DOI QR코드

DOI QR Code

Detrimental effects of lipopolysaccharides on maturation of bovine oocytes

  • Zhao, Shanjiang (Embryo Biotechnology and Reproduction Laboratory, Institute of Animal Science, Chinese Academy of Agricultural Sciences) ;
  • Pang, Yunwei (Embryo Biotechnology and Reproduction Laboratory, Institute of Animal Science, Chinese Academy of Agricultural Sciences) ;
  • Zhao, Xueming (Embryo Biotechnology and Reproduction Laboratory, Institute of Animal Science, Chinese Academy of Agricultural Sciences) ;
  • Du, Weihua (Embryo Biotechnology and Reproduction Laboratory, Institute of Animal Science, Chinese Academy of Agricultural Sciences) ;
  • Hao, Haisheng (Embryo Biotechnology and Reproduction Laboratory, Institute of Animal Science, Chinese Academy of Agricultural Sciences) ;
  • Zhu, Huabin (Embryo Biotechnology and Reproduction Laboratory, Institute of Animal Science, Chinese Academy of Agricultural Sciences)
  • 투고 : 2018.07.20
  • 심사 : 2018.10.16
  • 발행 : 2019.08.01

초록

Objective: Gram-negative bacteria lipopolysaccharide (LPS) has been reported to be associated with uterine impairment, embryonic resorption, ovarian dysfunction, and follicle retardation. Here, we aimed to investigate the toxic effects of LPS on the maturation ability and parthenogenetic developmental competence of bovine oocytes. Methods: First, we developed an in vitro model to study the response of bovine cumulusoocyte complexes (COCs) to LPS stress. After incubating germinal vesicle COCs in $10{\mu}g/mL$ of LPS, we analyzed the following three aspects: the expression levels of the LPS receptor toll-like receptor 4 (TLR4) in COCs, activities of intracellular signaling protein p38 mitogen-activated protein kinase (p38 MAPK) and nuclear factor-kappa B (NF-${\kappa}B$); and the concentrations of interleukin (IL)-$1{\beta}$, tumor necrosis factor (TNF)-${\alpha}$, and IL-6. Furthermore, we determined the effects of LPS on the maturation ability and parthenogenetic developmental competence of bovine oocytes. Results: The results revealed that LPS treatment significantly elevated TLR4 mRNA and protein expression levels in COCs. Exposure of COCs to LPS also resulted in a marked increase in activity of the intracellular signaling protein p-p38 MAPK and NF-${\kappa}B$. Furthermore, oocytes cultured in maturation medium containing LPS had significantly higher concentrations of the proinflammatory cytokines IL-$1{\beta}$, TNF-${\alpha}$, and IL-6. LPS exposure significantly decreased the first polar body extrusion rate. The cytoplasmic maturation, characterized by polar body extrusion and distribution of peripheral cortical granules, was significantly impaired in LPS-treated oocytes. Moreover, LPS exposure significantly increased intracellular reactive oxygen species levels and the relative mRNA abundance of the antioxidants thioredoxin (Trx), Trx2, and peroxiredoxin 1 in oocytes. Moreover, the early apoptotic rate and the release of cytochrome C were significantly increased in response to LPS. The cleavage, morula, and blastocyst formation rates were significantly lower in parthenogenetically activated oocytes exposed to LPS, while the incidence of apoptotic nuclei in blastocysts was significantly increased. Conclusion: Together, these results provide an underlying mechanism by which LPS impairs maturation potential in bovine oocytes.

