorndahl, L, Coomarasamy, A. Accuracy of sperm-cervical mucus penetration tests in evaluating sperm motility in semen: a systematic quantitative review. Hum Reprod 2003;18:1037–46.Google Scholar
Kesseru, E. In vivo sperm penetration and in vitro sperm migration tests. Fertil Steril 1973;24:584–91.Google Scholar
Alexander, NJ. Evaluation of male infertility with an in vitro cervical mucus penetration test. Fertil Steril 1981;36:201–8.Google Scholar
Mortimer, D, Mortimer, ST, Shu, MA, Swart, R. A simplified approach to sperm-cervical mucus interaction testing using a hyaluronate migration test. Hum Reprod 1990;5:835–41.Google Scholar
Ivic, A, Onyeaka, H, Girling, A, et al. Critical evaluation of methylcellulose as an alternative medium in sperm migration tests. Hum Reprod 2002;17:143–9.Google Scholar
David, MP, Amit, A, Bergman, A, Yedwab, G, Paz, GF, Hommonai, ZT. Sperm penetration in vitro: correlations between parameters of sperm quality and the penetration capacity. Fertil Steril 1979;32:676–80.Google Scholar
Katz, DF, Overstreet, JW, Hanson, FW. A new quantitative test for sperm penetration into cervical mucus. Fertil Steril 1980;33:179–86.Google Scholar
Overstreet, JW, Coats, C, Katz, DF, Hanson, FW. The importance of seminal plasma for sperm penetration of cervical mucus. Fertil Steril 1980;34:569–72.Google Scholar
Makler, A. A new multiple exposure photography method for objective human spermatozoal motility determination. Fertil Steril 1978;30:192–9.Google Scholar
Overstreet, JW, Katz, DF, Hanson, FW, Fonseca, JR. A simple inexpensive method for objective assessment of human sperm movement characteristics. Fertil Steril 1979;31:162–72.Google Scholar
Katz, DF, Overstreet, JW, Hanson, FW. Variations within and amongst normal men of movement characteristics of seminal spermatozoa. J Reprod Fertil 1981;62:221–8.Google Scholar
Aitken, RJ, Best, FSM, Richardson, DW, et al. An analysis of sperm function in cases of unexplained infertility: conventional criteria, movement characteristics, and fertilizing capacity. Fertil Steril 1982;38:212–21.Google Scholar
Katz, DF, Overstreet, JW. Sperm motility assessment by videomicrography. Fertil Steril 1981;35:188–93.Google Scholar
Mathur, S, Carlton, M, Ziegler, J, Rust, PF, Williamson, HO. A computerized sperm motion analysis. Fertil Steril 1986;46:484–8.Google Scholar
Holt, WV, Moore, HDM, Hillier, SG. Computer-assisted measurement of sperm swimming speed in human semen: correlation of results with in vitro fertilization assays. Fertil Steril 1985;44:112–19.Google Scholar
Barratt, CLR, Tomlinson, MJ, Cooke, ID. Prognostic significance of computerized motility analysis for in vivo fertility. Fertil Steril 1993;60:520–5.Google Scholar
Irvine, DS, Macleod, IC, Templeton, AA, Masterton, A, Taylor, A. A prospective clinical study of the relationship between the computer-assisted assessment of human semen quality and the achievement of pregnancy in vivo. Hum Reprod 1994;9:2324–34.Google Scholar
Macleod, IC, Irvine, DS. The predictive value of computerized-assisted semen analysis in the context of a donor insemination programme. Hum Reprod 1995;10:580–6.Google Scholar
Larsen, L, Scheike, T, Jensen, TK, et al.; The Danish First Pregnancy Planner Study Team. Computer-assisted semen analysis parameters as predictors for fertility of men from the general population. Hum Reprod 2000;15:1562–7.Google Scholar
Burke, RK, Kapinos, LJ. The effect of in vitro capacitation on sperm velocity and motility as measured by an in-office, integrated, microcomputerized system for semen analysis. Int J Fertil 1985;30:1017.Google Scholar
Kay, VJ, Robertson, L. Hyperactivated motility of human spermatozoa: a review of physiological function and application in assisted reproduction. Hum Reprod Update 1998;776–86.Google Scholar
Mortimer, ST, van der Horst, G, Mortimer, D. The future of computer-aided sperm analysis. Asian J Androl 2015;17:545–53.Google Scholar
Mortimer, ST, De Jonge, CJ. CASA—Computer-aided sperm analysis. In: Skinner, MK, ed. Encyclopedia of Reproduction, 2nd ed, Vol. 5. Boston, MA: Academic Press, Elsevier, 2018; pp. 5963. Available from: dx.doi.org/10.1016/B978-0-12-801238-3.64935-8.Google Scholar
Wang, C, Lee, GS, Leung, A, Surrey, ES, Chan, SY. Human sperm hyperactivation and acrosome reaction and their relationships to human in vitro fertilization. Fertil Steril 1993;59:1221–7.Google Scholar
Sukcharoen, N, Keith, J, Irvine, DS, Aitken, RJ. Definition of the optimal criteria for identifying hyperactivated human spermatozoa at 25 Hz using in-vitro fertilization as a functional end-point. Hum Reprod 1995;10:2928–37.Google Scholar
Wang, C, Leung, A, Tsoi, W-L, et al. Evaluation of human sperm hyperactivated motility and its relationship with the zona-free hamster oocyte sperm penetration assay. J Androl 1991;12:253–7.Google Scholar
Macleod, IC, Irvine, DS, Masterton, A, Taylor, A, Templeton, AA. Assessment of the conventional criteria of semen quality by computer-assisted image analysis: evaluation of the Hamilton-Thorn motility analyser in the context of a service andrology laboratory. Hum Reprod 1994;9:310–19.Google Scholar
ESHRE Andrology Special Interest Group. Consensus workshop on advanced diagnostic andrology techniques. Hum Reprod 1996;11:1463–79.Google Scholar
Yanagimachi, R. In vitro acrosome reaction and capacitation of golden hamster spermatozoa by bovine follicular fluid and its fractions. J Exp Zool 1969;170:269–80.Google Scholar
Bavister, BD. Environmental factors important for in vitro fertilization in the hamster. J Reprod Fertil 1969;18:S544–5.Google Scholar
Edwards, RG, Steptoe, PC, Purdy, JM. Fertilization and cleavage in vitro of preovulatory human oocytes. Nature 1970;227:1307–9.Google Scholar
