Skip to main content
Top
Published in: Journal of Assisted Reproduction and Genetics 2/2020

01-02-2020 | Infertility | Gamete Biology

Decreased expression of MRE11 and RAD50 in testes from humans with spermatogenic failure

Authors: Minhao Hu, Lejun Li, Shuyuan Liu, Yiyun Lou, Liya Wang, Fang Le, Hongping Li, Qijing Wang, Hangying Lou, Ning Wang, Fan Jin

Published in: Journal of Assisted Reproduction and Genetics | Issue 2/2020

Login to get access

Abstract

Purpose

To assess testicular mRNA and protein expression levels of MRE11 and RAD50 in human azoospermia patients.

Methods

Patients diagnosed with maturation arrest at the spermatocyte stage (MA) and Sertoli cell-only syndrome (SCOS) were recruited through diagnostic testicular biopsy. Patients with normal spermatogenesis were studied as controls. In addition, knockdown of MRE11 and RAD50 was performed in GC-2spd(ts) cells to investigate their roles in cellular proliferation and apoptosis.

Results

mRNA and protein expression levels of MRE11 and RAD50 were measured using quantitative polymerase chain reaction, western blotting, and immunohistochemistry, respectively. Knockdown of both MRE11 and RAD50 utilized transfection with small interfering RNAs.

