DOI QR코드

DOI QR Code

Development of a Test Method for the Evaluation of DNA Damage in Mouse Spermatogonial Stem Cells

  • Jeon, Hye Lyun (Toxicological Screening and Testing Division, National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Osong Health Technology Administration Complex) ;
  • Yi, Jung-Sun (Toxicological Screening and Testing Division, National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Osong Health Technology Administration Complex) ;
  • Kim, Tae Sung (Toxicological Screening and Testing Division, National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Osong Health Technology Administration Complex) ;
  • Oh, Youkyung (Toxicological Screening and Testing Division, National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Osong Health Technology Administration Complex) ;
  • Lee, Hye Jeong (Department of Stem Cell Biology, Konkuk University School of Medicine) ;
  • Lee, Minseong (Department of Stem Cell Biology, Konkuk University School of Medicine) ;
  • Bang, Jin Seok (Department of Stem Cell Biology, Konkuk University School of Medicine) ;
  • Ko, Kinarm (Department of Stem Cell Biology, Konkuk University School of Medicine) ;
  • Ahn, Il Young (Toxicological Screening and Testing Division, National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Osong Health Technology Administration Complex) ;
  • Ko, Kyungyuk (Toxicological Screening and Testing Division, National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Osong Health Technology Administration Complex) ;
  • Kim, Joohwan (Toxicological Screening and Testing Division, National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Osong Health Technology Administration Complex) ;
  • Park, Hye-Kyung (Toxicological Evaluation and Research Department, National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Osong Health Technology Administration Complex) ;
  • Lee, Jong Kwon (Toxicological Screening and Testing Division, National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Osong Health Technology Administration Complex) ;
  • Sohn, Soo Jung (Toxicological Research Division, National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Osong Health Technology Administration Complex)
  • Received : 2017.01.31
  • Accepted : 2017.03.07
  • Published : 2017.04.15

Abstract

Although alternative test methods based on the 3Rs (Replacement, Reduction, Refinement) are being developed to replace animal testing in reproductive and developmental toxicology, they are still in an early stage. Consequently, we aimed to develop alternative test methods in male animals using mouse spermatogonial stem cells (mSSCs). Here, we modified the OECD TG 489 and optimized the in vitro comet assay in our previous study. This study aimed to verify the validity of in vitro tests involving mSSCs by comparing their results with those of in vivo tests using C57BL/6 mice by gavage. We selected hydroxyurea (HU), which is known to chemically induce male reproductive toxicity. The 50% inhibitory concentration ($IC_{50}$) value of HU was 0.9 mM, as determined by the MTT assay. In the in vitro comet assay, % tail DNA and Olive tail moment (OTM) after HU administration increased significantly, compared to the control. Annexin V, PI staining and TUNEL assays showed that HU caused apoptosis in mSSCs. In order to compare in vitro tests with in vivo tests, the same substances were administered to male C57BL/6 mice. Reproductive toxicity was observed at 25, 50, 100, and 200 mg/kg/day as measured by clinical measures of reduction in sperm motility and testicular weight. The comet assay, DCFH-DA assay, H&E staining, and TUNEL assay were also performed. The results of the test with C57BL/6 mice were similar to those with mSSCs for HU treatment. Finally, linear regression analysis showed a strong positive correlation between results of in vitro tests and those of in vivo. In conclusion, the present study is the first to demonstrate the effect of HU-induced DNA damage, ROS formation, and apoptosis in mSSCs. Further, the results of the current study suggest that mSSCs could be a useful model to predict male reproductive toxicity.

