Skip to main content
Log in

The effect of human sperm chromatin maturity on ICSI outcomes

  • Research Article
  • Published:
Human Cell Aims and scope Submit manuscript

Abstract

Because sperm chromatin may play a key role in reproductive success, we verify the associations between sperm chromatin abnormalities, embryo development and the ability to achieve pregnancy. The evaluation of sperm chromatin maturity using aniline blue (AB), chromomycin A3 (CMA3) and toluidine blue (TB) staining were carried out in group of males from infertile couples that underwent ICSI. Low levels of sperm chromatin abnormalities (< 16%) were found in most subjects (> 50%). A higher percentage of TB-positive sperm cells were discovered in the men from couples who achieved ≤ 50% fertilized oocytes compared to men who achieved > 50%. No significant differences were discovered by the applied tests between the men from couples who achieved ≤ 50% and those who achieved > 50% high-quality embryos on the 3rd or 5th day after fertilization, nor between the men from couples who achieved pregnancy and those who failed. The sperm chromatin maturity did not correlate with the ICSI results. However, the ROC analysis revealed a significant predictive value of TB-positive spermatozoa only for fertilization. Therefore, the TB assay can be considered as a useful test for the prediction of fertilization. Our findings suggest that the level of sperm chromatin abnormalities of the examined men was not clinically significant. No found associations between sperm chromatin maturity and embryo development and the ability to achieve pregnancy. We could not exclude the effects of the repairing processes in the fertilized oocyte. The use of complementary tests that verify the status of the sperm chromatin seems justified.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2

Similar content being viewed by others

References

  1. Bungum M, Bungum L, Giwercman A. Sperm chromatin structure assay (SCSA): a tool in diagnosis and treatment of infertility. Asian J Androl. 2011;13:69–75. https://doi.org/10.1038/aja.2010.73.

    Article  PubMed  CAS  Google Scholar 

  2. Esteves SC. Novel concepts in male factor infertility: clinical and laboratory perspectives. J Assist Reprod Genet. 2016;33:1319–35. https://doi.org/10.1007/s10815-016-0763-8.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Esteves SC, Miyaoka R, Agarwal A. An update on the clinical assessment of the infertile male. Clinics (Sao Paulo). 2011;66:691–700. https://doi.org/10.1590/s1807-59322011000400026.

    Article  Google Scholar 

  4. Tahmasbpour E, Balasubramanian D, Agarwal A. A multi-faceted approach to understanding male infertility: gene mutations, molecular defects and assisted reproductive techniques (ART). J Assist Reprod Genet. 2014;31:1115–37. https://doi.org/10.1007/s10815-014-0280-6.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Wdowiak A, Wdowiak A, Bakalczuk S, Bakalczuk G. Relationship between alcohol consumption and a sperm nuclear DNA fragmentation and pregnancy. Postep Androl Online. 2016;3:14–21.

    Google Scholar 

  6. Aydos OS, Yükselten Y, Kaplan F, Sunguroğlu A, Aydos K. Analysis of the correlation between sperm DNA integrity and conventional semen parameters in infertile men. Turk J Urol. 2015;241:191–7. https://doi.org/10.5152/tud.2015.98475.

    Article  Google Scholar 

  7. Fernandez-Encinas A, García-Peiró A, Ribas-Maynou J, et al. Characterization of nuclease activity in human seminal plasma and its relationship to semen parameters, sperm DNA fragmentation and male infertility. J Urol. 2016;195:213–9. https://doi.org/10.1016/j.juro.2015.07.089.

    Article  PubMed  CAS  Google Scholar 

  8. Omran HM, Bakhiet M, Dashti MG. DNA integrity is a critical molecular indicator for the assessment of male infertility. Mol Med Rep. 2013;7:1631–5. https://doi.org/10.3892/mmr.2013.1390.

    Article  PubMed  CAS  Google Scholar 

  9. 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. https://doi.org/10.1016/j.fertnstert.2010.09.029.