Conclusion

Our findings demonstrated altered expression levels of MRE11 and RAD50 in human testes with MA and SCOS, and showed that these alterations might be associated with impaired spermatogenesis. These results offer valuable new perspectives into the molecular mechanisms of male infertility.
Appendix
Available only for authorised users
Literature
1.
go back to reference Boivin J, Bunting L, Collins JA, Nygren KG. International estimates of infertility prevalence and treatment-seeking: potential need and demand for infertility medical care. Hum Reprod. 2007;22:1506–12.PubMed Boivin J, Bunting L, Collins JA, Nygren KG. International estimates of infertility prevalence and treatment-seeking: potential need and demand for infertility medical care. Hum Reprod. 2007;22:1506–12.PubMed
2.
go back to reference Massart A, Lissens W, Tournaye H, Stouffs K. Genetic causes of spermatogenic failure. Asian J Androl. 2012;14:40–8.PubMed Massart A, Lissens W, Tournaye H, Stouffs K. Genetic causes of spermatogenic failure. Asian J Androl. 2012;14:40–8.PubMed
3.
go back to reference Matzuk MM, Lamb DJ. The biology of infertility: research advances and clinical challenges. Nat Med. 2008;14:1197–213.PubMedPubMedCentral Matzuk MM, Lamb DJ. The biology of infertility: research advances and clinical challenges. Nat Med. 2008;14:1197–213.PubMedPubMedCentral
4.
go back to reference Zhu Z, Li C, Yang S, Tian R, Wang J, Yuan Q, et al. Dynamics of the transcriptome during human spermatogenesis: predicting the potential key genes regulating male gametes generation. Sci Rep. 2016;6:19069.PubMedPubMedCentral Zhu Z, Li C, Yang S, Tian R, Wang J, Yuan Q, et al. Dynamics of the transcriptome during human spermatogenesis: predicting the potential key genes regulating male gametes generation. Sci Rep. 2016;6:19069.PubMedPubMedCentral
5.
go back to reference Wong WY, Thomas CM, Merkus JM, Zielhuis GA, Steegers-Theunissen RP. Male factor subfertility: possible causes and the impact of nutritional factors. Fertil Steril. 2000;73:435–42.PubMed Wong WY, Thomas CM, Merkus JM, Zielhuis GA, Steegers-Theunissen RP. Male factor subfertility: possible causes and the impact of nutritional factors. Fertil Steril. 2000;73:435–42.PubMed
6.
go back to reference Cannarella R, Condorelli RA, Duca Y, La Vignera S, Calogero AE. New insights into the genetics of spermatogenic failure: a review of the literature. Hum Genet. 2019;138:125–40.PubMed Cannarella R, Condorelli RA, Duca Y, La Vignera S, Calogero AE. New insights into the genetics of spermatogenic failure: a review of the literature. Hum Genet. 2019;138:125–40.PubMed
7.
go back to reference Schultz N, Hamra FK, Garbers DL. A multitude of genes expressed solely in meiotic or postmeiotic spermatogenic cells offers a myriad of contraceptive targets. Proc Natl Acad Sci U S A. 2003;100:12201–6.PubMedPubMedCentral Schultz N, Hamra FK, Garbers DL. A multitude of genes expressed solely in meiotic or postmeiotic spermatogenic cells offers a myriad of contraceptive targets. Proc Natl Acad Sci U S A. 2003;100:12201–6.PubMedPubMedCentral
8.
go back to reference de Massy B. Initiation of meiotic recombination: how and where? Conservation and specificities among eukaryotes. Annu Rev Genet. 2013;47:563–99.PubMed de Massy B. Initiation of meiotic recombination: how and where? Conservation and specificities among eukaryotes. Annu Rev Genet. 2013;47:563–99.PubMed
9.
go back to reference Keeney S, Lange J, Mohibullah N. Self-organization of meiotic recombination initiation: general principles and molecular pathways. Annu Rev Genet. 2014;48:187–214.PubMedPubMedCentral Keeney S, Lange J, Mohibullah N. Self-organization of meiotic recombination initiation: general principles and molecular pathways. Annu Rev Genet. 2014;48:187–214.PubMedPubMedCentral
10.