Keywords

References

  1. Rovida, C. and Hartung, T. (2009) Re-evaluation of animal numbers and costs for in vivo tests to accomplish REACH legislation requirements for chemicals - a report by the transatlantic think tank for toxicology (t(4)). ALTEX, 26, 187-208.
  2. Schulte, R.T., Ohl, D.A., Sigman, M. and Smith, G.D. (2010) Sperm DNA damage in male infertility: etiologies, assays, and outcomes. J. Assist. Reprod. Genet., 27, 3-12. https://doi.org/10.1007/s10815-009-9359-x
  3. Makker, K., Agarwal, A. and Sharma, R. (2009) Oxidative stress & male infertility. Indian J. Med. Res., 129, 357-367.
  4. Adler, S., Basketter, D., Creton, S., Pelkonen, O., van Benthem, J., Zuang, V., Andersen, K.E., Angers-Loustau, A., Aptula, A., Bal-Price, A., Benfenati, E., Bernauer, U., Bessems, J., Bois, F.Y., Boobis, A., Brandon, E., Bremer, S., Broschard, T., Casati, S., Coecke, S., Corvi, R., Cronin, M., Daston, G., Dekant, W., Felter, S., Grignard, E., Gundert- Remy, U., Heinonen, T., Kimber, I., Kleinjans, J., Komulainen, H., Kreiling, R., Kreysa, J., Leite, S.B., Loizou, G., Maxwell, G., Mazzatorta, P., Munn, S., Pfuhler, S., Phrakonkham, P., Piersma, A., Poth, A., Prieto, P., Repetto, G., Rogiers, V., Schoeters, G., Schwarz, M., Serafimova, R., Tähti, H., Testai, E., van Delft, J., van Loveren, H., Vinken, M., Worth, A. and Zaldivar, J.M. (2011) Alternative (non-animal) methods for cosmetics testing: current status and future prospects-2010. Arch. Toxicol., 85, 367-485. https://doi.org/10.1007/s00204-011-0693-2
  5. Tegelenbosch, R.A. and de Rooij, D.G. (1993) A quantitative study of spermatogonial multiplication and stem cell renewal in the C3H/101 F1 hybrid mouse. Mutat. Res., 290, 193-200. https://doi.org/10.1016/0027-5107(93)90159-D
  6. Ko, K., Arauzo-Bravo, M.J., Kim, J., Stehling, M. and Scholer, R.H. (2010) Conversion of adult mouse unipotent germline stem cells into pluripotent stem cells. Nat. Protoc., 5, 921-928. https://doi.org/10.1038/nprot.2010.44
  7. Kanatsu-Shinohara, M., Ogonuki, N., Inoue, K., Miki, H., Ogura, A., Toyokuni, S. and Shinohara, T. (2003) Long-term proliferation in culture and germline transmission of mouse male germline stem cells. Biol. Reprod., 69, 612-616. https://doi.org/10.1095/biolreprod.103.017012
  8. Ko, K., Tapia, N., Wu, G., Kim, J.B., Arauzo Bravo, M.J., Sasse, P., Glaser, T., Ruau, D., Han, D.W., Greber, B., Hausdorfer, K., Sebastiano, V., Stehling, M., Fleischmann, B.K., Brustle, O., Zenke, M. and Scholer, H.R. (2009) Induction of pluripotency in adult unipotent germline stem cells. Cell Stem Cell, 5, 87-96. https://doi.org/10.1016/j.stem.2009.05.025
  9. Meng, Y.U., Long, W., Yue, H., Zhi-min, L. and Jin-lian, H. (2013) ALK family inhibitor A83-01 promotes the proliferation of mouse male germline stem cells (mGSCs) under serum-and feeder-free conditions. J. Integr. Agric., 12, 1839- 1846. https://doi.org/10.1016/S2095-3119(13)60413-X
  10. Lei, Z., Qi-sheng, Z., Dong, L., Chao, L., Ahmed, K.E., Beibei, T., Jiu-zhou, S., Ya-ni, Z. and Bi-chun, L. (2015) Study on the role of JAK/STAT signaling pathway during chicken spermatogonial stem cells generation based on RNA-Seq. J. Integr. Agric., 14, 939-948. https://doi.org/10.1016/S2095-3119(14)60938-2
  11. Mahaldashtian, M., Naghdi, M., Ghorbanian, M.T., Makoolati, Z., Movahedin, M. and Mohamadi, S.M. (2016) In vitro effects of date palm (Phoenix dactylifera L.) pollen on colonization of neonate mouse spermatogonial stem cells. J. Ethnopharmacol., 186, 362-368. https://doi.org/10.1016/j.jep.2016.04.013