    Article  PubMed  Google Scholar 

  10. Avendaño C, Franchi A, Duran H, Oehninger S. DNA fragmentation of normal spermatozoa negatively impacts embryo quality and intracytoplasmic sperm injection outcome. Fertil Steril. 2010;94:549–55. https://doi.org/10.1016/j.fertnstert.2009.02.050.

    Article  PubMed  Google Scholar 

  11. Agarwal A, Majzoub A, Esteves SC, Ko E, Ramasamy R, Zini A. Clinical utility of sperm DNA fragmentation testing: practice recommendations based on clinical scenarios. Transl Androl Urol. 2016;5:935–50. https://doi.org/10.21037/tau.2016.10.03.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Bieniek JM, Drabovich AP, Lo KC. Seminal biomarkers for the evaluation of male infertility. Asian J Androl. 2016;18:426–33. https://doi.org/10.4103/1008-682x.175781.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. van der Horst G, du Plessis S. Not just the marriage of Figaro: but the marriage of WHO/ESHRE semen analysis criteria with sperm functionality. Postep Androl Online. 2017;4(1):6–21.

    Google Scholar 

  14. Iranpour FG. Impact of sperm chromatin evaluation on fertilization rate in intracytoplasmic sperm injection. Adv Biomed Res. 2013;3:229. https://doi.org/10.4103/2277-9175.145719.

    Article  CAS  Google Scholar 

  15. Rubino P, Viganò P, Luddi A, Piomboni P. The ICSI procedure from past to future: a systematic review of the more controversial aspects. Hum Reprod Update. 2016;22:194–227. https://doi.org/10.1093/humupd/dmv050.

    Article  PubMed  Google Scholar 

  16. Sakkas D, Ramalingam M, Garrido N, Barratt CL. Sperm selection in natural conception: what can we learn from Mother Nature to improve assisted reproduction outcomes? Hum Reprod Update. 2015;21:711–26. https://doi.org/10.1093/humupd/dmv042.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Utsuno H, Miyamoto T, Oka K, Shiozawa T. Morphological alterations in protamine-deficient spermatozoa. Hum Reprod. 2014;29:2374–81. https://doi.org/10.1093/humrep/deu225.

    Article  PubMed  CAS  Google Scholar 

  18. Bounartzi T, Dafopoulos K, Anifandis G, et al. Pregnancy prediction by free sperm DNA and sperm DNA fragmentation in semen specimens of IVF/ICSI-ET patients. Hum Fertil (Camb). 2016;19:56–62. https://doi.org/10.3109/14647273.2016.1157629.

    Article  CAS  Google Scholar 

  19. Irez T, Sahmay S, Ocal P, et al. Investigation of the association between the outcomes of sperm chromatin condensation and decondensation tests, and assisted reproduction techniques. Andrologia. 2015;47:438–47. https://doi.org/10.1111/and.12286.

    Article  PubMed  CAS  Google Scholar 

  20. Simon L, Murphy K, Shamsi MB, et al. Paternal influence of sperm DNA integrity on early embryonic development. Hum Reprod. 2014;29:2402–12. https://doi.org/10.1093/humrep/deu228.

    Article  PubMed  CAS  Google Scholar 

  21. 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. https://doi.org/10.1093/humrep/des261.

    Article  PubMed  CAS  Google Scholar 

  22. Talebi AR, Fesahat F, Mangoli E, Ghasemzadeh J, Nayeri M, Sadeghian-Nodoshan F. Relationship between sperm protamine deficiency and apoptosis in couples with unexplained repeated spontaneous abortions. Int J Reprod Biomed (Yazd). 2016;14:199–204.

    Article  CAS  Google Scholar 

  23. Asmarinah, Syauqy A, Umar LA, et al. Sperm chromatin maturity and integrity correlated to zygote development in ICSI program. Syst Biol Reprod Med. 2016;62:309–16. https://doi.org/10.1080/19396368.2016.1210695.

    Article  PubMed  CAS  Google Scholar 

  24. Carrell DT, Liu L, Peterson CM, et al. Sperm DNA fragmentation is increased in couples with unexplained recurrent pregnancy loss. Arch Androl. 2003;49:49–55.

