Skip to main content
Erschienen in: Journal of Assisted Reproduction and Genetics 2/2019

31.10.2018 | Reproductive Physiology and Disease

Reactive oxygen species-induced alterations in H19-Igf2 methylation patterns, seminal plasma metabolites, and semen quality

verfasst von: Mahsa Darbandi, Sara Darbandi, Ashok Agarwal, Saradha Baskaran, Sulagna Dutta, Pallav Sengupta, Hamid Reza Khorram Khorshid, Sandro Esteves, Kambiz Gilany, Mehdi Hedayati, Fatemeh Nobakht, Mohammad Mehdi Akhondi, Niknam Lakpour, Mohammad Reza Sadeghi

Erschienen in: Journal of Assisted Reproduction and Genetics | Ausgabe 2/2019

Einloggen, um Zugang zu erhalten

Abstract

Purpose

This study was conducted in order to investigate the effects of reactive oxygen species (ROS) levels on the seminal plasma (SP) metabolite milieu and sperm dysfunction.

Methods

Semen specimens of 151 normozoospermic men were analyzed for ROS by chemiluminescence and classified according to seminal ROS levels [in relative light units (RLU)/s/106 sperm]: group 1 (n = 39): low (ROS < 20), group 2 (n = 38): mild (20 ≤ ROS < 40), group 3 (n = 31): moderate (40 ≤ ROS < 60), and group 4 (n = 43): high (ROS ≥ 60). A comprehensive analysis of SP and semen parameters, including conventional semen characteristics, measurement of total antioxidant capacity (TAC), sperm DNA fragmentation index (DFI), chromatin maturation index (CMI), H19-Igf2 methylation status, and untargeted seminal metabolic profiling using nuclear magnetic resonance spectroscopy (1H-NMR), was carried out.

Result(s)

The methylation status of H19 and Igf2 was significantly different in specimens with high ROS (P < 0.005). Metabolic fingerprinting of these SP samples showed upregulation of trimethylamine N-oxide (P < 0.001) and downregulations of tryptophan (P < 0.05) and tyrosine/tyrosol (P < 0.01). High ROS significantly reduced total sperm motility (P < 0.05), sperm concentration (P < 0.001), and seminal TAC (P < 0.001) but increased CMI and DFI (P < 0.005). ROS levels have a positive correlation with Igf2 methylation (r = 0.19, P < 0.05), DFI (r = 0.40, P < 0.001), CMI (r = 0.39, P < 0.001), and trimethylamine N-oxide (r = 0.45, P < 0.05) and a negative correlation with H19 methylation (r = − 0.20, P < 0.05), tryptophan (r = − 0.45, P < 0.05), sperm motility (r = − 0.20, P < 0.05), sperm viability (r = − 0.23, P < 0.01), and sperm concentration (r = − 0.30, P < 0.001).

Conclusion(s)