Bavister, BD. Early history of in vitro fertilization. Reproduction 2002;124:181–96.Google Scholar
Yanagimachi, R, Yanagimachi, H, Rogers, BJ. The use of zona-free animal ova as a test-system for the assessment of the fertilizing capacity of Human spermatozoa. Biol Reprod 1976;15:471–6.Google Scholar
Overstreet, JW, Hembree, WC. Penetration of the zona pellucida of nonliving human oocytes by human spermatozoa in vitro. Fertil Steril 1976;27:815–31.Google Scholar
Rogers, BJ. The sperm penetration assay: its usefulness reevaluated. Fertil Steril 1985;43:821–40.Google Scholar
Yanagimachi, R, Lopata, A, Odom, CB, Bronson, RA, Mahi, CA, Nicolson, GL. Retention of biologic characteristics of zona pellucida in highly concentrated salt solution: the use of salt-stored eggs for assessing the fertilizing capacity of spermatozoa. Fertil Steril 1979;31:562–74.Google Scholar
Burkman, LJ, Coddington, CC, Franken, DR, Krugen, TF, Rosenwaks, Z, Hogen, GD. The hemizona assay (HZA): development of a diagnostic test for the binding of human spermatozoa to the human hemizona pellucida to predict fertilization potential. Fertil Steril 1988;49:688–97.Google Scholar
Franken, DR, Burkman, LJ, Oehninger, SC, et al. Hemizona assay using salt-stored human oocytes: evaluation of zona pellucida capacity for binding human spermatozoa. Gamete Res 1989;22:1526.Google Scholar
Franken, DR, Oosthuizen, WT, Cooper, S, et al. Electron microscopic evidence on the acrosomal status of bound sperm and their penetration into human hemizonae pellucida after storage in a buffered salt solution. Andrologia 1991;23:205–8.Google Scholar
Franken, DR, Windt, ML, Kruger, TF, Oehninger, S, Hodgen, GD. Comparison of sperm binding potential of uninseminated, inseminated-unfertilized, and fertilized-noncleaved human oocytes under hemizona assay conditions. Mol Reprod Dev 1991;30:5661.Google Scholar
Franken, DR, Coddington, CC, Burkman, LJ, et al. Defining the valid hemizona assay: accounting for binding variability within zonae pellucidae and within semen samples from fertile males. Fertil Steril 1991;56:1156–61.Google Scholar
Oehninger, S, Mahony, M, Ozgür, K, Kolm, P, Kruger, T, Franken, D. Clinical significance of human sperm-zona pellucida binding. Fertil Steril 1997;67:1121–7.Google Scholar
Liu, DY, Lopata, A, Johnston, WI, Baker, HW. A human sperm-zona pellucida binding test using oocytes that failed to fertilize in vitro. Fertil Steril 1988;50:782–8.Google Scholar
Liu, DY, Clarke, GN, Lopata, A, Johnston, WI, Baker, HW. A sperm–zona pellucida binding test and in vitro fertilization. Fertil Steril 1989;52:281–7.Google Scholar
Liu, DY, Baker, HW. A new test for the assessment of sperm-zona pellucida penetration: relationship with results of other sperm tests and fertilization in vitro. Hum Reprod 1994;9:489–96.Google Scholar
Liu, DY, Baker, HW. Defective sperm-zona pellucida interaction: a major cause of failure of fertilization in clinical in-vitro fertilization. Hum Reprod 2000;15:702–8.Google Scholar
Sosa, CM, Pavarotti, MA, Zanetti, MN, Zoppino, FC, De Blas, GA, Mayorga, LS. Kinetics of human sperm acrosomal exocytosis. Mol Hum Reprod 2015;21:244–54.Google Scholar
De Jonge, CJ, Barratt, CL. Methods for the assessment of sperm capacitation and acrosome reaction excluding the sperm penetration assay. Methods Mol Biol 2013;927:113–18.Google Scholar
Overstreet, JW, Yanagimachi, R, Katz, DF, Hayashi, K, Hanson, FW. Penetration of human spermatozoa into the human zona pellucida and the zona-free hamster egg: a study of fertile donors and infertile patients. Fertil Steril 1980;33:534–42.Google Scholar
Aitken, RJ, Ross, A, Hargreave, T, Richardson, D, Best, F. Analysis of human sperm function following exposure to the ionophore A23187. Comparison of normospermic and oligozoospermic men. J Androl 1984;5:321–9.Google Scholar
De Jonge, CJ, Mack, SR, Zaneveld, LJD. Synchronous assay for human sperm capacitation and the acrosome reaction. J Androl 1989;10:232–9.Google Scholar
Cummins, JM, Pember, SM, Jequier, AM, Yovich, JL, Hartmann, PE. A test of the human sperm acrosome reaction following ionophore challenge. Relationship to fertility and other seminal parameters. J Androl 1991;12:98103.Google Scholar
Aitken, RJ, Thatcher, S, Glasier, AF, Clarkson, JS, Wu, FC, Baird, DT. Relative ability of modified versions of the hamster oocyte penetration test, incorporating hyperosmotic medium or the ionophore A23187, to predict IVF outcome. Hum Reprod 1987;2:227–31.Google Scholar
Pampiglione, JS, Tan, SL, Campbell, S.The use of the stimulated acrosome reaction test as a test of fertilizing ability in human spermatozoa. Fertil Steril 1993;59:1280–4.Google Scholar
Makkar, G, Ng, EHY, Yeung, WSBY, Ho, PK. The significance of the ionophore-challenged acrosome reaction in the prediction of successful outcome of controlled ovarian stimulation and intrauterine insemination. Hum Reprod 2003;18:534–9.Google Scholar
Katsuki, T, Hara, T, Ueda, K, Tanaka, J, Ohama, K. Prediction of outcomes of assisted reproduction treatment using calcium ionophore-induced acrosome reaction. Hum Reprod 2005;20:469–75.Google Scholar
Krausz, C, Bonaccorsi, L, Luconi, M, et al. Intracellular calcium increase and acrosome reaction in response to progesterone in human spermatozoa are correlated with in-vitro fertilization. Hum Reprod 1995;10:120–4.Google Scholar
Krausz, C, Bonaccorsi, L, Maggio, P, et al. Two functional assays of sperm responsiveness to progesterone and their predictive values in in-vitro fertilization. Hum Reprod 1996;11:1661–7.Google Scholar
Cross, NL, Morales, P, Overstreet, JW, Hanson, FW. Induction of acrosome reactions by the human zona pellucida. Biol Reprod 1988;38:235–44.Google Scholar