    Article  PubMed  CAS  Google Scholar 

  25. Ioannou D, Miller D, Griffin DK, Tempest HG. Impact of sperm DNA chromatin in the clinic. J Assist Reprod Genet. 2016;33:157–66. https://doi.org/10.1007/s10815-015-0624-x.

    Article  PubMed  Google Scholar 

  26. Kazerooni T, Asadi N, Jadid L, et al. Evaluation of sperm’s chromatin quality with acridine orange test, chromomycin A3 and aniline blue staining in couples with unexplained recurrent abortion. J Assist Reprod Genet. 2009;26:591–6. https://doi.org/10.1007/s10815-009-9361-3.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Leach M, Aitken RJ, Sacks G. Sperm DNA fragmentation abnormalities in men from couples with a history of recurrent miscarriage. Aust N Z J Obstet Gynaecol. 2015;55:379–83. https://doi.org/10.1111/ajo.12373.

    Article  PubMed  Google Scholar 

  28. Sadeghi MR, Hodjat M, Lakpour N, Arefi S, Amirjannati N, Modarresi T. Effects of sperm chromatin integrity on fertilization rate and embryo quality following intracytoplasmic sperm injection. AJMB. 2009;1:173–80.

    PubMed  PubMed Central  Google Scholar 

  29. Talebi AR, Vahidi S, Aflatoonian A, et al. Cytochemical evaluation of sperm chromatin and DNA integrity in couples with unexplained recurrent spontaneous abortions. Andrologia. 2012;44:462–70. https://doi.org/10.1111/j.1439-0272.2011.01206.x.

    Article  PubMed  CAS  Google Scholar 

  30. Tarozzi N, Nadalini M, Stronati A, et al. Anomalies in sperm chromatin packaging: implications for assisted reproduction techniques. Reprod Biomed Online. 2009;18:486–95.

    Article  PubMed  Google Scholar 

  31. Tavalaee M, Deemeh MR, Arbabian M, Kiyani A, Nasr-Esfahani MH. Relationship between fertilization rate and early apoptosis in sperm population of infertile individuals. Andrologia. 2012;46:36–41. https://doi.org/10.1111/and.12038.

    Article  PubMed  CAS  Google Scholar 

  32. Bach PV, Schlegel PN. Sperm DNA damage and its role in IVF and ICSI. Basic Clin Androl. 2016. https://doi.org/10.1186/s12610-016-0043-6.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Bronet F, Martínez E, Gaytán M, et al. Sperm DNA fragmentation index does not correlate with the sperm or embryo aneuploidy rate in recurrent miscarriage or implantation failure patients. Hum Reprod. 2012;27:1922–9. https://doi.org/10.1093/humrep/des148.

    Article  PubMed  CAS  Google Scholar 

  34. Coughlan C, Clarke H, Cutting R, et al. Sperm DNA fragmentation, recurrent implantation failure and recurrent miscarriage. Asian J Androl. 2015;17:681–5. https://doi.org/10.4103/1008-682X.144946.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Tavalaee M, Razavi S, Nasr-Esfahani MH. Influence of sperm chromatin anomalies on assisted reproductive technology outcome. Fertil Steril. 2009;91:1119–26. https://doi.org/10.1016/j.fertnstert.2008.01.063.

    Article  PubMed  CAS  Google Scholar 

  36. 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;32:17–26. https://doi.org/10.1007/s10815-014-0374-1.

    Article  PubMed  Google Scholar 

  37. Chan PJ, Orzylowska EM, Corselli JU, Jacobson JD, Wei AK. A simple sperm DNA toroid integrity test and risk of miscarriage. Biomed Res Int. 2015. https://doi.org/10.1155/2015/780983.

    Article  PubMed  PubMed Central  Google Scholar 

  38. World Health Organization. WHO Laboratory Manual for the Examination and Processing of Human Semen. 5th ed. Geneva: World Health Organization Press; 2010.

  39. Talebi AR, Khalili MA, Vahidi S, Ghasemzadeh J, Tabibnejad N. Sperm chromatin condensation, DNA integrity, and apoptosis in men with spinal cord injury. J Spinal Cord Med. 2013;36:140–6. https://doi.org/10.1179/2045772312y.0000000055.