Results showed significant correlation between ROS levels and H19-Igf2 gene methylation as well as semen parameters. These findings are critical to identify idiopathic male infertility and its management through assisted reproduction technology (ART).
Anhänge
Nur mit Berechtigung zugänglich
Literatur
1.
Zurück zum Zitat Jarow JP, Sharlip ID, Belker AM, Lipshultz LI, Sigman M, Thomas AJ, et al. Best practice policies for male infertility. J Urol. 2002;167:2138–44.CrossRefPubMed Jarow JP, Sharlip ID, Belker AM, Lipshultz LI, Sigman M, Thomas AJ, et al. Best practice policies for male infertility. J Urol. 2002;167:2138–44.CrossRefPubMed
2.
Zurück zum Zitat Darbandi S, Darbandi M. Lifestyle modifications on further reproductive problems. Cresco J Reprod Sci. 2016;1:1–2. Darbandi S, Darbandi M. Lifestyle modifications on further reproductive problems. Cresco J Reprod Sci. 2016;1:1–2.
3.
Zurück zum Zitat Hamada A, Esteves SC, Nizza M, Agarwal A. Unexplained male infertility: diagnosis and management. Int Braz J Urol. 2012;38:576–94.CrossRefPubMed Hamada A, Esteves SC, Nizza M, Agarwal A. Unexplained male infertility: diagnosis and management. Int Braz J Urol. 2012;38:576–94.CrossRefPubMed
4.
Zurück zum Zitat Rajender S, Avery K, Agarwal A. Epigenetics, spermatogenesis and male infertility. Mutat Res-Rev Mutat. 2011;727:62–71.CrossRef Rajender S, Avery K, Agarwal A. Epigenetics, spermatogenesis and male infertility. Mutat Res-Rev Mutat. 2011;727:62–71.CrossRef
6.
Zurück zum Zitat Kovac JR, Pastuszak AW, Lamb DJ. The use of genomics, proteomics, and metabolomics in identifying biomarkers of male infertility. Fertil Steril. 2013;99:998–1007.CrossRefPubMedPubMedCentral Kovac JR, Pastuszak AW, Lamb DJ. The use of genomics, proteomics, and metabolomics in identifying biomarkers of male infertility. Fertil Steril. 2013;99:998–1007.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Gannon JR, Emery BR, Jenkins TG, Carrell DT. The sperm epigenome: implications for the embryo. New York: Springer; 2014. Gannon JR, Emery BR, Jenkins TG, Carrell DT. The sperm epigenome: implications for the embryo. New York: Springer; 2014.
8.
Zurück zum Zitat Li J-Y, Lees-Murdock DJ, Xu G-L, Walsh CP. Timing of establishment of paternal methylation imprints in the mouse. Genomics. 2004;84:952–60.CrossRefPubMed Li J-Y, Lees-Murdock DJ, Xu G-L, Walsh CP. Timing of establishment of paternal methylation imprints in the mouse. Genomics. 2004;84:952–60.CrossRefPubMed
9.
Zurück zum Zitat Gomes MVM, Pelosi GG. Epigenetic vulnerability and the environmental influence on health. Exp Biol Med. 2013;238:859–65.CrossRef Gomes MVM, Pelosi GG. Epigenetic vulnerability and the environmental influence on health. Exp Biol Med. 2013;238:859–65.CrossRef
10.
11.
Zurück zum Zitat Klenova EM, Morse HC, Ohlsson R, Lobanenkov VV. The novel BORIS+CTCF gene family is uniquely involved in the epigenetics of normal biology and cancer. Semin Cancer Biol. 2002;12:399–414.CrossRefPubMed Klenova EM, Morse HC, Ohlsson R, Lobanenkov VV. The novel BORIS+CTCF gene family is uniquely involved in the epigenetics of normal biology and cancer. Semin Cancer Biol. 2002;12:399–414.CrossRefPubMed
12.
Zurück zum Zitat Gabory A, Ripoche MA, Le Digarcher A, Watrin F, Ziyyat A, Forne T, et al. H19 acts as a trans regulator of the imprinted gene network controlling growth in mice. Dev. 2009;136:3413–21.CrossRef Gabory A, Ripoche MA, Le Digarcher A, Watrin F, Ziyyat A, Forne T, et al. H19 acts as a trans regulator of the imprinted gene network controlling growth in mice. Dev. 2009;136:3413–21.CrossRef
13.
Zurück zum Zitat Poplinski A, Tuttelmann F, Kanber D, Horsthemke B, Gromoll J. Idiopathic male infertility is strongly associated with aberrant methylation of MEST and IGF2/H19 ICR1. Int J Androl. 2010;33:642–9.PubMed Poplinski A, Tuttelmann F, Kanber D, Horsthemke B, Gromoll J. Idiopathic male infertility is strongly associated with aberrant methylation of MEST and IGF2/H19 ICR1. Int J Androl. 2010;33:642–9.PubMed
14.
Zurück zum Zitat Olszewska M, Barciszewska MZ, Fraczek M, Huleyuk N, Chernykh VB, Zastavna D, et al. Global methylation status of sperm DNA in carriers of chromosome structural aberrations. Asian J Androl. 2016;19:117–24.PubMedCentral Olszewska M, Barciszewska MZ, Fraczek M, Huleyuk N, Chernykh VB, Zastavna D, et al. Global methylation status of sperm DNA in carriers of chromosome structural aberrations. Asian J Androl. 2016;19:117–24.PubMedCentral