  12. Hashemi, E., Akhavan, O., Shamsara, M., Daliri, M., Dashtizad, M. and Farmacy, A. (2016) Synthesis and cytogenotoxicity evaluation of graphene on mice spermatogonial stem cells. Colloids Surf. B Biointerfaces, 146, 770-776. https://doi.org/10.1016/j.colsurfb.2016.07.019
  13. Tice, R.R., Agurell, E., Anderson, D., Burlinson, B., Hartmann, A., Kobayashi, H., Miyamae, Y., Rojas, E., Ryu, J.C. and Sasaki, Y.F. (2000) Single cell gel/comet assay: Guidelines for in vitro and in vivo genetic toxicology testing. Environ. Mol. Mutagen., 35, 206-221. https://doi.org/10.1002/(SICI)1098-2280(2000)35:3<206::AID-EM8>3.0.CO;2-J
  14. Lewis, S.E., John Aitken, R., Conner, S.J., Iuliis, G.D., Evenson, D.P., Henkel, R., Giwercman, A. and Gharagozloo, P. (2013) The imopact of sperm DNA damage in assisted conception and beyond: recent advances in diagnosis and treatment. Reprod. Biomed. Online, 27, 325-327. https://doi.org/10.1016/j.rbmo.2013.06.014
  15. OECD. (2014) Test No. 489: In vivo Mammalian alkaline Comet assay, OECD Publishing, Paris, pp. 1-25.
  16. Mohamed, H.M. and Mohamed, M.A. (2014) Effect of different doses of nandrolone decanoate on lipid peroxidation, DNA fragmentation, sperm abnormality and histopathology of testes of male Wister rats. Exp. Toxicol. Pathol., 67, 1-11.
  17. Ceribasi, A.O., Sakin, F., Turk, G., Sonmez, M. and Atessahin, A. (2012) Impact of ellagic acid on adriamycin-induced testicular histopathological lesions, apoptosis, lipid peroxidation and sperm damages. Exp. Toxicol. Pathol., 64, 717-724. https://doi.org/10.1016/j.etp.2011.01.006
  18. La Maestra, S., De Flora, S. and Micale, R.T. (2015) Effect of cigarette smoke on DNA damage, oxidative stress, andmorphological alterations in mouse testis and spermatozoa. Int. J. Hyg. Environ. Health, 218, 117-122. https://doi.org/10.1016/j.ijheh.2014.08.006
  19. Mojica-Villegas, M.A., Izquierdo-Vega, J.A., Chamorro-Cevallos, G. and Sanchez-Gutierrez, M. (2014) Protective effect of resveratrol on biomarkers of oxidative stress induced by iron/ ascorbate in mouse spermatozoa. Nutrients, 6, 489-503. https://doi.org/10.3390/nu6020489
  20. Khan, S., Ahmad, T., Parekh, C.V., Trivedi, P.P., Kushwaha, S. and Jena, G. (2011) Investigation on sodium valproate induced germ cell damage, oxidative stress and genotoxicity in male Swiss mice. Reprod. Toxicol., 32, 385-394. https://doi.org/10.1016/j.reprotox.2011.09.007
  21. Rahimipour, M., Talebi, A.R., Anvari, M., Sarcheshmeh, A.A. and Omidi, M. (2013) Effects of different doses of ethanol on sperm parameters, chromatin structure and apoptosis in adult mice. Eur. J. Obstet. Gynecol. Reprod. Biol., 170, 423-428. https://doi.org/10.1016/j.ejogrb.2013.06.038
  22. Yarbro, J.W. (1992) Mechanism of action of hydroxyurea. Semin. Oncol., 19, 1-10.
  23. Donehower, R.C. (1992) An overview of the clinical experience with hydroxyurea. Semin. Oncol., 19, 11-19.
  24. Evenson, D.P. and Jost, L.K. (1993) Hydroxyurea exposure alters mouse testicular kinetics and sperm chromatin structure. Cell Prolif., 26, 147-159. https://doi.org/10.1111/j.1365-2184.1993.tb00015.x
  25. Jones, K.M., Niaz, M.S., Brooks, C.M., Roberson, S.I., Aguinaga, M.P., Hills, E.R., Rice, V.M., Bourne, P., Bruce, D. and Archibong, A.E. (2009) Adverse effects of a clinically relevant dose of hydroxyurea used for the treatment of sickle cell disease on male fertility endpoints. Int. J. Environ. Res. Public Health, 6, 1124-1144. https://doi.org/10.3390/ijerph6031124