go back to reference Lange J, Yamada S, Tischfield SE, Pan J, Kim S, Zhu X, et al. The landscape of mouse meiotic double-strand break formation, processing, and repair. Cell. 2016;167:695–708.PubMedPubMedCentral Lange J, Yamada S, Tischfield SE, Pan J, Kim S, Zhu X, et al. The landscape of mouse meiotic double-strand break formation, processing, and repair. Cell. 2016;167:695–708.PubMedPubMedCentral
11.
go back to reference Zakharyevich K, Tang S, Ma Y, Hunter N. Delineation of joint molecule resolution pathways in meiosis identifies a crossover-specific resolvase. Cell. 2012;149:334–47.PubMedPubMedCentral Zakharyevich K, Tang S, Ma Y, Hunter N. Delineation of joint molecule resolution pathways in meiosis identifies a crossover-specific resolvase. Cell. 2012;149:334–47.PubMedPubMedCentral
12.
go back to reference Handel MA, Schimenti JC. Genetics of mammalian meiosis: regulation, dynamics and impact on fertility. Nat Rev Genet. 2010;11:124–36.PubMed Handel MA, Schimenti JC. Genetics of mammalian meiosis: regulation, dynamics and impact on fertility. Nat Rev Genet. 2010;11:124–36.PubMed
13.
go back to reference Gunes S, Al-Sadaan M, Agarwal A. Spermatogenesis, DNA damage and DNA repair mechanisms in male infertility. Reprod BioMed Online. 2015;31:309–19.PubMed Gunes S, Al-Sadaan M, Agarwal A. Spermatogenesis, DNA damage and DNA repair mechanisms in male infertility. Reprod BioMed Online. 2015;31:309–19.PubMed
14.
go back to reference Baker SM, Plug AW, Prolla TA, Bronner CE, Harris AC, Yao X, et al. Involvement of mouse Mlh1 in DNA mismatch repair and meiotic crossing over. Nat Genet. 1996;13:336–42.PubMed Baker SM, Plug AW, Prolla TA, Bronner CE, Harris AC, Yao X, et al. Involvement of mouse Mlh1 in DNA mismatch repair and meiotic crossing over. Nat Genet. 1996;13:336–42.PubMed
15.
go back to reference Barlow C, Liyanage M, Moens PB, Tarsounas M, Nagashima K, Brown K, et al. Atm deficiency results in severe meiotic disruption as early as leptonema of prophase I. Development. 1998;125:4007–17.PubMed Barlow C, Liyanage M, Moens PB, Tarsounas M, Nagashima K, Brown K, et al. Atm deficiency results in severe meiotic disruption as early as leptonema of prophase I. Development. 1998;125:4007–17.PubMed
16.
go back to reference Romanienko PJ, Camerini-Otero RD. The mouse Spo11 gene is required for meiotic chromosome synapsis. Mol Cell. 2000;6:975–87.PubMed Romanienko PJ, Camerini-Otero RD. The mouse Spo11 gene is required for meiotic chromosome synapsis. Mol Cell. 2000;6:975–87.PubMed
17.
go back to reference Kuznetsov S, Pellegrini M, Shuda K, Fernandez-Capetillo O, Liu Y, Martin BK, et al. RAD51C deficiency in mice results in early prophase I arrest in males and sister chromatid separation at metaphase II in females. J Cell Biol. 2007;176:581–92.PubMedPubMedCentral Kuznetsov S, Pellegrini M, Shuda K, Fernandez-Capetillo O, Liu Y, Martin BK, et al. RAD51C deficiency in mice results in early prophase I arrest in males and sister chromatid separation at metaphase II in females. J Cell Biol. 2007;176:581–92.PubMedPubMedCentral
18.
go back to reference Stracker TH, Theunissen JW, Morales M, Petrini JH. The Mre11 complex and the metabolism of chromosome breaks: the importance of communicating and holding things together. DNA Repair (Amst). 2004;3:845–54. Stracker TH, Theunissen JW, Morales M, Petrini JH. The Mre11 complex and the metabolism of chromosome breaks: the importance of communicating and holding things together. DNA Repair (Amst). 2004;3:845–54.
19.
go back to reference Borde V. The multiple roles of the Mre11 complex for meiotic recombination. Chromosom Res. 2007;15:551–63. Borde V. The multiple roles of the Mre11 complex for meiotic recombination. Chromosom Res. 2007;15:551–63.
20.
go back to reference Rupnik A, Lowndes NF, Grenon M. MRN and the race to the break. Chromosoma. 2010;119:115–35.PubMed Rupnik A, Lowndes NF, Grenon M. MRN and the race to the break. Chromosoma. 2010;119:115–35.PubMed
21.