15.
Zurück zum Zitat Kerjean A, Dupont JM, Vasseur C, Le Tessier D, Cuisset L, Paldi A, et al. Establishment of the paternal methylation imprint of the human H19 and MEST/PEG1 genes during spermatogenesis. Hum Mol Genet. 2000;9:2183–7.CrossRefPubMed Kerjean A, Dupont JM, Vasseur C, Le Tessier D, Cuisset L, Paldi A, et al. Establishment of the paternal methylation imprint of the human H19 and MEST/PEG1 genes during spermatogenesis. Hum Mol Genet. 2000;9:2183–7.CrossRefPubMed
16.
Zurück zum Zitat Boissonnas CC, El Abdalaoui H, Haelewyn V, Fauque P, Dupont JM, Gut I, et al. Specific epigenetic alterations of IGF2-H19 locus in spermatozoa from infertile men. Eur J Hum Genet. 2010;18:73–80.CrossRefPubMed Boissonnas CC, El Abdalaoui H, Haelewyn V, Fauque P, Dupont JM, Gut I, et al. Specific epigenetic alterations of IGF2-H19 locus in spermatozoa from infertile men. Eur J Hum Genet. 2010;18:73–80.CrossRefPubMed
17.
Zurück zum Zitat Gunes S, Agarwal A, Henkel R, Mahmutoglu A, Sharma R, Esteves S, et al. Association between promoter methylation of MLH1 and MSH2 and reactive oxygen species in oligozoospermic men—a pilot study. Andrologia. 2017;50(3):1–6. Gunes S, Agarwal A, Henkel R, Mahmutoglu A, Sharma R, Esteves S, et al. Association between promoter methylation of MLH1 and MSH2 and reactive oxygen species in oligozoospermic men—a pilot study. Andrologia. 2017;50(3):1–6.
18.
Zurück zum Zitat Juyena NS, Stelletta C. Seminal plasma: an essential attribute to spermatozoa. J Androl. 2012;33:536–51.CrossRefPubMed Juyena NS, Stelletta C. Seminal plasma: an essential attribute to spermatozoa. J Androl. 2012;33:536–51.CrossRefPubMed
19.
Zurück zum Zitat Minai-Tehrani A, Jafarzadeh N, Gilany K. Metabolomics: a state-of-the-art technology for better understanding of male infertility. Andrologia. 2016;48(6):609–16.CrossRefPubMed Minai-Tehrani A, Jafarzadeh N, Gilany K. Metabolomics: a state-of-the-art technology for better understanding of male infertility. Andrologia. 2016;48(6):609–16.CrossRefPubMed
20.
Zurück zum Zitat Wishart DS, Jewison T, Guo AC, Wilson M, Knox C, Liu Y, et al. HMDB 3.0—the human metabolome database in 2013. Biochem Biophys Res Commun. 2012:D801–7. Wishart DS, Jewison T, Guo AC, Wilson M, Knox C, Liu Y, et al. HMDB 3.0—the human metabolome database in 2013. Biochem Biophys Res Commun. 2012:D801–7.
21.
Zurück zum Zitat Deepinder F, Chowdary HT, Agarwal A. Role of metabolomic analysis of biomarkers in the management of male infertility. Expert Rev Mol Diagn. 2007;7:351–8.CrossRefPubMed Deepinder F, Chowdary HT, Agarwal A. Role of metabolomic analysis of biomarkers in the management of male infertility. Expert Rev Mol Diagn. 2007;7:351–8.CrossRefPubMed
22.
Zurück zum Zitat Kobayashi H, Hiura H, John RM, Sato A, Otsu E, Kobayashi N, et al. DNA methylation errors at imprinted loci after assisted conception originate in the parental sperm. Eur J Hum Genet. 2009;17:1582–91.CrossRefPubMedPubMedCentral Kobayashi H, Hiura H, John RM, Sato A, Otsu E, Kobayashi N, et al. DNA methylation errors at imprinted loci after assisted conception originate in the parental sperm. Eur J Hum Genet. 2009;17:1582–91.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Kagami M, Nagai T, Fukami M, Yamazawa K, Ogata T. Silver-Russell syndrome in a girl born after in vitro fertilization: partial hypermethylation at the differentially methylated region of PEG1/MEST. J Assist Reprod Gen. 2007;24:131–6.CrossRef Kagami M, Nagai T, Fukami M, Yamazawa K, Ogata T. Silver-Russell syndrome in a girl born after in vitro fertilization: partial hypermethylation at the differentially methylated region of PEG1/MEST. J Assist Reprod Gen. 2007;24:131–6.CrossRef
24.
Zurück zum Zitat Carrell DT, Aston KI, Oliva R, Emery B, De Jonge C. The “omics” of human male infertility: integrating big data in a systems biology approach. Cell Tissue Res. 2016;363:295–312.CrossRefPubMed Carrell DT, Aston KI, Oliva R, Emery B, De Jonge C. The “omics” of human male infertility: integrating big data in a systems biology approach. Cell Tissue Res. 2016;363:295–312.CrossRefPubMed