    Article  PubMed  PubMed Central  Google Scholar 

  40. O’Flaherty CM, Chan PT, Hales BF, Robaire B. Sperm chromatin structure components are differentially repaired in cancer survivors. J Androl. 2012;33:629–36. https://doi.org/10.2164/jandrol.111.015388.

    Article  PubMed  Google Scholar 

  41. Scott L, Alvero R, Leondires M, Miller B. The morphology of human pronuclear embryos is positively related to blastocyst development and implantation. Hum Reprod. 2000;15:2394–403. https://doi.org/10.1093/humrep/15.11.2394.

    Article  PubMed  CAS  Google Scholar 

  42. Gardner DK, Lane M, Stevens J, Schlenker T, Schoolcraft WB. Blastocyst score affects implantation and pregnancy outcome: towards a single blastocyst transfer. Fertil Steril. 2000;73:1155–8.

    Article  PubMed  CAS  Google Scholar 

  43. Bączkowski T, Kurzawa R, Głąbowski W. Methods of embryo scoring in in vitro fertilization. Reprod Biol. 2004;4:5–22.

    PubMed  Google Scholar 

  44. Heitmann RJ, Hill MJ, Richter KS, DeCherney AH, Widra EA. The simplified SART embryo scoring system is highly correlated to implantation and live birth in single blastocyst transfers. J Assist Reprod Genet. 2013;4:563–7. https://doi.org/10.1007/s10815-013-9932-1.

    Article  Google Scholar 

  45. Tandara M, Bajić A, Tandara L, et al. Sperm DNA integrity testing: big halo is a good predictor of embryo quality and pregnancy after conventional IVF. Andrology. 2014;2:678–86. https://doi.org/10.1111/j.2047-2927.2014.00234.x.

    Article  PubMed  CAS  Google Scholar 

  46. Razavi S, Nasr-Esfahani MH, Mardani M, Mafi A, Moghdam A. Effect of human sperm chromatin anomalies on fertilization outcome post-ICSI. Andrologia. 2003;35:238–43. https://doi.org/10.1046/j.1439-0272.2003.00566.

    Article  PubMed  CAS  Google Scholar 

  47. Carrell DT, Emery BR, Hammoud S. Altered protamine expression and diminished spermatogenesis: what is the link? Hum Reprod Updat. 2007;13:313–27. https://doi.org/10.1111/j.2047-2927.2014.00234.x.

    Article  CAS  Google Scholar 

  48. Francis S, Yelumalai S, Jones C, Coward K. Aberrant protamine content in sperm and consequential implications for infertility treatment. Hum Fertil (Camb). 2014;17:80–9. https://doi.org/10.3109/14647273.2014.915347.

    Article  Google Scholar 

  49. Oliva R, Castillo J. Proteomics and genetics of sperm chromatin condensation. Asian J Androl. 2011;13:24–30. https://doi.org/10.1038/aja.2010.65.

    Article  PubMed  Google Scholar 

  50. Ward WS. Function of sperm chromatin structural elements in fertilization and development. Mol Hum Reprod. 2010;16:30–6. https://doi.org/10.1093/molehr/gap080.

    Article  PubMed  CAS  Google Scholar 

  51. Agarwal A, Cho CL, Majzoub A, Esteves SC. The Society for Translational Medicine: clinical practice guidelines for sperm DNA fragmentation testing in male infertility. Transl Androl Urol. 2017;6(Suppl 4):720–33. https://doi.org/10.21037/tau.2017.08.06.

    Article  Google Scholar 

  52. Evenson DP. Evaluation of sperm chromatin structure and DNA strand breaks is an important part of clinical male fertility assessment. Transl Androl Urol. 2017;6:495. https://doi.org/10.21037/tau.2017.07.20.

    Article  Google Scholar 

  53. Piasecka M, Gaczarzewicz D, Laszczyńska M, Starczewski A, Brodowska A. Flow cytometry application in the assessment of sperm DNA integrity of men with asthenozoospermia. Folia Histochem Cytobiol. 2007;45(1):127–36.