go back to reference Lavin MF, Kozlov S, Gatei M, Kijas AW. ATM-dependent phosphorylation of all three members of the MRN complex: from sensor to adaptor. Biomolecules. 2015;5:2877–902.PubMedPubMedCentral Lavin MF, Kozlov S, Gatei M, Kijas AW. ATM-dependent phosphorylation of all three members of the MRN complex: from sensor to adaptor. Biomolecules. 2015;5:2877–902.PubMedPubMedCentral
22.
go back to reference de Jager M, Dronkert ML, Modesti M, Beerens CE, Kanaar R, van Gent DC. DNA-binding and strand-annealing activities of human Mre11: implications for its roles in DNA double-strand break repair pathways. Nucleic Acids Res. 2001;29:1317–25.PubMedPubMedCentral de Jager M, Dronkert ML, Modesti M, Beerens CE, Kanaar R, van Gent DC. DNA-binding and strand-annealing activities of human Mre11: implications for its roles in DNA double-strand break repair pathways. Nucleic Acids Res. 2001;29:1317–25.PubMedPubMedCentral
23.
go back to reference Hopfner KP, Karcher A, Craig L, Woo TT, Carney JP, Tainer JA. Structural biochemistry and interaction architecture of the DNA double-strand break repair Mre11 nuclease and Rad50-ATPase. Cell. 2001;105:473–85.PubMed Hopfner KP, Karcher A, Craig L, Woo TT, Carney JP, Tainer JA. Structural biochemistry and interaction architecture of the DNA double-strand break repair Mre11 nuclease and Rad50-ATPase. Cell. 2001;105:473–85.PubMed
24.
go back to reference Stewart GS, Maser RS, Stankovic T, Bressan DA, Kaplan MI, Jaspers NG, et al. The DNA double-strand break repair gene hMRE11 is mutated in individuals with an ataxia-telangiectasia-like disorder. Cell. 1999;99:577–87.PubMed Stewart GS, Maser RS, Stankovic T, Bressan DA, Kaplan MI, Jaspers NG, et al. The DNA double-strand break repair gene hMRE11 is mutated in individuals with an ataxia-telangiectasia-like disorder. Cell. 1999;99:577–87.PubMed
25.
go back to reference Waltes R, Kalb R, Gatei M, Kijas AW, Stumm M, Sobeck A, et al. Human RAD50 deficiency in a Nijmegen breakage syndrome-like disorder. Am J Hum Genet. 2009;84:605–16.PubMedPubMedCentral Waltes R, Kalb R, Gatei M, Kijas AW, Stumm M, Sobeck A, et al. Human RAD50 deficiency in a Nijmegen breakage syndrome-like disorder. Am J Hum Genet. 2009;84:605–16.PubMedPubMedCentral
26.
go back to reference Cherry SM, Adelman CA, Theunissen JW, Hassold TJ, Hunt PA, Petrini JH. The Mre11 complex influences DNA repair, synapsis, and crossing over in murine meiosis. Curr Biol. 2007;17:373–8.PubMedPubMedCentral Cherry SM, Adelman CA, Theunissen JW, Hassold TJ, Hunt PA, Petrini JH. The Mre11 complex influences DNA repair, synapsis, and crossing over in murine meiosis. Curr Biol. 2007;17:373–8.PubMedPubMedCentral
27.
go back to reference Bender CF, Sikes ML, Sullivan R, Huye LE, Le Beau MM, Roth DB, et al. Cancer predisposition and hematopoietic failure in Rad50(S/S) mice. Genes Dev. 2002;16:2237–51.PubMedPubMedCentral Bender CF, Sikes ML, Sullivan R, Huye LE, Le Beau MM, Roth DB, et al. Cancer predisposition and hematopoietic failure in Rad50(S/S) mice. Genes Dev. 2002;16:2237–51.PubMedPubMedCentral
28.
go back to reference Bonache S, Algaba F, Franco E, Bassas L, Larriba S. Altered gene expression signature of early stages of the germ line supports the pre-meiotic origin of human spermatogenic failure. Andrology. 2014;2:596–606.PubMed Bonache S, Algaba F, Franco E, Bassas L, Larriba S. Altered gene expression signature of early stages of the germ line supports the pre-meiotic origin of human spermatogenic failure. Andrology. 2014;2:596–606.PubMed
29.
go back to reference Hofmann MC, Hess RA, Goldberg E, Millan JL. Immortalized germ cells undergo meiosis in vitro. Proc Natl Acad Sci U S A. 1994;91:5533–7.PubMedPubMedCentral Hofmann MC, Hess RA, Goldberg E, Millan JL. Immortalized germ cells undergo meiosis in vitro. Proc Natl Acad Sci U S A. 1994;91:5533–7.PubMedPubMedCentral
30.