25.
Zurück zum Zitat Bhusari SS, Dobosy JR, Fu V, Almassi N, Oberley T, Jarrard DF. Superoxide dismutase 1 knockdown induces oxidative stress and DNA methylation loss in the prostate. Epigenet. 2010;5:402–9.CrossRef Bhusari SS, Dobosy JR, Fu V, Almassi N, Oberley T, Jarrard DF. Superoxide dismutase 1 knockdown induces oxidative stress and DNA methylation loss in the prostate. Epigenet. 2010;5:402–9.CrossRef
26.
Zurück zum Zitat Kasperczyk A, Dobrakowski M, Czuba ZP, Horak S, Kasperczyk S. Environmental exposure to lead induces oxidative stress and modulates the function of the antioxidant defense system and the immune system in the semen of males with normal semen profile. Toxicol Appl Pharmacol. 2015;284:339–44.CrossRefPubMed Kasperczyk A, Dobrakowski M, Czuba ZP, Horak S, Kasperczyk S. Environmental exposure to lead induces oxidative stress and modulates the function of the antioxidant defense system and the immune system in the semen of males with normal semen profile. Toxicol Appl Pharmacol. 2015;284:339–44.CrossRefPubMed
27.
Zurück zum Zitat Moazamian R, Polhemus A, Connaughton H, Fraser B, Whiting S, Gharagozloo P, et al. Oxidative stress and human spermatozoa: diagnostic and functional significance of aldehydes generated as a result of lipid peroxidation. MHR: Basic Sci Reprod Med. 2015;21:502–15. Moazamian R, Polhemus A, Connaughton H, Fraser B, Whiting S, Gharagozloo P, et al. Oxidative stress and human spermatozoa: diagnostic and functional significance of aldehydes generated as a result of lipid peroxidation. MHR: Basic Sci Reprod Med. 2015;21:502–15.
28.
Zurück zum Zitat WHO. WHO laboratory manual for the examination and processing of human semen. Geneva: World Health Organization; 2010. WHO. WHO laboratory manual for the examination and processing of human semen. Geneva: World Health Organization; 2010.
29.
Zurück zum Zitat Homa ST, Vessey W, Perez-Miranda A, Riyait T, Agarwal A. Reactive oxygen species (ROS) in human semen: determination of a reference range. J Assist Reprod Genet. 2015;32:757–64.CrossRefPubMedPubMedCentral Homa ST, Vessey W, Perez-Miranda A, Riyait T, Agarwal A. Reactive oxygen species (ROS) in human semen: determination of a reference range. J Assist Reprod Genet. 2015;32:757–64.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Shamsi MB, Venkatesh S, Pathak D, Deka D, Dada R. Sperm DNA damage & oxidative stress in recurrent spontaneous abortion (RSA). Indian J Med Res. 2011;133:550–1.PubMedPubMedCentral Shamsi MB, Venkatesh S, Pathak D, Deka D, Dada R. Sperm DNA damage & oxidative stress in recurrent spontaneous abortion (RSA). Indian J Med Res. 2011;133:550–1.PubMedPubMedCentral
31.
Zurück zum Zitat Jiang L, Zheng T, Huang J, Mo J, Zhou H, Liu M, et al. Association of semen cytokines with reactive oxygen species and histone transition abnormalities. J Assist Reprod Gen. 2016;33:1239–46.CrossRef Jiang L, Zheng T, Huang J, Mo J, Zhou H, Liu M, et al. Association of semen cytokines with reactive oxygen species and histone transition abnormalities. J Assist Reprod Gen. 2016;33:1239–46.CrossRef
32.
Zurück zum Zitat Agarwal A, Sharma R, Gupta S, Harlev A, Ahmad G, du Plessis SS, et al. Oxidative stress in human reproduction: shedding light on a complicated phenomenon. Cham: Springer International Publishing; 2017.CrossRef Agarwal A, Sharma R, Gupta S, Harlev A, Ahmad G, du Plessis SS, et al. Oxidative stress in human reproduction: shedding light on a complicated phenomenon. Cham: Springer International Publishing; 2017.CrossRef
33.
Zurück zum Zitat Mahfouz R, Sharma R, Sharma D, Sabanegh E, Agarwal A. Diagnostic value of the total antioxidant capacity (TAC) in human seminal plasma. Fertil Steril. 2009;91:805–11.CrossRefPubMed Mahfouz R, Sharma R, Sharma D, Sabanegh E, Agarwal A. Diagnostic value of the total antioxidant capacity (TAC) in human seminal plasma. Fertil Steril. 2009;91:805–11.CrossRefPubMed
34.
Zurück zum Zitat Fernández JL, Muriel L, Goyanes V, Segrelles E, Gosálvez J, Enciso M, et al. Halosperm® is an easy, available, and cost-effective alternative for determining sperm DNA fragmentation. Fertil Steril. 2005;84:860.CrossRefPubMed Fernández JL, Muriel L, Goyanes V, Segrelles E, Gosálvez J, Enciso M, et al. Halosperm® is an easy, available, and cost-effective alternative for determining sperm DNA fragmentation. Fertil Steril. 2005;84:860.CrossRefPubMed