Bielfeld, P, Faridi, A, Zaneveld, LJ, De Jonge, CJ. The zona pellucida-induced acrosome reaction of human spermatozoa is mediated by protein kinases. Fertil Steril 1994;61:536–41.Google Scholar
Franken, DR, Bastiaan, HS, Oehninger, SC. Physiological induction of the acrosome reaction in human sperm: validation of a microassay using minimal volumes of solubilized, homologous zona pellucida. J Assist Reprod Genet 2000;17:374–8.Google Scholar
Franken, DR, Bastiaan, HS, Kidson, A, Wranz, P, Habenicht, UF. Zona pellucida mediated acrosome reaction and sperm morphology. Andrologia 1997;29:311–17.Google Scholar
De Jonge, CJ. The diagnostic significance of the induced acrosome reaction. Reprod Med Rev 1994;3:159–78.Google Scholar
Buttke, DE, Nelson, JL, Schlegel, PN, Hunnicutt, GR, Travis, AJ. Visualization of GM1 with cholera toxin B in live epididymal versus ejaculated bull, mouse, and human spermatozoa. Biol Reprod 2006;74:889–95.Google Scholar
Selvaraj, V, Asano, A, Buttke, DE, Sengupta, P, Weiss, RS, Travis, AJ. Mechanisms underlying the micron-scale segregation of sterols and GM1 in live mammalian sperm. J Cell Physiol 2009;218:522–36.Google Scholar
Moody, MA, Cardona, C, Simpson, AJ, Smith, TT, Travis, AJ, Ostermeier, GC. Validation of a laboratory-developed test of human sperm capacitation. Mol Reprod Dev 2017;84:408–22.Google Scholar
Cardona, C, Neri, QV, Simpson, AJ, et al. Localization patterns of the ganglioside GM1 in human sperm are indicative of male fertility and independent of traditional semen measures. Mol Reprod Dev 2017;84:423–35.Google Scholar
Babigumira, JB, Sharara, FI, Garrison, LP Jr. Projecting the potential impact of the Cap-Score on clinical pregnancy, live births, and medical costs in couples with unexplained infertility. J Assist Reprod Genet 2018;35:99106.Google Scholar
Schinfeld, J, Sharara, F, Morris, R, et al. Cap-Score prospectively predicts probability of pregnancy. Mol Reprod Dev 2018;85:654–64.Google Scholar
Kovalski, NN, de Lamirande, E, Gagnon, C. Reactive oxygen species generated by human neutrophils inhibit sperm motility: protective effect of seminal plasma and scavengers. Fertil Steril 1992;58:809–16.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 Fert 1987;81:459–69.Google Scholar
Aitken, RJ, Clarkson, JS, Fishel, S. Generation of reactive oxygen species, lipid peroxidation, and human sperm function. Biol Reprod 1989;41:183–97.Google Scholar
Aitken, J, Krausz, C, Buckingham, D. Relationships between biochemical markers for residual sperm cytoplasm, reactive oxygen species generation, and the presence of leukocytes and precursor germ cells in human sperm suspensions. Mol Reprod Dev 1994;39:268–79.Google Scholar
de Lamirande, E, Gagnon, C. A positive role for the superoxide anion in triggering hyperactivation and capacitation of human spermatozoa. Int J Androl 1993;16:21–5.Google Scholar
Aitken, RJ, Gordon, E, Harkiss, D, et al. Relative impact of oxidative stress on the functional competence and genomic integrity of human spermatozoa. Biol Reprod 1998;59:1037–46.Google Scholar
Tremellen, K. Oxidative stress and male infertility – a clinical perspective. Hum Reprod Update 2008;14:243–58.Google Scholar
Aitken, RJ, Smith, TB, Jobling, MS, Baker, MA, De Iuliis, GN. Oxidative stress and male reproductive health. Asian J Androl 2014;16:31–8.Google Scholar
Aitken, RJ, Gibb, Z, Baker, MA, Drevet, J, Gharagozloo, P. Causes and consequences of oxidative stress in spermatozoa. Reprod Fertil Devel 2016;28:110.Google Scholar
Agarwal, A, Majzoub, A. Laboratory tests for oxidative stress. Indian J Urol 2017;33:199206.Google Scholar
Wagner, H, Cheng, JW, Ko, EY. Role of reactive oxygen species in male infertility: an updated review of literature. Arab J Urol 2018;16:3543.Google Scholar
De Jonge, CJ. A clinical assay for reactive oxygen species – ready for primetime? Reprod Biomed Online 2018;36:88–9.Google Scholar
Balhorn, R. A model for the structure of chromatin in mammalian sperm. J Cell Biol 1982;93:298305.Google Scholar
Ward, WS, Coffey, DS. DNA packaging and organization in mammalian spermatozoa: comparison with somatic cells. Biol Reprod 1991;44:569–74.Google Scholar
Fuentes-Mascorro, G, Serrano, H, Rosado, A. Sperm chromatin. Arch Androl 2000;45:215–25.Google Scholar
Sakkas, D, Moffatt, O, Manicardi, GC, Mariethoz, E, Tarozzi, N, Bizzaro, D. Nature of DNA damage in ejaculated human spermatozoa and the possible involvement of apoptosis. Biol Reprod 2002;66:1061–7.Google Scholar
Aitken, RJ, De Iuliis, GN. On the possible origins of DNA damage in human spermatozoa. Mol Hum Reprod 2010;16:313.Google Scholar
Sakkas, D, Alvarez, JG. Sperm DNA fragmentation: mechanisms of origin, impact on reproductive outcome, and analysis. Fertil Steril 2010;93:1027–36.Google Scholar
Aitken, RJ, Bronson, R, Smith, TB, De Iuliis, GN. The source and significance of DNA damage in human spermatozoa; a commentary on diagnostic strategies and straw man fallacies. Mol Hum Reprod 2013;19:475–85.Google Scholar
Barratt, CL, Aitken, RJ, Björndahl, L, et al. Sperm DNA: organization, protection and vulnerability: from basic science to clinical applications – a position report. Hum Reprod 2010;25:824–38.Google Scholar
Pacey, AA. Environmental and lifestyle factors associated with sperm DNA damage. Hum Fertil 2010;13:189–93.Google Scholar
Evenson, DP, Jost, LK, Marshall, D, et al. Utility of the sperm chromatin structure assay as a diagnostic and prognostic tool in the human fertility clinic. Hum Reprod 1999;14:1039–49.Google Scholar
Spanò, M, Bonde, JP, Hjøllund, HI, Kolstad, HA, Cordelli, E, Leter, G. Sperm chromatin damage impairs human fertility. The Danish First Pregnancy Planner Study Team. Fertil Steril 2000;73:4350.Google Scholar
Evenson, DP, Darzynkiewicz, Z, Melamed, MR. Relation of mammalian sperm chromatin heterogeneity to fertility. Science 1980;210:1131–3.Google Scholar