go back to reference Wang H, Wen L, Yuan Q, Sun M, Niu M, He Z. Establishment and applications of male germ cell and Sertoli cell lines. Reproduction. 2016;152:R31–40.PubMed Wang H, Wen L, Yuan Q, Sun M, Niu M, He Z. Establishment and applications of male germ cell and Sertoli cell lines. Reproduction. 2016;152:R31–40.PubMed
31.
go back to reference Le F, Wang LY, Wang N, Li L, Li LJ, Zheng YM, et al. In vitro fertilization alters growth and expression of Igf2/H19 and their epigenetic mechanisms in the liver and skeletal muscle of newborn and elder mice. Biol Reprod. 2013;88:75.PubMed Le F, Wang LY, Wang N, Li L, Li LJ, Zheng YM, et al. In vitro fertilization alters growth and expression of Igf2/H19 and their epigenetic mechanisms in the liver and skeletal muscle of newborn and elder mice. Biol Reprod. 2013;88:75.PubMed
32.
go back to reference Cheng YS, Kuo PL, Teng YN, Kuo TY, Chung CL, Lin YH, et al. Association of spermatogenic failure with decreased CDC25A expression in infertile men. Hum Reprod. 2006;21:2346–52.PubMed Cheng YS, Kuo PL, Teng YN, Kuo TY, Chung CL, Lin YH, et al. Association of spermatogenic failure with decreased CDC25A expression in infertile men. Hum Reprod. 2006;21:2346–52.PubMed
33.
go back to reference Schrader M, Muller-Tidow C, Ravnik S, Muller M, Schulze W, Diederichs S, et al. Cyclin A1 and gametogenesis in fertile and infertile patients: a potential new molecular diagnostic marker. Hum Reprod. 2002;17:2338–43.PubMed Schrader M, Muller-Tidow C, Ravnik S, Muller M, Schulze W, Diederichs S, et al. Cyclin A1 and gametogenesis in fertile and infertile patients: a potential new molecular diagnostic marker. Hum Reprod. 2002;17:2338–43.PubMed
34.
go back to reference Weikert S, Schrader M, Muller M, Schulze W, Krause H, Miller K. Expression levels of the inhibitor of apoptosis survivin in testes of patients with normal spermatogenesis and spermatogenic failure. Fertil Steril. 2005;83(Suppl 1):1100–5.PubMed Weikert S, Schrader M, Muller M, Schulze W, Krause H, Miller K. Expression levels of the inhibitor of apoptosis survivin in testes of patients with normal spermatogenesis and spermatogenic failure. Fertil Steril. 2005;83(Suppl 1):1100–5.PubMed
35.
go back to reference Jaiswal D, Trivedi S, Agrawal NK, Singh K. Dysregulation of apoptotic pathway candidate genes and proteins in infertile azoospermia patients. Fertil Steril. 2015;104:736–43.PubMed Jaiswal D, Trivedi S, Agrawal NK, Singh K. Dysregulation of apoptotic pathway candidate genes and proteins in infertile azoospermia patients. Fertil Steril. 2015;104:736–43.PubMed
36.
go back to reference Kuo PL, Wang ST, Lin YM, Lin YH, Teng YN, Hsu CC. Expression profiles of the DAZ gene family in human testis with and without spermatogenic failure. Fertil Steril. 2004;81:1034–40.PubMed Kuo PL, Wang ST, Lin YM, Lin YH, Teng YN, Hsu CC. Expression profiles of the DAZ gene family in human testis with and without spermatogenic failure. Fertil Steril. 2004;81:1034–40.PubMed
37.
go back to reference Li LJ, Zhang FB, Liu SY, Tian YH, Le F, Lou HY, et al. Decreased expression of SAM68 in human testes with spermatogenic defects. Fertil Steril. 2014;102:61–7.PubMed Li LJ, Zhang FB, Liu SY, Tian YH, Le F, Lou HY, et al. Decreased expression of SAM68 in human testes with spermatogenic defects. Fertil Steril. 2014;102:61–7.PubMed
38.
go back to reference Lardone MC, Parada-Bustamante A, Ebensperger M, Valdevenito R, Kakarieka E, Martinez D, et al. DAX-1 and DAX-1A expression in human testicular tissues with primary spermatogenic failure. Mol Hum Reprod. 2011;17:739–46.PubMed Lardone MC, Parada-Bustamante A, Ebensperger M, Valdevenito R, Kakarieka E, Martinez D, et al. DAX-1 and DAX-1A expression in human testicular tissues with primary spermatogenic failure. Mol Hum Reprod. 2011;17:739–46.PubMed
39.