    CAS  Google Scholar 

  54. Simon L, Emery BR, Carrell DT. Review: impact of sperm DNA damage in assisted reproduction. Best Pract Res Clin Obstet Gynaecol. https://doi.org/10.1016/j.bpobgyn.2017.07.003 (Published online ahead of print 24 July 2017).

  55. Wiweko B, Utami P. Predictive value of sperm deoxyribonucleic acid (DNA) fragmentation index in male infertility. Basic Clin Androl. 2017;27:1. https://doi.org/10.1186/s12610-016-0046-3.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Zheng WW, Song G, Wang QL, et al. Sperm DNA damage has a negative effect on early embryonic development following in vitro fertilization. Asian J Androl. 2017; https://doi.org/10.4103/aja.aja_19_17 (Published online ahead of print Jun 30).

  57. Tsarev I, Bungum M, Giwercman A, et al. Evaluation of male fertility potential by Toluidine Blue test for sperm chromatin structure assessment. Hum Reprod. 2009;24:1569–74. https://doi.org/10.1093/humrep/dep068.

    Article  PubMed  CAS  Google Scholar 

  58. Nasr-Esfahani MH, Razavi S, Mardani M. Relation between different human sperm nuclear maturity tests and in vitro fertilization. J Assist Reprod Genet. 2001;18:219–25.

    Article  PubMed  CAS  Google Scholar 

  59. Karydis S, Asimakopoulos B, Papadopoulos N, Vakalopoulos I, Al-Hasani S, Nikolettos N. ICSI outcome is not associated with the incidence of spermatozoa with abnormal chromatin condensation. In Vivo. 2005;19:921–5.

    PubMed  Google Scholar 

  60. Sadeghi MR, Lakpour N, Heidari-Vala H, et al. Relationship between sperm chromatin status and ICSI outcome in men with obstructive azoospermia and unexplained infertile normozoospermia. Rom J Morphol Embryol. 2011;52:645–51.

    PubMed  CAS  Google Scholar 

  61. Tavares RS, Silva AF, Lourenço B, Almeida-Santos T, Sousa AP, Ramalho-Santos J. Evaluation of human sperm chromatin status after selection using a modified Diff-Quik stain indicates embryo quality and pregnancy outcomes following in vitro fertilization. Andrology. 2013;1:830–7. https://doi.org/10.1111/j.2047-2927.2013.00127.x.

    Article  PubMed  CAS  Google Scholar 

  62. 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;19:80–90. https://doi.org/10.4103/1008-682x.182822.

    Article  PubMed  Google Scholar 

  63. Agarwal A, Cho CL, Esteves SC. Should we evaluate and treat sperm DNA fragmentation? Curr Opin Obstet Gynecol. 2016;28:164–71. https://doi.org/10.1097/gco.0000000000000271.

    Article  PubMed  Google Scholar 

  64. 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. https://doi.org/10.1371/journal.pone.0165125.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  65. Ward SW. Eight tests for sperm DNA fragmentation and their roles in the clinic. Transl Androl Urol. 2017. https://doi.org/10.21037/tau.2017.03.78 (in press, Published online ahead of print).

  66. Nasr-Esfahani MH, Razavi S, Mozdarani H, Mardani M, Azvagi H. Relationship between protamine deficiency with fertilization rate and incidence of sperm premature chromosomal condensation post-ICSI. Andrologia. 2004;36:95. https://doi.org/10.1111/j.1439-0272.2004.00612.x.

    Article  PubMed  CAS  Google Scholar 

  67. Ribas-Maynou J, García-Peiró A, Fernández-Encinas A, et al. Comprehensive analysis of sperm DNA fragmentation by five different assays: TUNEL assay, SCSA, SCD test and alkaline and neutral Comet assay. Andrology. 2013;1:715–22. https://doi.org/10.1111/j.2047-2927.2013.00111.x.

    Article  PubMed  CAS  Google Scholar 

  68. Borini A, Tarozzi N, Bizzaro D, Bonu MA, Fava L, Flamigni C, Coticchio G. Sperm DNA fragmentation: paternal effect on early post-implantation embryo development in ART. Hum Reprod. 2006;21:2876–81. https://doi.org/10.1093/humrep/del251.