Evenson, DP, Larson, KL, Jost, LK. Sperm chromatin structure assay: its clinical use for detecting sperm DNA fragmentation in male infertility and comparisons with other techniques. J Androl 2002;23:2543.Google Scholar
Hughes, CM, Lewis, SE, McKelvey-Martin, VJ, Thompson, W. A comparison of baseline and induced DNA damage in human spermatozoa from fertile and infertile men, using a modified comet assay. Mol Hum Reprod 1996;2:613–19.Google Scholar
Hughes, CM, Lewis, SE, McKelvey-Martin, VJ, Thompson, W. Reproducibility of human sperm DNA measurements using the alkaline single cell gel electrophoresis assay. Mutat Res 1997;374:261–8.Google Scholar
Sun, JG, Jurisicova, A, Casper, RF. Detection of deoxyribonucleic acid fragmentation in human sperm: correlation with fertilization in vitro. Biol Reprod 1997;56:602–7.Google Scholar
Sharma, RK, Sabanegh, E, Mahfouz, R, Gupta, S, Thiyagarajan, A, Agarwal, A. TUNEL as a test for sperm DNA damage in the evaluation of male infertility. Urology 2010;76:1380–6:Google Scholar
Fernández, JL, Muriel, L, Rivero, MT, Goyanes, V, Vazquez, R, Alvarez, JG. The sperm chromatin dispersion test: a simple method for the determination of sperm DNA fragmentation. J Androl 2003;24:5966.Google Scholar
Fernández, JL, Muriel, L, Goyanes, V, et al. Simple determination of human sperm DNA fragmentation with an improved sperm chromatin dispersion (SCD) test. Fertil Steril 2005;84:833–42.Google Scholar
Fernández, JL, Muriel, L, Goyanes, V, et al. Halosperm® is an easy, available, and cost-effective alternative for determining sperm DNA fragmentation. Fertil Steril 2005;84:860.Google Scholar
Li, Z, Wang, L, Cai, J, Huang, H. Correlation of sperm DNA damage with IVF and ICSI outcomes: a systematic review and meta-analysis. J Assist Reprod Genet 2006;23:367–76.Google Scholar
Evenson, D, Wixon, R. Meta-analysis of sperm DNA fragmentation using the sperm chromatin structure assay. Reprod Biomed Online 2006;12:466–72.Google Scholar
Zini, A, Boman, JM, Belzile, E, Ciampi, A. Sperm DNA damage is associate with an increased risk of pregnany after IVF and ICSI: systematic review and meta-analysis. Hum Reprod 2008;23:2663–8.Google Scholar
Shamsi, MB, Imam, SN, Dada, R. Sperm DNA integrity assays: diagnostic and prognostic challenges and implications in management of infertility. J Assist Reprod Genet 2011;28:1073–85.Google Scholar
Robinson, L, Gallos, ID, Conner, SJ, et al. The effect of sperm DNA fragmentation on miscarriage rates: a systematic review and meta-analysis. Hum Reprod 2012;27:2908–17.Google Scholar
Zhao, J, Zhang, Q, Wang, Y, Li, Y. Whether sperm deoxyribonucleic acid fragmentation has an effect on pregnancy and miscarriage after in vitro fertilization/intracytoplasmic sperm injection: a systematic review and meta- analysis. Fertil Steril 2014;102:9981005Google Scholar
Zhang, Z, Zhu, L, Jiang, H, Chen, H, Chen, Y, Dai, Y. Sperm DNA fragmentation index and pregnancy outcome after IVF or ICSI: a meta-analysis. J Assist Reprod Genet 2015;32:1726.Google Scholar
Osman, A, Alsomait, H, Seshadri, S, El-Toukhy, T, Khalaf, Y. The effect of sperm DNA fragmentation on live birth rate after IVF or ICSI: a systematic review and meta-analysis. Reprod Biomed Online 2015;30:120–7.Google Scholar
Cissen, M, Wely, MV, Scholten, I, et al. Measuring sperm DNA fragmentation and clinical outcomes of medically assisted reproduction: a systematic review and meta-analysis. PLoS ONE 2016;11:e0165125.Google Scholar
Simon, L, Zini, A, Dyachenko, A, Ciampi, A, Carrell, DT. A systematic review and meta-analysis to determine the effect of sperm DNA damage on in vitro fertilization and intracytoplasmic sperm injection outcome. Asian J Androl 2017;19:8090.Google Scholar
Pacey, A. Is sperm DNA fragmentation a useful test that identifies a treatable cause of male infertility? Best Pract Res Clin Obstet Gynaecol 2018;53:1119.Google Scholar
Sugihara, A, Van Avermaete, F, Roelant, E, Punjabi, U, De Neubourg, D. The role of sperm DNA fragmentation testing in predicting intra-uterine insemination outcome: a systematic review and meta-analysis. Eur J Obstet Gynecol Reprod Biol 2019;244:815.Google Scholar
McQueen, DB, Zhang, J, Robins, JC. Sperm DNA fragmentation and recurrent pregnancy loss: a systematic review and meta-analysis. Fertil Steril 2019;112:5460.Google Scholar
Carrell, DT, De Jonge, CJ. The troubling state of the semen analysis. Andrology 2016;4:761–2.Google Scholar
De Jonge, CJ. Semen analysis: Looking for an upgrade in class. Fertil Steril 2012;97:260–6.Google Scholar
Palermo, G, Joris, H, Devroey, P, Van Steirteghem, AC. Pregnancies after intracytoplasmic injection of single spermatozoon into an oocyte. Lancet 1992;340:1718.Google Scholar
Carrell, DT, Nyboe Andersen, A, Lamb, DJ. The need to improve patient care through discriminate use of intracytoplasmic sperm injection (ICSI) and improved understanding of spermatozoa, oocyte and embryo biology. Andrology 2015;3:143–6.Google Scholar
Barratt, CLR, De Jonge, CJ, Sharpe, RM. ‘Man up’: the importance and strategy for placing male reproductive health centre stage in the political and research agenda. Hum Reprod 2018;33:541–45.Google Scholar
Li, Z, Wang, AY, Bowman, M, et al. ICSI does not increase the cumulative live birth rate in non-male factor infertility. Hum Reprod 2018;33:1322–30.Google Scholar
The Practice Committees of the American Society for Reproductive Medicine and Society for Assisted Reproductive Technology. Intracytoplasmic sperm injection (ICSI) for non-male factor infertility: a committee opinion. Fertil Steril 2012;98:1395–9.Google Scholar
De Jonge, C, Barratt, CLR. The present crisis in male reproductive health: an urgent need for a political, social, and research roadmap. Andrology 2019;7:762–8.Google Scholar