35.
Zurück zum Zitat Esteves SC, Sharma RK, Gosálvez J, Agarwal A. A translational medicine appraisal of specialized andrology testing in unexplained male infertility. Int Urol Nephrol. 2014;46:1037–52.CrossRefPubMed Esteves SC, Sharma RK, Gosálvez J, Agarwal A. A translational medicine appraisal of specialized andrology testing in unexplained male infertility. Int Urol Nephrol. 2014;46:1037–52.CrossRefPubMed
36.
Zurück zum Zitat Barnard L, Aston KI. Spermatogenesis: methods and protocols. New York: Humana; 2012. Barnard L, Aston KI. Spermatogenesis: methods and protocols. New York: Humana; 2012.
37.
Zurück zum Zitat Zini A, Agarwal A. Sperm chromatin: biological and clinical applications in male infertility and assisted reproduction. New York: Springer; 2011.CrossRef Zini A, Agarwal A. Sperm chromatin: biological and clinical applications in male infertility and assisted reproduction. New York: Springer; 2011.CrossRef
38.
Zurück zum Zitat Paiva C, Amaral A, Rodriguez M, Canyellas N, Correig X, Ballescà J, et al. Identification of endogenous metabolites in human sperm cells using proton nuclear magnetic resonance (1H-NMR) spectroscopy and gas chromatography-mass spectrometry (GC-MS). Andrology. 2015;3:496–505.CrossRefPubMed Paiva C, Amaral A, Rodriguez M, Canyellas N, Correig X, Ballescà J, et al. Identification of endogenous metabolites in human sperm cells using proton nuclear magnetic resonance (1H-NMR) spectroscopy and gas chromatography-mass spectrometry (GC-MS). Andrology. 2015;3:496–505.CrossRefPubMed
39.
Zurück zum Zitat Brown FF, Campbell ID, Kuchel PW. Human erythrocyte metabolism studies by 1H spin echo NMR. FEBS Lett. 1977;82:12–6.CrossRefPubMed Brown FF, Campbell ID, Kuchel PW. Human erythrocyte metabolism studies by 1H spin echo NMR. FEBS Lett. 1977;82:12–6.CrossRefPubMed
40.
Zurück zum Zitat Viant MR. Improved methods for the acquisition and interpretation of NMR metabolomic data. Biochem Biophys Res Commun. 2003;310:943–8.CrossRefPubMed Viant MR. Improved methods for the acquisition and interpretation of NMR metabolomic data. Biochem Biophys Res Commun. 2003;310:943–8.CrossRefPubMed
41.
Zurück zum Zitat Niederberger C. The “omics” of human male infertility: integrating big data in a systems biology approach. J Urol. 2016;196:295–312.CrossRef Niederberger C. The “omics” of human male infertility: integrating big data in a systems biology approach. J Urol. 2016;196:295–312.CrossRef
42.
Zurück zum Zitat Kostidis S, Addie RD, Morreau H, Mayboroda OA, Giera M. Quantitative NMR analysis of intra- and extracellular metabolism of mammalian cells: a tutorial. Anal Chim Acta. 2017;980:1–24.CrossRefPubMed Kostidis S, Addie RD, Morreau H, Mayboroda OA, Giera M. Quantitative NMR analysis of intra- and extracellular metabolism of mammalian cells: a tutorial. Anal Chim Acta. 2017;980:1–24.CrossRefPubMed
43.
Zurück zum Zitat Pechlivanis A, Kostidis S, Saraslanidis P, Petridou A, Tsalis G, Mougios V, et al. 1H NMR-based metabonomic investigation of the effect of two different exercise sessions on the metabolic fingerprint of human urine. J Proteome Res. 2010;9:6405–16.CrossRefPubMed Pechlivanis A, Kostidis S, Saraslanidis P, Petridou A, Tsalis G, Mougios V, et al. 1H NMR-based metabonomic investigation of the effect of two different exercise sessions on the metabolic fingerprint of human urine. J Proteome Res. 2010;9:6405–16.CrossRefPubMed
44.
45.
Zurück zum Zitat Höcker M, Rosenberg I, Xavier R, Henihan RJ, Wiedenmann B, Rosewicz S, et al. Oxidative stress activates the human histidine decarboxylase promoter in AGS gastric cancer cells. J Biol Chem. 1998;273:23046–54.CrossRefPubMed Höcker M, Rosenberg I, Xavier R, Henihan RJ, Wiedenmann B, Rosewicz S, et al. Oxidative stress activates the human histidine decarboxylase promoter in AGS gastric cancer cells. J Biol Chem. 1998;273:23046–54.CrossRefPubMed
46.
Zurück zum Zitat Ravanbakhsh S, Liu P, Bjordahl TC, Mandal R, Grant JR, Wilson M, et al. Seminal plasma metabolomics approach for the diagnosis of unexplained male infertility. PLoS One. 2015;10:1–13. Ravanbakhsh S, Liu P, Bjordahl TC, Mandal R, Grant JR, Wilson M, et al. Seminal plasma metabolomics approach for the diagnosis of unexplained male infertility. PLoS One. 2015;10:1–13.