  26. Choi, N.Y., Park, Y.S., Ryu, J.S., Lee, H.J., Arauzo-Bravo, M.J., Ko, K., Han, D.W., Scholer, H.R. and Ko, K. (2014) A novel feeder-free culture system for expansion of mouse spermatogonial stem cells. Mol. Cells, 37, 473-479. https://doi.org/10.14348/molcells.2014.0080
  27. Guthrie, H.D. and Welch, G.R. (2012) Effects of reactive oxygen species on sperm function. Theriogenology, 78, 1700-1708. https://doi.org/10.1016/j.theriogenology.2012.05.002
  28. Sanocka, D. and Kurpisz, M. (2004) Reactive oxygen species and sperm cells. Reprod. Biol. Endocrinol, 2, 12. https://doi.org/10.1186/1477-7827-2-12
  29. Sakano, K., Oikawa, S., Hasegawa, K. and Kawanishi, S. (2001) Hydroxyurea induces site-specific DNA damage via formation of hydrogen peroxide and nitric oxide. Jpn. J. Cancer Res., 92, 1166-1174. https://doi.org/10.1111/j.1349-7006.2001.tb02136.x
  30. Hu, J., Song, D., Luo, G., Xu, S., Cao, Y. and Sun, Z. (2015) Activation of Toll like receptor 3 induces spermatogonial stem cell apoptosis. Cell Biochem. Funct., 33, 415-420. https://doi.org/10.1002/cbf.3133
  31. Zhou, L., Wu, C.Q., Luo, Y.W., Liao, M.Y. and Sun, Z.Y. (2015) Studies on the characteristics and mechanisms of testicular toxicity induced by Hydroxyurea. Toxicol. Mech. Methods, 25, 396-401. https://doi.org/10.3109/15376516.2015.1045657
  32. Sakai, T., Takahashi, M., Mitsumori, K., Yasuhara, K., Kawashima, K., Mayahara, H. and Ohno, Y. (2000) Collaborative work to evaluate toxicity on male reproductive organs by repeated dose studies in rats. J. Toxicol. Sci., 25, 1-21.
  33. Ozawa, S., Yokoi, R., Kitamura, T., Kuriyama, K., Kobayashi, K. and Shibata, N. (2000) Collaborative work to evaluate toxicity on male reproductive organs by repeated dose studies in rats 15) two-week and 4-week administration study of methyl methanesulfonate (MMS). J. Toxicol. Sci., 25, 155-162. https://doi.org/10.2131/jts.25.SpecialIssue_155
  34. Wiger, R., Hongslo, J.K., Evenson, D.P., De Angelis, P., Schwarze, P.E. and Holme, J.A. (1995) Effects of acetaminophen and hydroxyurea on spermatogenesis and sperm chromatin structure in laboratory mice. Reprod. Toxicol., 9, 21-33. https://doi.org/10.1016/0890-6238(94)00052-X
  35. Trivedi, P.P., Kushwaha, S., Tripathi, D.N. and Jena, G.B. (2010) Evaluation of male germ cell toxicity in rats: correlation between sperm head morphology and sperm comet assay. Mutat. Res., 703, 115-121. https://doi.org/10.1016/j.mrgentox.2010.08.005
  36. Steller, H. (1995) Mechanisms and genes of cellular suicide. Science, 267, 1445-1449. https://doi.org/10.1126/science.7878463
  37. Bejarano, I., Espino, J., Paredes, S.D., Ortiz, A., Lozano, G., Pariente, J.A. and Rodríguez, A.B. (2012) Apoptosis, ROS and calcium signaling in Human Spermatozoa: Relationship to Infertility (Bashamboo, A. Ed.). InTech, Croatia, pp. 51-76.
  38. Moline, J.M., Golden, A.L., Bar-Chama, N., Smith, E., Rauch, M.E., Chapin, R.E., Perreault, S.D., Schrader, S.M., Suk, W.A. and Landrigan, P.J. (2000) Exposure to hazardous substances and male reproductive health: a research framework. Environ. Health Perspect., 108, 803-813. https://doi.org/10.1289/ehp.00108s5803

Cited by

  1. Environmental Exposure of Sperm Sex-Chromosomes: A Gender Selection Technique vol.33, pp.4, 2017, https://doi.org/10.5487/TR.2017.33.4.315
  2. Sargassum serratifolium Extract Attenuates Interleukin-1β-Induced Oxidative Stress and Inflammatory Response in Chondrocytes by Suppressing the Activation of NF-κB, p38 MAPK, and PI3K/Akt vol.19, pp.8, 2018, https://doi.org/10.3390/ijms19082308