DOI QR코드

DOI QR Code

Effects of a mild heat treatment on mouse testicular gene expression and sperm quality

  • Zhao, Jun (School of Life Sciences, Northeast Normal University, Jilin Key Laboratory of Animal Resource Conservation and Utilization) ;
  • Zhang, Ying (Veterinary College, Jilin University) ;
  • Hao, Linlin (Veterinary College, Jilin University) ;
  • Wang, Jia (Veterinary College, Jilin University) ;
  • Zhang, Jiabao (Experiment Animal Centre, Jilin University) ;
  • Liu, Songcai (Veterinary College, Jilin University) ;
  • Ren, Bingzhong (School of Life Sciences, Northeast Normal University, Jilin Key Laboratory of Animal Resource Conservation and Utilization)
  • Received : 2010.06.18
  • Accepted : 2010.08.09
  • Published : 2010.12.31

Abstract

The decrease in sperm quality under heat stress causes a great loss in animal husbandry production. In order to reveal the mechanism underlying the sperm quality decrease caused by heat stress, we first established a mild heat-treated mouse model. Then, the sperm quality was identified. Further, the testicular proteome profile was mapped and compared with the control using 2D electrophoresis and mass spectrometry. Finally, the differential expressed proteins involved in the heat stress response were identified by real-time PCR and Western blotting. The results showed that heat stress caused a significant reduction in mouse sperm quality (P<0.05). Further, 52 protein spots on the 2D gel were found to differ between the heat-shocked tissues and the control. Of these spots, some repair proteins which might provide some explanation for the influence on sperm quality were found. We then focused on Bag-1, Hsp40, Hsp60 and Hsp70, which were found to be differently expressed after heat shock (P<0.05). Further analysis in this heat-shocked model suggests numerous potential mechanisms for heat shock-induced spermatogenic disorders.