    Article  PubMed  CAS  Google Scholar 

  69. Ozmen B, Caglar GS, Koster F, Schopper B, Diedrich K, Al-Hasani S. Relationship between sperm DNA damage, induced acrosome reaction and viability in ICSI patients. Reprod Biomed Online. 2007;15:208–14.

    Article  PubMed  CAS  Google Scholar 

  70. Nasr-Esfahani MH, Salehi M, Razavi S, Mardani M, Bahramian H, Steger K, Oreizi F. Effect of protamine-2 deficiency on ICSI outcome. Reprod Biomed Online. 2004;9:652–8.

    Article  PubMed  CAS  Google Scholar 

  71. Nasr-Esfahani MH, Salehi M, Razavi S, et al. Effect of sperm DNA damage and sperm protamine deficiency on fertilization and embryo development post-ICSI. Reprod Biomed Online. 2005;11:198–205.

    Article  PubMed  CAS  Google Scholar 

  72. Nasr-Esfahani MH, Razavi S, Tavalaee M. Failed fertilization after ICSI and spermiogenic defects. Fertil Steril. 2008;89:892–8. https://doi.org/10.1016/j.fertnstert.2007.04.012.

    Article  PubMed  Google Scholar 

  73. Lord T, Aitken RJ. Fertilization stimulates 8-hydroxy-2′-deoxyguanosine repair and antioxidant activity to prevent mutagenesis in the embryo. Dev Biol. 2015;406:1–13. https://doi.org/10.1016/j.ydbio.2015.07.024.

    Article  PubMed  CAS  Google Scholar 

  74. Ménézo Y, Dale B, Cohen M. DNA damage and repair in human oocytes and embryos: a review. Zygote. 2010;18:357–65. https://doi.org/10.1017/s0967199410000286.

    Article  PubMed  Google Scholar 

  75. Perry M. Chemically induced DNA damage and sperm and oocyte repair machinery: the story gets more interesting. Asian J Androl. 2015. https://doi.org/10.4103/1008-682x.156118 (Published online ahead of print).

    Article  PubMed  Google Scholar 

  76. Tesarik J, Greco E, Mendoza C. Late, but not early, paternal effect on human embryo development is related to sperm DNA fragmentation. Human Reprod. 2004;19:611–5. https://doi.org/10.1093/humrep/deh127.

    Article  CAS  Google Scholar 

  77. Tesarik J. Paternal effects on cell division in the human preimplantation embryo. Reprod Biomed Online. 2005;10:370–5.

    Article  PubMed  Google Scholar 

  78. Aitken RJ, De Iuliis GN. Origins and consequences of DNA damage in male germ cells. Reprod Biomed Online. 2007;14:727–33.

    Article  PubMed  CAS  Google Scholar 

  79. Gavriliouk D, Aitken RJ. Damage to sperm DNA mediated by reactive oxygen species: its impact on human reproduction and the health trajectory of offspring. Adv Exp Med Biol. 2015;868:23–47. https://doi.org/10.1007/978-3-319-18881-2_2.

    Article  PubMed  Google Scholar 

  80. Bohrer RC, Coutinho AR, Duggavathi R, Bordignon V. The incidence of DNA double-strand breaks is higher in late-cleaving and less developmentally competent porcine embryos. Biol Reprod. 2015;93:59. https://doi.org/10.1095/biolreprod.115.130542.

    Article  PubMed  CAS  Google Scholar 

  81. He M, Tan L. Correlation between sperm ultrastructure in infertile patients with abnormal sperm morphology and DNA damage. Genet Mol Res. 2015;14:17000–6. https://doi.org/10.4238/2015.december.15.6.

    Article  PubMed  CAS  Google Scholar 

  82. 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. https://doi.org/10.1093/molehr/gat025.

    Article  PubMed  CAS  Google Scholar 

  83. Dada R, Kumar M, Jesudasan R, Fernández JL, Gosálvez J, Agarwal A. Epigenetics and its role in male infertility. J Assist Reprod Genet. 2012;29:213–23. https://doi.org/10.1007/s10815-012-9715-0.