47.
Zurück zum Zitat Alonso A, Marsal S, Julià A. Analytical methods in untargeted metabolomics: state of the art in 2015. Front Bioeng Biotechnol. 2015;3:1–20.CrossRef Alonso A, Marsal S, Julià A. Analytical methods in untargeted metabolomics: state of the art in 2015. Front Bioeng Biotechnol. 2015;3:1–20.CrossRef
48.
Zurück zum Zitat Darbandi M, Darbandi S, Khorshid HRK, Akhondi MM, Mokarram P, Sadeghi MR. A simple, rapid and economic manual method for human sperm DNA extraction in genetic and epigenetic studies. Middle East Fertil Soc J, 2017. Darbandi M, Darbandi S, Khorshid HRK, Akhondi MM, Mokarram P, Sadeghi MR. A simple, rapid and economic manual method for human sperm DNA extraction in genetic and epigenetic studies. Middle East Fertil Soc J, 2017.
49.
Zurück zum Zitat Li Y, Tollefsbol TO. DNA methylation detection: bisulfite genomic sequencing analysis. Epigenet Protoc. 2011;79:11–21.CrossRef Li Y, Tollefsbol TO. DNA methylation detection: bisulfite genomic sequencing analysis. Epigenet Protoc. 2011;79:11–21.CrossRef
50.
Zurück zum Zitat Schuebel KE, Chen W, Cope L, Glöckner SC, Suzuki H, Yi J-M, et al. Comparing the DNA hypermethylome with gene mutations in human colorectal cancer. PLoS Genet. 2007;3:1709–23.CrossRefPubMed Schuebel KE, Chen W, Cope L, Glöckner SC, Suzuki H, Yi J-M, et al. Comparing the DNA hypermethylome with gene mutations in human colorectal cancer. PLoS Genet. 2007;3:1709–23.CrossRefPubMed
51.
Zurück zum Zitat Mokarram P, Kumar K, Brim H, Naghibalhossaini F, Saberi-firoozi M, Nouraie M, et al. Distinct high-profile methylated genes in colorectal cancer. PLoS One. 2009;4:1–9.CrossRef Mokarram P, Kumar K, Brim H, Naghibalhossaini F, Saberi-firoozi M, Nouraie M, et al. Distinct high-profile methylated genes in colorectal cancer. PLoS One. 2009;4:1–9.CrossRef
53.
Zurück zum Zitat Mahadevan S, Shah SL, Marrie TJ, Slupsky CM. Analysis of metabolomic data using support vector machines. Anal Chem. 2008;80:7562–70.CrossRefPubMed Mahadevan S, Shah SL, Marrie TJ, Slupsky CM. Analysis of metabolomic data using support vector machines. Anal Chem. 2008;80:7562–70.CrossRefPubMed
54.
Zurück zum Zitat Moore GE, Ishida M, Demetriou C, Al-Olabi L, Leon LJ, Thomas AC, et al. The role and interaction of imprinted genes in human fetal growth. Philos Trans R Soc B. 2015;370:1–12.CrossRef Moore GE, Ishida M, Demetriou C, Al-Olabi L, Leon LJ, Thomas AC, et al. The role and interaction of imprinted genes in human fetal growth. Philos Trans R Soc B. 2015;370:1–12.CrossRef
55.
Zurück zum Zitat Nordin M, Bergman D, Halje M, Engström W, Ward A. Epigenetic regulation of the Igf2/H19 gene cluster. Cell Prolif. 2014;47:189–99.CrossRefPubMed Nordin M, Bergman D, Halje M, Engström W, Ward A. Epigenetic regulation of the Igf2/H19 gene cluster. Cell Prolif. 2014;47:189–99.CrossRefPubMed
56.
Zurück zum Zitat Bartolomei MS, Ferguson-Smith AC. Mammalian genomic imprinting. Cold Spring Harb Perspect Biol. 2011;3:1–17.CrossRef Bartolomei MS, Ferguson-Smith AC. Mammalian genomic imprinting. Cold Spring Harb Perspect Biol. 2011;3:1–17.CrossRef
57.
Zurück zum Zitat Kanduri C. Long noncoding RNAs: lessons from genomic imprinting. BBA-Gene Regul Mech. 1859;2016:102–11. Kanduri C. Long noncoding RNAs: lessons from genomic imprinting. BBA-Gene Regul Mech. 1859;2016:102–11.
58.
Zurück zum Zitat Kingsley SL, Deyssenroth MA, Kelsey KT, Awad YA, Kloog I, Schwartz JD, et al. Maternal residential air pollution and placental imprinted gene expression. Environ Int. 2017;108:204–11.CrossRefPubMedPubMedCentral Kingsley SL, Deyssenroth MA, Kelsey KT, Awad YA, Kloog I, Schwartz JD, et al. Maternal residential air pollution and placental imprinted gene expression. Environ Int. 2017;108:204–11.CrossRefPubMedPubMedCentral
59.
Zurück zum Zitat Gabory A, Ripoche M-A, Yoshimizu T, Dandolo L. The H19 gene: regulation and function of a non-coding RNA. Cytogenet Genome Res. 2006;113:188–93.CrossRefPubMed Gabory A, Ripoche M-A, Yoshimizu T, Dandolo L. The H19 gene: regulation and function of a non-coding RNA. Cytogenet Genome Res. 2006;113:188–93.CrossRefPubMed
61.