Barratt, CLR, Björndahl, L, De Jonge, CJ, et al. The diagnosis of male infertility: an analysis of the evidence to support the development of global WHO guidance – challenges and future research opportunities. Hum Reprod Update 2017;23:660–80.Google Scholar
Esteves, SC, Roque, M, Bedoschi, G, Haahr, T, Humaidan, P. Intracytoplasmic sperm injection for male infertility and consequences for offspring. Nat Rev Urol 2018;15:535–62.Google Scholar
Barratt, CL, Anderson, RA, De Jonge, C. Male fertility: a window on the health of this generation and the next. Reprod Biomed Online 2019;39:721–3.Google Scholar

References

Howard, SS. Antoine van Leeuwenhoek and the discovery of sperm. Fertil Steril 1997;67:1617.Google Scholar
Jensen, K. Reproduction: The Cycle of Life. Pinchot, PB, ed. US News Books, 1957.Google Scholar
Bunge, RG, Sherman, JK. Fertilizing capacity of human spermatozoa. Nature 1953; 172:767–8.Google Scholar
Sherman, JK. Synopsis on the use of frozen human semen since 1964: state of the art of human semen banking. Fertil Steril 1973;24:397.Google Scholar
Bunge, RG, Kettel, WC, Sherman, JK. Clinical use of frozen semen: report of four cases. Fertil Steril 1954;5:520–9.Google Scholar
Edwards, RG, Steptoe, PC, Purdy, JM. Establishing full-term human pregnancies using cleaving embryos grown in vitro. Br J Obstet Gynaecol 1980;87:737–56.Google Scholar
Palermo, G, Joris, H, Devroy, P, Van Steirteghem, AC. Pregnancies after intracytoplasmic injection of single spermatozoon into an oocyte. Lancet 1992; 340:1718.Google Scholar
Keel, BA, Black, JB. Reduced motility longevity in thawed human spermatozoa. Arch Androl 1980;4:213–15.Google Scholar
Lovelock, JE. The haemolysis of human red blood-cells by freezing and thawing. Biochim Biophys Acta 1953;10:414–26.Google Scholar
Meryman, H. Modified model for the mechanism of freezing injury in erythrocytes. Nature 1968;218:333–6.Google Scholar
Mazur, P, Leibo, S, Chu, E. A two-factor hypothesis of freezing injury. evidence from Chinese hamster tissue-culture cells. Exp Cell Res 1972;71:345–55.Google Scholar
Gao, D, Mazur, P, Critser, JK. Fundamental cryobiology of mammalian spermatozoa. In: Karow, AM, Critser, JK, eds. Reproductive Tissue Banking: Scientific Principles. San Diego, CA: Academic Press, 1997; pp. 263328.Google Scholar
Pegg, DE. The history and principles of cryopreservation. Sem Reprod Med 2002;20:513.Google Scholar
Mortimer, D. Concepts and future concepts and practices in human sperm cryobanking. Reprod BioMed Online 2004;9:134–51.Google Scholar
Walters, EM, et al. The history of sperm cryopreservation. In: Pacey, AA, Tomlinson, MJ, eds. Sperm Banking: Theory and Practice. Cambridge: Cambridge University Press, 2009; pp. 117.Google Scholar
Behjati-Ardakani, Z, Akhondi, MM, Mahmoodzadeh, H, Hosseini, SH. An evaluation of the historical importance of fertility and its reflection in ancient mythology. J Reprod Infertil 2016;17:29.Google Scholar
Sherman, JK. Research on frozen human sperm: past, present and future. Fertil Steril 1964;15:485–99.Google Scholar
Ombelet, W, van Robays, J. Artificial insemination history: hurdles and milestones. Facts Views Vis Obgyn 2015;7:137–43.Google Scholar
Hard, AD. Artificial impregnation. Med World 1909;27:163.Google Scholar
Shettles, LB. The respiration of human spermatozoa and their response to various gases and low temperatures. Am J Physiol 1940;128:408.Google Scholar
Polge, C, Smith, UA, Parkes, AS. Revival of spermatozoa after vitrification and dehydration at low temperatures. Nature 1949;164:666–76.Google Scholar
Smith, AU, Polge, C. Survival of sperm at low temperatures. Nature 1950;166:668–9.Google Scholar
Polge, C, Lovelock, JE. Preservation of bull sperm at −70C. Vet Rec 1952;64:296–7.Google Scholar
Smith, AU. Behaviour of fertilized rabbit eggs exposed to glycerol and to low temperatures. Nature 1952;170:374–5.Google Scholar
Whittingham, DG, Leibo, SP, Mazur, P. Survival of mouse embryos frozen at −196°C and −269°C. Science 1972;178:411–14.Google Scholar
Mazur, P. Freezing of living cells: mechanisms and implications. Am J Physiol 1984;247:C125–42.Google Scholar
Macomber, D, Sanders, MB. The sperm count. N Engl J Med 1929;200:981–4.Google Scholar
Snowden, R, Mitchell, GD (eds). Artificial Family: Consideration of Artificial Insemination by Donor. London: Allen and Unwin, 1981.Google Scholar
Wolstenholme, GEW, Fitzsimons, DW (eds). Law and Ethics of A.I.D. and Embryo Transfer. Amsterdam: Ciba Foundation Symposium 17, Associated Scientific Publishers, 1973.Google Scholar
Swanson, KW. The birth of the sperm bank. The Annals of Iowa 2012;71:241–76.Google Scholar
Sherman, JK. Current status of clinical cryobanking of human semen. In: Paulson, JD, Negro-Vilar, A, Lucena, E, Martini, L, eds. Andrology: Male Fertility and Sterility. Orlando, FL: Academic Press, 1986; pp. 517–47.Google Scholar
Muller, HJ. Human evolution by voluntary choice of germ plasma service. Science 1961; 134:643–9.Google Scholar
People of the State of California, Plantiff and Respondent v. Folmer J. Sorensen, Defendant and Appellent, Cr. 6420, September 29, 1967.Google Scholar
Rothman, CA. Method of obtaining viable sperm in the post-mortem state. Fertil Steril 1980;34:512.Google Scholar
Los Angeles Times. Baby is born using sperm from dead father. March 27, 1999.Google Scholar
Rall, WF, Reid, DS, Farrant, J. Innocuous biological freezing during warming. Nature 1980;286:511–14.Google Scholar
Morris, GJ. Rapidly cooled human sperm: no evidence of intracellular ice formation. Hum Reprod 2006;21:2075–83.Google Scholar
Benson, JD, Woods, EJ, Walters, EM, Critser, JK. The cryobiology of spermatozoa. Theriogenology 2012; 78:1682–99.Google Scholar