    Article  PubMed  PubMed Central  Google Scholar 

  84. Simon L, Liu L, Murphy K, et al. Comparative analysis of three sperm DNA damage assays and sperm nuclear protein content in couples undergoing assisted reproduction treatment. Hum Reprod. 2014;29:904–17. https://doi.org/10.1093/humrep/deu040.

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

This study was supported by the Pomeranian Medical University, Szczecin, Poland Grants No. WNoZ-322-04/S/2016 and No. FSN-322-5/2016.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Malgorzata Piasecka.

Ethics declarations

Conflict of interest

The authors declare there is no conflict of interest that could be perceived as prejudicing the impartiality of the research reported.

Electronic supplementary material

Below is the link to the electronic supplementary material.

13577_2018_203_MOESM1_ESM.tif

Supplemental Fig. 1a–c. Non-invasive morphological evaluation of human embryo after intracytoplasmic sperm injection. Fertilized oocyte assessed within the first 24 h (a), embryo development on the 3rd day (b) and on the 5th day (c). The embryo was rated as follows: 2PN – zygote with visible female and male pronuclei containing nucleoli; 8A1 – a top-quality embryo with eight symmetrical blastomeres without cytoplasmic defects and fragmentation (according to Bączkowski et al. [43]); AA – a top-quality embryo with blastocoel filling and a well-developed inner cell mass and trophectoderm (according to Gardner et al. [42]). Original microphotographs obtained from infertile couples treated with ICSI; scale bar = 25 µm. (TIFF 3731 kb)

13577_2018_203_MOESM2_ESM.tif

Supplemental Fig. 2. Dynamics of the chromatin-remodeling process during spermatogenesis and epididymal sperm maturation. Chromatin remodeling is associated with chromosome reorganization, compaction and stabilization. Its condensation is mediated by protamination, characterized by the massive incorporation of protamines into the minor groove of DNA. Protamination is a complex process and is involved in the replacement of the following: the somatic histones by testicular-specific histones (Ht) (1), Ht by transition proteins (TP) (2) and TP by protamines (3). The chemical modifications of histone peptides and the formation of DNA strand breaks by topoisomerase facilitate the incorporation of these proteins to the DNA strand. Stabilization of the chromatin structure is mediated by the binding of zinc ions to the protamines at the end of the spermiogenesis (4). The depletion of these ions from the protamines takes place during epididymal sperm maturation (5). The mature sperm chromatin contains ≤ 15% of testicular histones. Asterisks indicate cytochemical tests that have been applied to reveal nuclear sperm compaction abnormalities at the different stages of nuclear remodeling; aniline blue staining (AB test) is able to detect sperm cells with excessive presence of histones, the TUNEL test (terminal deoxynucleotidyl transferase dUTP nick end labeling) reveals persisting DNA strand breaks, chromomycin A3 staining (CMA3 test) is used to display spermatozoa with protamine deficiency, and toluidine blue (TB test) allows for the discrimination between sperm cells with immature chromatin structure and normal chromatin. According to Carrell et al. [47], Ward [50], Oliva and Castillo [49] and Francis et al. [48] with own modifications. (TIFF 39105 kb)

13577_2018_203_MOESM3_ESM.tif

Supplemental Fig. 3. Predicted embryo development after fertilization with DNA-damaged sperm cell (details in the text). BER – base excision repair, NER – nucleotide excision repair, MMR – DNA mismatch repair, TDP 1 and 2 – tyrosyl-DNA phosphodiesterase 1 and 2. (TIFF 42007 kb)

Supplementary material 4 (DOCX 14 kb)

Supplementary material 5 (DOCX 38 kb)

Supplementary material 6 (DOCX 37 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gill, K., Rosiak, A., Gaczarzewicz, D. et al. The effect of human sperm chromatin maturity on ICSI outcomes. Human Cell 31, 220–231 (2018). https://doi.org/10.1007/s13577-018-0203-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13577-018-0203-4

Keywords

Navigation