Zurück zum Zitat DeBaun MR, Niemitz EL, Feinberg AP. Association of in vitro fertilization with Beckwith-Wiedemann syndrome and epigenetic alterations of LIT1 and H19. Am J Hum Genet. 2003;72:156–60.CrossRefPubMed DeBaun MR, Niemitz EL, Feinberg AP. Association of in vitro fertilization with Beckwith-Wiedemann syndrome and epigenetic alterations of LIT1 and H19. Am J Hum Genet. 2003;72:156–60.CrossRefPubMed
62.
Zurück zum Zitat Hammoud SS, Purwar J, Pflueger C, Cairns BR, Carrell DT. Alterations in sperm DNA methylation patterns at imprinted loci in two classes of infertility. Fertil Steril. 2010;94:1728–33.CrossRefPubMed Hammoud SS, Purwar J, Pflueger C, Cairns BR, Carrell DT. Alterations in sperm DNA methylation patterns at imprinted loci in two classes of infertility. Fertil Steril. 2010;94:1728–33.CrossRefPubMed
63.
Zurück zum Zitat Marques CJ, Francisco T, Sousa S, Carvalho F, Barros A, Sousa M. Methylation defects of imprinted genes in human testicular spermatozoa. Fertil Steril. 2010;94:585–94.CrossRefPubMed Marques CJ, Francisco T, Sousa S, Carvalho F, Barros A, Sousa M. Methylation defects of imprinted genes in human testicular spermatozoa. Fertil Steril. 2010;94:585–94.CrossRefPubMed
64.
Zurück zum Zitat Stouder C, Somm E, Paoloni-Giacobino A. Prenatal exposure to ethanol: a specific effect on the H19 gene in sperm. Reprod Toxicol. 2011;31:507–12.CrossRefPubMed Stouder C, Somm E, Paoloni-Giacobino A. Prenatal exposure to ethanol: a specific effect on the H19 gene in sperm. Reprod Toxicol. 2011;31:507–12.CrossRefPubMed
65.
Zurück zum Zitat Maher AD, Patki P, Lindon JC, Want EJ, Holmes E, Craggs M, et al. Seminal oligouridinosis: low uridine secretion as a biomarker for infertility in spinal neurotrauma. Clin Chem. 2008;54:2063–6.CrossRefPubMed Maher AD, Patki P, Lindon JC, Want EJ, Holmes E, Craggs M, et al. Seminal oligouridinosis: low uridine secretion as a biomarker for infertility in spinal neurotrauma. Clin Chem. 2008;54:2063–6.CrossRefPubMed
66.
Zurück zum Zitat Gupta A, Mahdi AA, Ahmad MK, Shukla KK, Jaiswer SP, Shankhwar SN. 1H NMR spectroscopic studies on human seminal plasma: a probative discriminant function analysis classification model. J Pharm Biomed Anal. 2011;54:106–13.CrossRefPubMed Gupta A, Mahdi AA, Ahmad MK, Shukla KK, Jaiswer SP, Shankhwar SN. 1H NMR spectroscopic studies on human seminal plasma: a probative discriminant function analysis classification model. J Pharm Biomed Anal. 2011;54:106–13.CrossRefPubMed
67.
Zurück zum Zitat Vander Heiden MG, DeBerardinis RJ. Understanding the intersections between metabolism and cancer biology. Cell. 2017;168:657–69.CrossRefPubMed Vander Heiden MG, DeBerardinis RJ. Understanding the intersections between metabolism and cancer biology. Cell. 2017;168:657–69.CrossRefPubMed
68.
Zurück zum Zitat Son DO, Satsu H, Shimizu M. Histidine inhibits oxidative stress- and TNF-α-induced interleukin-8 secretion in intestinal epithelial cells. FEBS Lett. 2005;579:4671–7.CrossRefPubMed Son DO, Satsu H, Shimizu M. Histidine inhibits oxidative stress- and TNF-α-induced interleukin-8 secretion in intestinal epithelial cells. FEBS Lett. 2005;579:4671–7.CrossRefPubMed
69.
Zurück zum Zitat Peterson JW, Boldogh I, Popov VL, Saini SS, Chopra AK. Anti-inflammatory and antisecretory potential of histidine in Salmonella-challenged mouse small intestine. Lab Investig. 1998;78:523–34.PubMed Peterson JW, Boldogh I, Popov VL, Saini SS, Chopra AK. Anti-inflammatory and antisecretory potential of histidine in Salmonella-challenged mouse small intestine. Lab Investig. 1998;78:523–34.PubMed
70.
Zurück zum Zitat Jiménez-Trejo F, Tapia-Rodríguez M, Cerbón M, Kuhn DM, Manjarrez-Gutiérrez G, Mendoza-Rodríguez CA, et al. Evidence of 5-HT components in human sperm: implications for protein tyrosine phosphorylation and the physiology of motility. Reproduction. 2012;144:677–85.CrossRefPubMedPubMedCentral Jiménez-Trejo F, Tapia-Rodríguez M, Cerbón M, Kuhn DM, Manjarrez-Gutiérrez G, Mendoza-Rodríguez CA, et al. Evidence of 5-HT components in human sperm: implications for protein tyrosine phosphorylation and the physiology of motility. Reproduction. 2012;144:677–85.CrossRefPubMedPubMedCentral
71.