키워드

참고문헌

  1. Sheldon IM, Noakes DE, Rycroft AN, Pfeiffer DU, Dobson H. Influence of uterine bacterial contamination after parturition on ovarian dominant follicle selection and follicle growth and function in cattle. Reproduction 2002;123:837-45. https://doi.org/10.1530/rep.0.1230837
  2. Williams EJ, Sibley K, Miller AN, et al. The effect of Escherichia coli lipopolysaccharide and tumour necrosis factor alpha on ovarian function. Am J Reprod Immunol 2008;60:462-73. https://doi.org/10.1111/j.1600-0897.2008.00645.x
  3. Herath S, Williams EJ, Lilly ST, et al. Ovarian follicular cells have innate immune capabilities that modulate their endocrine function. Reproduction 2007;134:683-93. https://doi.org/10.1530/REP-07-0229
  4. Borsberry S, Dobson H. Periparturient diseases and their effect on reproductive performance in five dairy herds. Vet Rec 1989;124:217-9. https://doi.org/10.1136/vr.124.9.217
  5. LeBlanc SJ, Duffield TF, Leslie KE, et al. Defining and diagnosing postpartum clinical endometritis and its impact on reproductive performance in dairy cows. J Dairy Sci 2002;85:2223-36. https://doi.org/10.3168/jds.S0022-0302(02)74302-6
  6. Karsch FJ, Battaglia DF, Breen KM, Debus N, Harris TG. Mechanisms for ovarian cycle disruption by immune/inflammatory stress. Stress 2002;5:101-12. https://doi.org/10.1080/10253890290027868
  7. Sheldon IM, Cronin J, Goetze L, Donofrio G, Schuberth HJ. Defining postpartum uterine disease and the mechanisms of infection and immunity in the female reproductive tract in cattle. Biol Reprod 2009;81:1025-32. https://doi.org/10.1095/biolreprod.109.077370
  8. Ibrahim S, Salilew-Wondim D, Rings F, et al. Expression pattern of inflammatory response genes and their regulatory micrornas in bovine oviductal cells in response to lipopolysaccharide: implication for early embryonic development. PLoS One 2015;10:e0119388. https://doi.org/10.1371/journal.pone.0119388
  9. Raetz CR, Whitfield C. Lipopolysaccharide endotoxins. Annu Rev Biochem 2002;71:635-700. https://doi.org/10.1146/annurev.biochem.71.110601.135414
  10. Beutler B. Inferences, questions and possibilities in Toll-like receptor signalling. Nature 2004;430:257-63. https://doi.org/10.1038/nature02761
  11. Doyle SL, O'Neill LA. Toll-like receptors: from the discovery of NFkappaB to new insights into transcriptional regulations in innate immunity. Biochem Pharmacol 2006;72:1102-13. https://doi.org/10.1016/j.bcp.2006.07.010
  12. Blum JW, Dosogne H, Hoeben D, et al. Tumor necrosis factor-alpha and nitrite/nitrate responses during acute mastitis induced by Escherichia coli infection and endotoxin in dairy cows. Domest Anim Endocrinol 2000;19:223-35. https://doi.org/10.1016/S0739-7240(00)00079-5
  13. Bromfield JJ, Sheldon IM. Lipopolysaccharide initiates inflammation in bovine granulosa cells via the TLR4 pathway and perturbs oocyte meiotic progression in vitro. Endocrinology 2011;152:5029-40. https://doi.org/10.1210/en.2011-1124
  14. Raza H, John A, Shafarin J. Potentiation of LPS-induced apoptotic cell death in human hepatoma HepG2 cells by aspirin via ROS and mitochondrial dysfunction: protection by N-acetyl cysteine. PLoS One 2016;11:e0159750. https://doi.org/10.1371/journal.pone.0159750
  15. Coticchio G, Dal Canto M, Mignini Renzini M, et al. Oocyte maturation: gamete-somatic cells interactions, meiotic resumption, cytoskeletal dynamics and cytoplasmic reorganization. Hum Reprod Update 2015;21:427-54. https://doi.org/10.1093/humupd/dmv011
  16. Coticchio G, Sereni E, Serrao L, Mazzone S, Iadarola I, Borini A. What criteria for the definition of oocyte quality? Ann NY Acad Sci 2004;1034:132-44. https://doi.org/10.1196/annals.1335.016