Wowk, B. Thermodynamic aspects of vitrification. Cryobiology 2010;60:1122.Google Scholar
Nawroth, F, Isachenko, V, Dessole, S, et al. Vitrification of human spermatozoa without cryoprotectants. Cryo Letters 2002;23:93102.Google Scholar
Isachenko, E, Isachenko, V, Katkov, II, et al. Vitrification of mammalian spermatozoa in he absence of cryoprotectants: from past practical difficulties to present success. Reprod BioMed Online 2003;6:191200.Google Scholar
Mahadevan, M, Trounson, AO. Effect of cryoprotective media and dilution methods on the preservation of human spermatozoa. Andrologia 1983;15:355–66.Google Scholar
Gao, DY, Liu, J, Liu, C, et al. Prevention of osmotic injury to human spermatozoa during addition and removal of glycerol. Hum Reprod 1995;10:1109–22.Google Scholar
Gilmore, J, Liu, J, Gao, DY, Critser, JK. Determination of optimal cryoprotectants and procedures for their addition and removal from human spermatozoa. Hum Reprod 1997;12:112–18.Google Scholar
Gilmore, JA, Liu, J, Woods, EJ, Peter, AT, Critser, JK. Cryoprotective agent and temperature effects on human sperm membrane permeabilities: convergence of theoretical and empirical approaches for optimal cryopreservation methods. Hum Reprod 2000;15:335–43.Google Scholar
Jeyendran, RS, Van der Ven, HH, Kennedy, W, Perez-Pelaez, M, Zaneveld, LJD. Comparison of glycerol and a zwitter ion buffer system as cryoprotective media for human spermatozoa. J Androl 1984;5:17.Google Scholar
Keel, BA, Webster, BW, Roberts, DK. Effects of cryopreservation on the motility characteristics of human spermatozoa. J Reprod Fert 1987;81:213–20.Google Scholar
Weidel, L, Prins, GS. Cryosurvival of human spermatozoa frozen in eight different buffer systems. J Androl 1987;8:41–7.Google Scholar
Stanic, P, Tandara, M, Sonicki, Z, et al. Comparison of protective media and freezing techniques for cryopreservation of human semen. Eur J Obstet Gynecol Reprod Biol 2000;91:6570.Google Scholar
Critser, JK, Huse-Benda, AR, Aaker, DV, Arneson, BW, Ball, GD. Cryopreservation of human spermatozoa. I. Effects of holding procedure and seeding on motility, fertilizability, and acrosome reaction. Fertil Steril 1987;47:656–63.Google Scholar
Critser, JK, Arneson, BW, Aaker, DV, Huse-Benda, AR, Ball, GD. Cryopreservation of human spermatozoa. II. Post-thaw chronology of motility and of zona-free hamster ova penetration. Fertil Steril 1987;47:980–4.Google Scholar
Critser, JK, Huse-Benda, AR, Aaker, DV, Arneson, BW, Ball, GD. Cryopreservation of human spermatozoa. III. The effect of cryoprotectants on motility. Fertil Steril 1988;50:314–20.Google Scholar
Henry, MA, Noiles, EE, Gao, D, Mazur, P, Critser, JK. Cryopreservation of human spermatozoa. IV. The effects of cooling rate and warming rate on the maintenance of motility, plasma membrane integrity, and mitochondrial function. Fertil Steril 1993;60:911–18.Google Scholar
Punyatanasakchai, P, Sophonsritsuk, A, Weerakiet, S, Wansumrit, S, Chompurat, D. Comparison of cryopreserved human sperm in vapor and liquid phases of liquid nitrogen: effect on motility parameters, morphology, and sperm function. Fertil Steril 2008;90:1978–82.Google Scholar
Isachenko, E, Isachenko, V, Sanchez, R, Katkov, II, Kreienberg, R. Cryopreservation of spermatozoa: old routine and new perspectives. In: Donnez, J, Kim, SS, eds. Principles and Practice of Fertility Preservation. Cambridge: Cambridge University Press, 2011; pp. 176–98.Google Scholar
Isachenko, V, Maettner, R, Petrunkina, AM, et al. Cryoprotectant-free vitrification of human spermatozoa in large (up to 0.5 mL) volume: a novel technology. Clin Lab 2011;57(9–10):643–50.Google Scholar
Whittingham, DG, Wood, M, Farrant, J, Lee, H, Halsey, JA. Survival of frozen mouse embryos after rapid thawing from −196 degrees C. J Reprod Fertil 1979;56:1121.Google Scholar
Willadsen, SM, Polge, C, Trounson, AO, Rowson, LEA. Transplantation of sheep and cattle embryos after storage at −196°C. In: Elliott, K, Whelan, J, eds. The Freezing of Mammalian Embryos. Amsterdam: Elsevier Excerpta Medica, 1977; pp. 190201.Google Scholar
Seki, S, Mazur, P. Effect of warming rate on the survival of vitrified mouse oocytes and on the recrystallization of intracellular ice. Biol Reprod 2008;79:727–37.Google Scholar
Seki, S, Mazur, P. The dominance of warming rate over cooling rate in the survival of mouse oocytes subjected to a vitrification procedure. Cryobiology 2009;59:7982.Google Scholar
Calamera, JC, Buffone, MG, Doncel, GF, et al. Effect of thawing temperature on the motility recovery of cryopreserved human spermatozoa. Fertil Steril 2010;93:789–94.Google Scholar
Whitney, JB, Anderson, RE, Schiewe, MC. Single center validation of routine blastocyst biopsy implementation. J Assist Reprod Genet 2016;33:1507–13.Google Scholar
Anderson, RE, Whitney, JB, Schiewe, MC. Clinical benefits of preimplantation genetic testing for aneuploidy (PGT-A) for all in vitro fertilization treatment cycles. J Eur Med Genet 2020;63:103731.Google Scholar
Manjunath, P. New insights into the understanding of the mechanism of sperm protection by extender components. Anim Reprod 2012;9:809–15.Google Scholar
Hezavehei, M, Shara, M, Kouchesfahani, HM, et al. Sperm cryopreservation: a review on current molecular cryobiology and advanced approaches. Reprod BioMed Online 2019;37:327–39.Google Scholar
Di Santo, M, Tarozzi, N, Nadalini, M, Borini, A. Human sperm cryopreservation: update on techniques, effect on DNA integrity, and implications for ART. Adv Urol 2012;854837:112.Google Scholar
Saritha, KR, Bongso, A. Comparative evaluation of fresh and washed human sperm cryopreserved in vapor and liquid phases of liquid nitrogen. J Androl 2001;22:857–62.Google Scholar