Zurück zum Zitat El-Sheshtawy RI, El-Nattat WS, Sabra HA. Effect of addition of catalase with or without L-tryptophan on cryopreservation of bull extended semen and conception rate. Glob Vet. 2013;11:280–4. El-Sheshtawy RI, El-Nattat WS, Sabra HA. Effect of addition of catalase with or without L-tryptophan on cryopreservation of bull extended semen and conception rate. Glob Vet. 2013;11:280–4.
72.
Zurück zum Zitat Naz RK, Rajesh PB. Role of tyrosine phosphorylation in sperm capacitation/acrosome reaction. Reprod Biol Endocrin. 2004;2:1–12.CrossRef Naz RK, Rajesh PB. Role of tyrosine phosphorylation in sperm capacitation/acrosome reaction. Reprod Biol Endocrin. 2004;2:1–12.CrossRef
73.
Zurück zum Zitat Pukazhenthi BS, Long JA, Wildt DE, Ottinger MA, Armstrong DL, Howard J. Regulation of sperm function by protein tyrosine phosphorylation in diverse wild felid species. J Androl. 1998;19:675–85.PubMed Pukazhenthi BS, Long JA, Wildt DE, Ottinger MA, Armstrong DL, Howard J. Regulation of sperm function by protein tyrosine phosphorylation in diverse wild felid species. J Androl. 1998;19:675–85.PubMed
74.
Zurück zum Zitat Sati L, Cayli S, Delpiano E, Sakkas D, Huszar G. The pattern of tyrosine phosphorylation in human sperm in response to binding to zona pellucida or hyaluronic acid. Reprod Sci. 2014;21:573–81.CrossRefPubMedPubMedCentral Sati L, Cayli S, Delpiano E, Sakkas D, Huszar G. The pattern of tyrosine phosphorylation in human sperm in response to binding to zona pellucida or hyaluronic acid. Reprod Sci. 2014;21:573–81.CrossRefPubMedPubMedCentral
75.
76.
Zurück zum Zitat Banihani SA. Semen quality as affected by olive oil. Int J Food Prop. 2017;20:1901–6. Banihani SA. Semen quality as affected by olive oil. Int J Food Prop. 2017;20:1901–6.
77.
Zurück zum Zitat Gomes VPM, Torres C, Rodriguez-Borges JE, Paiva-Martins F. A convenient synthesis of hydroxytyrosol monosulfate metabolites. J Agric Food Chem. 2015;63:9565–71.CrossRefPubMed Gomes VPM, Torres C, Rodriguez-Borges JE, Paiva-Martins F. A convenient synthesis of hydroxytyrosol monosulfate metabolites. J Agric Food Chem. 2015;63:9565–71.CrossRefPubMed
78.
Zurück zum Zitat Negri L, Benaglia R, Monti E, Morenghi E, Pizzocaro A, Setti PEL. Effect of superoxide dismutase supplementation on sperm DNA fragmentation. Arch Ital Urol Androl. 2017;89:212–8.CrossRefPubMed Negri L, Benaglia R, Monti E, Morenghi E, Pizzocaro A, Setti PEL. Effect of superoxide dismutase supplementation on sperm DNA fragmentation. Arch Ital Urol Androl. 2017;89:212–8.CrossRefPubMed
80.
Zurück zum Zitat Qiao S, Wu W, Chen M, Tang Q, Xia Y, Jia W, et al. Seminal plasma metabolomics approach for the diagnosis of unexplained male infertility. PLoS One. 2017;12:1–13. Qiao S, Wu W, Chen M, Tang Q, Xia Y, Jia W, et al. Seminal plasma metabolomics approach for the diagnosis of unexplained male infertility. PLoS One. 2017;12:1–13.
81.
Zurück zum Zitat Chen X, Hu C, Dai J, Chen L. Metabolomics analysis of seminal plasma in infertile males with kidney-yang deficiency: a preliminary study. Evid-Based Compl Alt. 2015;2015:1–8. Chen X, Hu C, Dai J, Chen L. Metabolomics analysis of seminal plasma in infertile males with kidney-yang deficiency: a preliminary study. Evid-Based Compl Alt. 2015;2015:1–8.
82.
Zurück zum Zitat Agarwal A, Ahmad G, Sharma R. Reference values of reactive oxygen species in seminal ejaculates using chemiluminescence assay. J Assist Reprod Gene. 2015;32:1721–9.CrossRef Agarwal A, Ahmad G, Sharma R. Reference values of reactive oxygen species in seminal ejaculates using chemiluminescence assay. J Assist Reprod Gene. 2015;32:1721–9.CrossRef
Metadaten
Titel
Reactive oxygen species-induced alterations in H19-Igf2 methylation patterns, seminal plasma metabolites, and semen quality
verfasst von
Mahsa Darbandi
Sara Darbandi
Ashok Agarwal
Saradha Baskaran
Sulagna Dutta
Pallav Sengupta
Hamid Reza Khorram Khorshid
Sandro Esteves
Kambiz Gilany
Mehdi Hedayati
Fatemeh Nobakht
Mohammad Mehdi Akhondi
Niknam Lakpour
Mohammad Reza Sadeghi
Publikationsdatum
31.10.2018
Verlag
Springer US
Erschienen in
Journal of Assisted Reproduction and Genetics / Ausgabe 2/2019
Print ISSN: 1058-0468
Elektronische ISSN: 1573-7330
DOI
https://doi.org/10.1007/s10815-018-1350-y

Weitere Artikel der Ausgabe 2/2019

Journal of Assisted Reproduction and Genetics 2/2019 Zur Ausgabe

Update Gynäkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert – ganz bequem per eMail.