Keywords

References

  1. Aktas C, Kanter M. 2009. A morphological study on Leydig cells of scrotal hyperthermia applied rats in short-term. J Mol Histol. 40:31-39. https://doi.org/10.1007/s10735-009-9210-9
  2. Barry MA, Behnke CA, Eastman A. 1990. Activation of programmed cell death (apoptosis) by cisplatin, other anticancer drugs, toxins and hyperthermia. Biochem Pharmacol. 40:2353-2362. https://doi.org/10.1016/0006-2952(90)90733-2
  3. Bravo MM, Aparicio IM, Garcia-Herreros M, Gil MC, Pena FJ, Garcia-Marin LJ. 2005. Changes in tyrosine phosphorylation associated with true capacitation and capacitation-like state in boar spermatozoa. Mol Reprod Dev. 71:88-96. https://doi.org/10.1002/mrd.20286
  4. Dix DJ. 1997. Hsp70 expression and function during gametogenesis. Cell Stress Chaperones. 2:73-77. https://doi.org/10.1379/1466-1268(1997)002<0073:HEAFDG>2.3.CO;2
  5. Dix DJ, Allen JW, Collins BW, Mori C, Nakamura N, Poorman-Allen P, Goulding EH, Eddy EM. 1996. Targeted gene disruption of Hsp70-2 results in failed meiosis, germ cell apoptosis, and male infertility. PNAS. 93:3264-3268. https://doi.org/10.1073/pnas.93.8.3264
  6. Dix DJ, Allen JW, Collins BW, Poorman-Allen P, Mori C, Blizard DR, Brown PR, Goulding EH, Strong BD, Eddy EM. 1997. HSP70-2 is required for desynapsis of synaptonemal complexes during meiotic prophase in juvenile and adult mouse spermatocytes. Development. 124:4595-4603.
  7. Georgopoulos C, Welch WJ. 1993. Role of the major heat shock proteins as molecular chaperones. Annu Rev Cell Biol. 9:601-634. https://doi.org/10.1146/annurev.cb.09.110193.003125
  8. Giwercman A, Richthoff J, Hjollund H, Bonde JP, Jepson K, Frohm B, Spano M. 2003. Correlation between sperm motility and sperm chromatin structure assay parameters. Fertil Steril. 80:1404-1412. https://doi.org/10.1016/S0015-0282(03)02212-X
  9. Hohfeld J, Jentsch S. 1997. GrpE-like regulation of the Hsc70 chaperone by the anti-apoptotic protein BAG-1. EMBO J. 16:6209-6216. https://doi.org/10.1093/emboj/16.20.6209
  10. Holstein AF, Schulze W, Davidoff M. 2003. Understanding spermatogenesis is a prerequisite for treatment. Reprod Biol Endocrinol. 1:107. https://doi.org/10.1186/1477-7827-1-107
  11. Jeong YJ, Kim MK, Song HJ, Kang EJ, Ock SA, Kumar BM, Balasubramanian S, Rho GJ. 2009. Effect of alphatocopherol supplementation during boar semen cryopreservation on sperm characteristics and expression of apoptosis related genes. Cryobiology. 58:181-189. https://doi.org/10.1016/j.cryobiol.2008.12.004
  12. Jolly C, Morimoto RI. 2000. Role of the heat shock response and molecular chaperones in oncogenesis and cell death. J Natl Cancer Inst. 92:1564-1572. https://doi.org/10.1093/jnci/92.19.1564
  13. Kowalowka M, Wysocki P, Fraser L, Strzezek J. 2008. Extracellular superoxide dismutase of boar seminal plasma. Reprod Domest Anim. 43:490-496. https://doi.org/10.1111/j.1439-0531.2007.00943.x
  14. Maxwell WMC, Johnson LA. 1997. Chlortetracycline analysis of boar spermatozoa after incubation, flow cytometric sorting, cooling, or cryopreservation. Mol Reprod Dev. 46:408-418. https://doi.org/10.1002/(SICI)1098-2795(199703)46:3<408::AID-MRD21>3.0.CO;2-T
  15. Muino-Blanco T, Perez-Pe R, Cebrian-Perez JA. 2008. Seminal plasma proteins and sperm resistance to stress. Reprod Domest Anim. 4:18-31.
  16. Naz RK, Rajesh PB. 2004. Role of tyrosine phosphorylation in sperm capacitation/acrosome reaction (Review). Reprod Biol Endocrinol. 2:75. https://doi.org/10.1186/1477-7827-2-75
  17. Nover L. 1991. Heat shock response. Boca Raton, FL: CRC Press.
  18. Okado-Matsumoto A, Fridovich I. 2001. Subcellular distribution of superoxide dismutases (SOD) in rat liver: Cu, Zn-SOD in mitochondria. J Biol Chem. 276:38388-38393. https://doi.org/10.1074/jbc.M105395200
  19. Rockett JC, Mapp FL, Garges JB, Luft JC, Mori C, Dix DJ. 2001. Effects of hyperthermia on spermatogenesis, apoptosis, gene expression, and fertility in adult male mice. Biol Reprod. 65:229-239. https://doi.org/10.1095/biolreprod65.1.229
  20. Setchell BP, D'Occhio MJ, Hall MJ, Laurie MS, Tucker MJ, Zupp JL. 1988. Is embryonic mortality increased in normal female rats mated to subfertile males? J Reprod Fertil. 82:567-574. https://doi.org/10.1530/jrf.0.0820567
  21. Setchell BP, Ekpe G, Zupp JL, Surani MA. 1998. Transient retardation in embryo growth in normal female mice made pregnant by males whose testes had been heated. Hum Reprod. 13:342-347. https://doi.org/10.1093/humrep/13.2.342
  22. Takayama S, Reed JC, Homa S. 2003. Heat-shock proteins as regulators of apoptosis. Oncogene. 22:9041-9047. https://doi.org/10.1038/sj.onc.1207114
  23. Wang L, Zhu YF, Guo XJ, Huo R, Ma X, Lin M, Zhou ZM, Sha JH. 2005. A two-dimensional electrophoresis reference map of human ovary. J Mol Med. 83:812-821. https://doi.org/10.1007/s00109-005-0676-y
  24. Watson PF. 2000. The causes of reduced fertility with cryopreserved semen. Anim Reprod Sci. 60:481-492. https://doi.org/10.1016/S0378-4320(00)00099-3
  25. WHO. 1999. Laboratory manual for the examination of human semen and semen-cervical mucus interaction. 4th ed. New York: Cambridge University Press. p. 55-62.
  26. Zhu Y, Cui Y, Guo X, Wang L, Bi Y, Hu Y, Zhao X, Liu Q, Huo R, Lin M, et al. 2006. Proteomic analysis of effect of hyperthermia on spermatogenesis in adult male mice. J Proteome Res. 5:2217-2225. https://doi.org/10.1021/pr0600733

Cited by

  1. Duzhong Butiansu Prescription Improves Heat Stress-Induced Spermatogenic Dysfunction by Regulating Sperm Formation and Heat Stress Pathway vol.2020, pp.None, 2010, https://doi.org/10.1155/2020/6723204
  2. Whole‐body heat exposure causes developmental stage‐specific apoptosis of male germ cells vol.87, pp.6, 2010, https://doi.org/10.1002/mrd.23348