go back to reference Parada-Bustamante A, Molina C, Valencia C, Florez M, Lardone MC, Argandona F, et al. Disturbed testicular expression of the estrogen-metabolizing enzymes CYP1A1 and COMT in infertile men with primary spermatogenic failure: possible negative implications on Sertoli cells. Andrology. 2017;5:486–94.PubMed Parada-Bustamante A, Molina C, Valencia C, Florez M, Lardone MC, Argandona F, et al. Disturbed testicular expression of the estrogen-metabolizing enzymes CYP1A1 and COMT in infertile men with primary spermatogenic failure: possible negative implications on Sertoli cells. Andrology. 2017;5:486–94.PubMed
40.
go back to reference Ferfouri F, Boitrelle F, Ghout I, Albert M, Molina GD, Wainer R, et al. A genome-wide DNA methylation study in azoospermia. Andrology. 2013;1:815–21.PubMed Ferfouri F, Boitrelle F, Ghout I, Albert M, Molina GD, Wainer R, et al. A genome-wide DNA methylation study in azoospermia. Andrology. 2013;1:815–21.PubMed
41.
go back to reference Ramasamy R, Ridgeway A, Lipshultz LI, Lamb DJ. Integrative DNA methylation and gene expression analysis identifies discoidin domain receptor 1 association with idiopathic nonobstructive azoospermia. Fertil Steril. 2014;102:968–73.PubMedPubMedCentral Ramasamy R, Ridgeway A, Lipshultz LI, Lamb DJ. Integrative DNA methylation and gene expression analysis identifies discoidin domain receptor 1 association with idiopathic nonobstructive azoospermia. Fertil Steril. 2014;102:968–73.PubMedPubMedCentral
42.
go back to reference Gabrielsen JS, Tanrikut C. Chronic exposures and male fertility: the impacts of environment, diet, and drug use on spermatogenesis. Andrology. 2016;4:648–61.PubMed Gabrielsen JS, Tanrikut C. Chronic exposures and male fertility: the impacts of environment, diet, and drug use on spermatogenesis. Andrology. 2016;4:648–61.PubMed
43.
go back to reference Neto FT, Bach PV, Najari BB, Li PS, Goldstein M. Spermatogenesis in humans and its affecting factors. Semin Cell Dev Biol. 2016;59:10–26.PubMed Neto FT, Bach PV, Najari BB, Li PS, Goldstein M. Spermatogenesis in humans and its affecting factors. Semin Cell Dev Biol. 2016;59:10–26.PubMed
44.
go back to reference Zhong ZH, Jiang WQ, Cesare AJ, Neumann AA, Wadhwa R, Reddel RR. Disruption of telomere maintenance by depletion of the MRE11/RAD50/NBS1 complex in cells that use alternative lengthening of telomeres. J Biol Chem. 2007;282:29314–22.PubMed Zhong ZH, Jiang WQ, Cesare AJ, Neumann AA, Wadhwa R, Reddel RR. Disruption of telomere maintenance by depletion of the MRE11/RAD50/NBS1 complex in cells that use alternative lengthening of telomeres. J Biol Chem. 2007;282:29314–22.PubMed
45.
go back to reference Kavitha CV, Choudhary B, Raghavan SC, Muniyappa K. Differential regulation of MRN (Mre11-Rad50-Nbs1) complex subunits and telomerase activity in cancer cells. Biochem Biophys Res Commun. 2010;399:575–80.PubMed Kavitha CV, Choudhary B, Raghavan SC, Muniyappa K. Differential regulation of MRN (Mre11-Rad50-Nbs1) complex subunits and telomerase activity in cancer cells. Biochem Biophys Res Commun. 2010;399:575–80.PubMed
46.
go back to reference Kim SK, Yoon YD, Park YS, Seo JT, Kim JH. Involvement of the Fas-Fas ligand system and active caspase-3 in abnormal apoptosis in human testes with maturation arrest and Sertoli cell-only syndrome. Fertil Steril. 2007;87:547–53.PubMed Kim SK, Yoon YD, Park YS, Seo JT, Kim JH. Involvement of the Fas-Fas ligand system and active caspase-3 in abnormal apoptosis in human testes with maturation arrest and Sertoli cell-only syndrome. Fertil Steril. 2007;87:547–53.PubMed
Metadata
Title
Decreased expression of MRE11 and RAD50 in testes from humans with spermatogenic failure
Authors
Minhao Hu
Lejun Li
Shuyuan Liu
Yiyun Lou
Liya Wang
Fang Le
Hongping Li
Qijing Wang
Hangying Lou
Ning Wang
Fan Jin
Publication date
01-02-2020
Publisher
Springer US
Published in
Journal of Assisted Reproduction and Genetics / Issue 2/2020
Print ISSN: 1058-0468
Electronic ISSN: 1573-7330
DOI
https://doi.org/10.1007/s10815-019-01686-5

Other articles of this Issue 2/2020

Journal of Assisted Reproduction and Genetics 2/2020 Go to the issue