  17. Platz-Christensen JJ, Mattsby-Baltzer I, Thomsen P, Wiqvist N. Endotoxin and interleukin-1 alpha in the cervical mucus and vaginal fluid of pregnant women with bacterial vaginosis. Am J Obstet Gynecol 1993;169:1161-6. https://doi.org/10.1016/0002-9378(93)90274-M
  18. Bannerman DD, Paape MJ, Hare WR, Sohn EJ. Increased levels of LPS-binding protein in bovine blood and milk following bacterial lipopolysaccharide challenge. J Dairy Sci 2003;86:3128-37. https://doi.org/10.3168/jds.S0022-0302(03)73914-9
  19. Thouas GA, Korfiatis NA, French AJ, Jones GM, Trounson AO. Simplified technique for differential staining of inner cell mass and trophectoderm cells of mouse and bovine blastocysts. Reprod Biomed Online 2001;3:25-9. https://doi.org/10.1016/S1472-6483(10)61960-8
  20. Shimada M, Hernandez-Gonzalez I, Gonzalez-Robanya I, Richards JS. Induced expression of pattern recognition receptors in cumulus oocyte complexes: novel evidence for innate immune-like functions during ovulation. Mol Endocrinol 2006;20:3228-39. https://doi.org/10.1210/me.2006-0194
  21. Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell 2010;140:805-20. https://doi.org/10.1016/j.cell.2010.01.022
  22. Roth Z, Asaf S, Furman O, et al. Subclinical mastitis disrupts oocyte cytoplasmic maturation in association with reduced developmental competence and impaired gene expression in preimplantation bovine embryos. Reprod Fertil Dev 2015 Apr 20 [Epub]. https://doi.org/10.1071/RD14431
  23. Asaf S, Leitner G, Furman O, et al. Effects of Escherichia coliand Staphylococcus aureus-induced mastitis in lactating cows on oocyte developmental competence. Reproduction 2014;147:33-43. https://doi.org/10.1530/REP-13-0383
  24. Jaja-Chimedza A, Gantar M, Mayer GD, Gibbs PD, Berry JP. Effects of cyanobacterial lipopolysaccharides from microcystis on glutathione-based detoxification pathways in the zebrafish (Danio rerio) embryo. Toxins (Basel) 2012;4:390-404. https://doi.org/10.3390/toxins4060390
  25. Sagar S, Kumar P, Behera RR, Pal A. Effects of CEES and LPS synergistically stimulate oxidative stress inactivates OGG1 signaling in macrophage cells. J Hazard Mater 2014;278:236-49. https://doi.org/10.1016/j.jhazmat.2014.05.096
  26. Qin T, Yin Y, Yu Q, Yang Q. Bursopentin (BP5) protects dendritic cells from lipopolysaccharide-induced oxidative stress for immunosuppression. PLoS One 2015;10:e0117477. https://doi.org/10.1371/journal.pone.0117477
  27. Li X, Wang X, Zheng M, Luan QX. Mitochondrial reactive oxygen species mediate the lipopolysaccharide-induced pro-inflammatory response in human gingival fibroblasts. Exp Cell Res 2016;347:212-21. https://doi.org/10.1016/j.yexcr.2016.08.007
  28. Fu C, Dai X, Yang Y, Lin M, Cai Y, Cai S. Dexmedetomidine attenuates lipopolysaccharide-induced acute lung injury by inhibiting oxidative stress, mitochondrial dysfunction and apoptosis in rats. Mol Med Rep 2017;15:131-8. https://doi.org/10.3892/mmr.2016.6012
  29. Zhao L, Chen YH, Wang H, et al. Reactive oxygen species contribute to lipopolysaccharide-induced teratogenesis in mice. Toxicol Sci 2008;103:149-57. https://doi.org/10.1093/toxsci/kfn027

피인용 문헌

  1. Effects of increasing lipopolysaccharide concentrations on in vitro developmental competence of ovine oocytes vol.17, pp.2, 2020, https://doi.org/10.1590/1984-3143-ar2019-0125
  2. In vitro and in vivo pharmacokinetic characterization of LMT-28 as a novel small molecular interleukin-6 inhibitor vol.33, pp.4, 2019, https://doi.org/10.5713/ajas.19.0463