Chaveiro, A, Liu, J, Engel, B, Critser, JK, Woelders, H. Significant variability among bulls in the sperm membrane permeability for water and glycerol: possible implications for semen freezing protocols for individual males. Cryobiology 2006;53:349–59.Google Scholar
Watson, PF. The preservation of semen in mammals. In: Finn, CA, ed. Oxford Reviews of Reproductive Biology. Oxford: Oxford University Press, 1979; pp. 283350.Google Scholar
Critser, JK. Current status of semen cryobanking in the USA. Hum Reprod 1998;13 (Suppl 2):5569.Google Scholar
Moutos, CP, Lahham, R, Phelps, JY. Cryostorage failures: a medicolegal review. J Assist Reprod Genet 2019;36:10411048.Google Scholar
Cassou, R. [La méthode des pailletes en plastique adaptée à la généralisation de la conqélation]. In: Proc 5th Int Congr Anim Reprod. Trento: Italy, 1964;4:540–6.Google Scholar
Schiewe, MC, Schmidt, PM, Pontbriand, D, Wildt, DE. Toxicity potential of residual ethylene oxide on embryos cryopreserved in gas-sterilized plastic straws. Gamete Res 1988;19:31–9.Google Scholar
Benifla, J-L, Letur-Konçrsch, H, Collin, G, et al. Safety of cryopreservation straws for human gametes or embryos: a preliminary study with human immunodeficiency virus-1. Hum Reprod 2000:15:2186–9.Google Scholar
Pomeroy, KO, Harris, S, Conaghan, J, et al. Storage of cryopreserved reproductive tissues: evidence that cross-contamination of infectious agents is a negligible risk. Fertil Steril 2010;94:1181–8.Google Scholar
Bielanski, A, Bergeron, H, Lau, PCK, Devenish, J. Microbial contamination of embryos and semen during long term banking in liquid nitrogen. Cryobiology 2003;46:146–52.Google Scholar
Tedder, RS, Zuckerman, MA, Goldstone, AH, et al. Hepatitis B transmission from contaminated cryopreservation tanks. Lancet 1995;346:137–40.Google Scholar
Fountain, D, Ralston, M, Higgins, N, et al. Liquid nitrogen freezers: a potential source of microbial contamination of hematopoietic stem cell components. Transfusion 1997;37:585–91.Google Scholar
Cobo, A, Romero, JL, Perez, S, et al. Storage of human oocytes in the vapor phase of nitrogen. Fertil Steril 2010;94:1903–7.Google Scholar
Pomeroy, K. Liquid nitrogen storage tank failure: can we improve the current system? 2018. Available from: www.fertstertdialog.com.Google Scholar
Schiewe, MC, Freeman, MF, Whitney, JB, et al. Comprehensive assessment of cryogenic storage risk and quality management concerns: best practice guidelines for ART labs. J Assist Reprod Genet 2019;36:514.Google Scholar
International Society for Biological and Environmental Repositories (ISBER). Best Practices Addendum: Liquid Nitrogen-Based Cryogenic Storage of Specimens, 4th ed. Vancouver, BC: ISBER, 2019.Google Scholar
Kupker, W, Schlegel, PN, Al-Hasani, S, et al. Use of frozen-thawed testicular sperm for intracytoplasmic sperm injection. Fertil Steril 2000;73:453–8.Google Scholar
Levran, D, Ginath, S, Farhi, J, Nahum, H, Glezerman, M, Weissman, A. Timing of testicular sperm retrieval procedures and in vitro fertilization – intracytoplasmic sperm injection outcome. Fertil Steril 2001; 76:380–3.Google Scholar
Schiewe, MC, Rothman, C, Spitz, A, Werthman, PE, Zeitlin, SI, Anderson, RE. Validation-verification of a highly effective, practical human testicular tissue in vitro culture-cryopreservation procedure aimed to optimize pre-freeze and post-thaw motility. J Assist Reprod Genet 2016;33:519–28.Google Scholar
Habermann, H, Seo, R, Cieslak, J, et al. In vitro fertilization outcomes after intracytoplasmic sperm injection with fresh or frozen-thawed testicular spermatozoa. Fertil Steril 2000;73:955–60.Google Scholar
Park, YS, Lee, SH, Song, SJ, Jun, JH, Koong, MK, Seo, JT. Influence of motility on the outcome of in vitro fertilization/intracytoplasmic sperm injection with fresh vs. frozen testicular sperm from men with obstructive azoospermia. Fertil Steril 2003;80:526–30.Google Scholar
Verheyen, G, De Croo, I, Tournaye, H, Pletinex, I, Devroey, P, Van Steirteghem, A. Comparison of four mechanical methods to retrieve spermatozoa from testicular tissue. Hum Reprod 1995;10:2956–9.Google Scholar
Crabbé, E, Verheyen, G, Silber, S, et al. Enzymatic digestion of testicular tissue may rescue the intracytoplasmic sperm injection in some patients with non-obstructive azoospermia. Hum Reprod 1998;13:2791–6.Google Scholar
Crabbé, E, Verheyen, G, Tournaye, H, Van Steirteghem, A. Freezing of testicular sperm as a minced suspension preserves sperm quality better than whole-biopsy freezing when glycerol is used as cryoprotectant. Int J Androl 1999:22:43–8.Google Scholar
Zhu, J, Tsirigotis, M, Pelekanos, M, Craft, I. In-vitro maturation of human testicular spermatozoa. Hum Reprod 1996;11:231–2.Google Scholar
Edirisinghe, WR, Junk, SM, Matson, PL, Yovich, JL. Changes in the motility patterns during in vitro culture of fresh and frozen-thawed testicular and epididymal spermatozoa: implications for planning treatment by intracytoplasmic sperm injection. Hum Reprod 1996;11:2474–6.Google Scholar
Aoki, VW, Wilcox, AL, Thorp, C, Hamiliton, BD, Carrell, DT. Improved in vitro fertilization, embryo quality and pregnancy rates with intracytoplasmic sperm injection of sperm from fresh testicular biopsy samples vs. frozen biopsy samples. Fertil Steril 2004;82:1532–5.Google Scholar
Allan, JA, Cotman, AS. A new method of freezing testicular biopsy sperm: three pregnancies with sperm extracted from cryopreserved sections of seminiferous tubule. Fertil Steril 1997;68:741–4.Google Scholar
Liu, J, Tsai, YL, Katz, E, Compton, G, Garcia, JE, Baramki, TA. Outcome of in-vitro culture of fresh and frozen-thawed human testicular spermatozoa. Hum Reprod 1997;12:1667–72.Google Scholar
Hu, Y, Maxson, WS, Hoffman, DI, Ory, SJ, Licht, MR, Eager, S. Clinical application of intracytoplasmic sperm injection using in vitro cultured testicular spermatozoa obtained the day before egg retrieval. Fertil Steril 1999;72:666–9.Google Scholar
Tasdemir, I, Tasdemir, M, Tavukeuoglu, S. Effect of pentoxyfylline on immotile testicular spermatozoa. J Assist Reprod Genet 1998;15:90–2.Google Scholar
Mangoli, V, Mangoli, R, Dandekar, S, Suri, K, Desai, S. Selection of viable spermatozoa from testicular biopsies: a comparative study between pentoxifylline and hypoosmotic swelling test. Fertil Steril 2011;95:631–4.Google Scholar
Angelopoulos, T, Adler, A, Krey, L, Licciardi, F, Noyes, N, McCullough, A. Enhancement or initiation of testicular sperm motility by in vitro culture of testicular tissue. Fertil Steril 1999;71:240–3.Google Scholar
Balaban, B, Urman, B, Sertac, A, et al. In-vitro culture of spermatozoa induces motility and increases implantation and pregnancy rates after testicular sperm extraction and intracytoplasmic sperm injection. Hum Reprod 1999;14:2808–11.Google Scholar
Navara, CS, Simerly, C, Zoran, S, Schatten, G. The sperm centrosome during fertilization in mammals: implications for fertility and reproduction. Reprod Fertil Dev 1995;7:747–54.Google Scholar
Luyet, BJ, Hodapp, EL. Revival of frog’s spermatozoa vitrified in liquid air. Proc Soc Exp Biol Med 1938;49:433.Google Scholar
Isachenko, V, Isachenko, E, Montag, M, et al. Clean technology for cryoprotectant-free vitrification of human spermatozoa. Reprod BioMed Online 2005;10:350–4.Google Scholar
Isachenko, E, Isachenko, V, Weiss, JM, et al. Acrosomal status and mitochondrial activity of human spermatozoa vitrified with sucrose. Reproduction 2008;136:167–73.Google Scholar
Kuznyetsov, V, Moskovtsev, SI, Crowe, M, Lulat, AG, Librach, CL. Vitrification of a small number of spermatozoa in normozoospermic and severely oligozoospermic samples. Syst Biol Reprod Med 2015;61:1317.Google Scholar
Isachenko, V, Isachenko, E, Katkov, II, et al. Cryoprotectant-free cryo-preservation of human spermatozoa by vitrification and freezing in vapor: effect on motility, DNA integrity, and fertilization ability. Biol Reprod 2004;71:1167–73.Google Scholar
Isachenko, E, Isachenko, V, Katkov, II, et al. DNA integrity and motility of human spermatozoa after standard slow freezing versus cryoprotectant-free vitrification. Hum Reprod 2004;19:932–9.Google Scholar
Isachenko, E, Isachenko, V, Weiss, JM. et al. Acrosomal status and mitochondrial activity of human spermatozoa vitrified with sucrose. Reproduction 2008;136:167–73.Google Scholar
Isachenko, V, Maettner, R, Petrunkina, AM, et al. Vitrification of human ICSI/IVF spermatozoa without cryoprotectants: new capillary technology. J Androl 2012;33:462–8.Google Scholar
Isachenko, E, Mallmann, P, Rahimi, G,et al. Vitrification technique: new possibilities for male gamete low temperature storage. In: Katkov, II, ed. Frontiers in Cryobiology. Croatia: InTech, 2012; pp. 4176.Google Scholar
Agha-Rahimi, A, Khalili, MA, Nabi, A, Ashourzadeh, S. Vitrification is not superior to rapid freezing of normozoospermic spermatozoa: effects on sperm parameters, DNA fragmentation and hyaluronan binding. Reprod BioMed Online 2014;28:352–8.Google Scholar
Isachenko, V, Isachenko, E, Petrunkina, AM, Sanchez, R. Human spermatozoa vitrified in the absence of permeable cryoprotectants: birth of two healthy babies. Reprod Fertil Dev 2011;24:323–6.Google Scholar
Mansilla, MA, Merino, O, Risopatrón, J, Isachenko, V, Isachenko, E, Sánchez, R. High temperature is essential for preserved human sperm function during the devitrification process. Andrologia 2015;48:13.Google Scholar
Sánchez, R, Isachenko, V, Petrunkina, AM, et al. Live birth after intrauterine insemination with spermatozoa from an oligoasthenozoospermic patient vitrified without permeable cryoprotectants. J Androl 2011;33:559–62.Google Scholar
Sánchez, R, Risopatrón, J, Schulz, M, et al. Vitrified sperm banks: the new aseptic technique for human spermatozoa allows cryopreservation at −86°C. Andrologia 2012;44:433–5.Google Scholar
Moskovtsev, SI, Lulat, AG-M, Librach, CL. Cryopreservation of human spermatozoa by vitrification vs slow freezing: Canadian experience. In: Katkov, II, ed. Frontiers in Cryobiology. Croatia: InTech, 2012; pp. 79100.Google Scholar
Slabbert, M, du Plessis, SS, Huyser, C. Large volume cryoprotectant-free vitrification: an alternative to conventional cryopreservation for human spermatozoa. Andrologia 2015;47:594–9.Google Scholar
Katkov, II, Bolyukh, VF, Chernetsov, OA, et al. Kinetic vitrification of spermatozoa of vertebrates: what can we learn from nature? In: Katkov, II, ed. Frontiers in Cryobiology. Croatia: InTech, 2012; pp. 340.Google Scholar
Wyns, C, Abou-Ghannam, G, Poels, J. Vitrification of human testicular tissue, spermatogonia, and spermatozoa. In: Tucker, MJ, Liebermann, J, eds. Vitrification in Assisted Reproduction, 2nd ed. Boca Raton, FL: CRC Press, 2015; pp. 197202.Google Scholar
Mohaqiq, M, Movahedin, M, Mazaheri, Z, Amirjannati, N. In vitro transplantation of spermatogonial stem cells isolated from human frozen–thawed testis tissue can induce spermatogenesis under 3-dimensional tissue culture conditions. Biol Res 2019;52:1625.Google Scholar
Sherman, JK. Freezing and freeze-drying of human spermatozoa. Fertil Steril 1954;5:357–71.Google Scholar
Loi, P, Iuso, D, Czernik, M, Zacchini, F, Ptak, G. Towards storage of cells and gametes in dry form. Trends Biotechnol 2013;31:688–95.Google Scholar
Gianaroli, L, Magli, MC, Stanghellini, I, et al. DNA integrity is maintained after freeze-drying of human spermatozoa. Fertil Steril 2012;97:1067–73.Google Scholar
Gil, L, Olaciregui, M, Luno, V, Maio, C, Gonzalez, N, Martinez, F. Current status of freeze-drying technology to preserve domestic animals sperm. Reprod Domest Anim 2014;49:7281.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
×