DOI QR코드

DOI QR Code

Inhibitors of apoptosis: expression and regulation in the endometrium during the estrous cycle and at the maternal-conceptus interface during pregnancy in pigs

  • Yoo, Inkyu (Department of Biological Science and Technology, Yonsei University) ;
  • Jung, Wonchul (Department of Biological Science and Technology, Yonsei University) ;
  • Lee, Soohyung (Department of Biological Science and Technology, Yonsei University) ;
  • Cheon, Yugyeong (Department of Biological Science and Technology, Yonsei University) ;
  • Ka, Hakhyun (Department of Biological Science and Technology, Yonsei University)
  • Received : 2021.07.07
  • Accepted : 2021.09.05
  • Published : 2022.04.01

Abstract

Objective: Caspase-mediated apoptosis plays a crucial role in the regulation of endometrial and placental function in females. Caspase activity is tightly controlled by members of the inhibitors of apoptosis proteins (IAPs) family. However, the expression and regulation of IAPs at the maternal-conceptus interface has not been studied in pigs. Therefore, we determined the expression of IAP family members baculovirus IAP repeat-containing 1 (BIRC1) to BIRC6 at the maternal-conceptus interface in pigs. Methods: We obtained endometrial tissues from pigs at various stages of the estrous cycle and pregnancy, conceptus tissues during early pregnancy, and chorioallantoic tissues during mid- to late pregnancy and analyzed the expression of IAPs. Furthermore, we determined the effects of the steroid hormones estradiol-17β (E2) and progesterone on the expression of IAPs in endometrial explant tissue cultures. Results: During the estrous cycle, BIRC2 and BIRC5 expression varied cyclically, and during pregnancy, endometrial BIRC1, BIRC2, BIRC3, BIRC4, and BIRC5 expression varied in a stage-specific manner. Conceptus and chorioallantoic tissues also expressed IAPs during pregnancy. The BIRC2 and BIR3 mRNAs were localized to luminal epithelial cells, and BIRC4 proteins to glandular epithelial cells in the endometrium. Exposure of endometrial tissues to E2 increased the expression of BIRC6, while progesterone increased the expression of BIRC1, BIRC4, and BIRC6 in a dose-dependent manner. Conclusion: These results indicated that IAPs were expressed in the endometrium during the estrous cycle and at the maternal-conceptus interface during pregnancy in a stage-specific manner. In addition, steroid hormones were found to be responsible for the expression of some IAPs in pigs. Together, the results suggested that IAPs may play important roles in endometrial and placental functions by regulating caspase action and apoptosis at the maternal-conceptus interface.

Keywords

Acknowledgement

This study was supported by the National Research Foundation grant funded by the Korean Government (NRF-2019R1 A2C1004670 to HK and 2020R1A6A3A01098213 to IY), Republic of Korea.

References

  1. Elmore S. Apoptosis: a review of programmed cell death. Toxicol Pathol 2007;35:495-516. https://doi.org/10.1080/01926230701320337
  2. Harada T, Kaponis A, Iwabe T, et al. Apoptosis in human endometrium and endometriosis. Hum Reprod Update 2004;10:29-38. https://doi.org/10.1093/humupd/dmh007
  3. Cohen M, Wuillemin C, Irion O, Bischof P. Role of decidua in trophoblastic invasion. Neuro Endocrinol Lett 2010;31:193-7.
  4. Annie L, Gurusubramanian G, Roy VK. Estrogen and progesterone dependent expression of visfatin/NAMPT regulates proliferation and apoptosis in mice uterus during estrous cycle. J Steroid Biochem Mol Biol 2019;185:225-36. https://doi.org/10.1016/j.jsbmb.2018.09.010
  5. Pampfer S, Donnay I. Apoptosis at the time of embryo implantation in mouse and rat. Cell Death Differ 1999;6:533-45. https://doi.org/10.1038/sj.cdd.4400516
  6. Tan J, Raja S, Davis MK, Tawfik O, Dey SK, Das SK. Evidence for coordinated interaction of cyclin D3 with p21 and cdk6 in directing the development of uterine stromal cell decidualization and polyploidy during implantation. Mech Dev 2002;111:99-113. https://doi.org/10.1016/s0925-4773(01)00614-1
  7. Okano A, Ogawa H, Takahashi H, Geshi M. Apoptosis in the porcine uterine endometrium during the estrous cycle, early pregnancy and post partum. J Reprod Dev 2007;53:923-30. https://doi.org/10.1262/jrd.18139
  8. Yoo I, Kye YC, Han J, et al. Uterine epithelial expression of the tumor necrosis factor superfamily: a strategy for immune privilege during pregnancy in a true epitheliochorial placentation species. Biol Reprod 2020;102:828-42. https://doi.org/10.1093/biolre/ioz233
  9. Han J, Gu MJ, Yoo I, et al. Analysis of cysteine-X-cysteine motif chemokine ligands 9, 10, and 11, their receptor CXCR3, and their possible role on the recruitment of immune cells at the maternal-conceptus interface in pigs. Biol Reprod 2017;97:69-80. https://doi.org/10.1093/biolre/iox074
  10. McIlwain DR, Berger T, Mak TW. Caspase functions in cell death and disease. Cold Spring Harb Perspect Biol 2013;5:a008656. https://doi.org/10.1101/cshperspect.a008656
  11. Green DR, Llambi F. Cell death signaling. Cold Spring Harb Perspect Biol 2015;7:a006080. https://doi.org/10.1101/cshperspect.a006080
  12. Galluzzi L, Lopez-Soto A, Kumar S, Kroemer G. Caspases connect cell-death signaling to organismal homeostasis. Immunity 2016;44:221-31. https://doi.org/10.1016/j.immuni.2016.01.020
  13. D'Arcy MS. Cell death: a review of the major forms of apoptosis, necrosis and autophagy. Cell Biol Int 2019;43:582-92. https://doi.org/10.1002/cbin.11137
  14. Jung W, Yoo I, Han J, et al. Expression of caspases in the pig endometrium throughout the estrous cycle and at the maternal-conceptus interface during pregnancy and regulation by steroid hormones and cytokines. Front Vet Sci 2021;8:641916. https://doi.org/10.3389/fvets.2021.641916
  15. Deveraux QL, Reed JC. IAP family proteins-suppressors of apoptosis. Genes Dev 1999;13:239-52. https://doi.org/10.1101/gad.13.3.239
  16. Kenneth NS, Duckett CS. IAP proteins: regulators of cell migration and development. Curr Opin Cell Biol 2012;24:871-5. https://doi.org/10.1016/j.ceb.2012.11.004
  17. Saleem M, Qadir MI, Perveen N, et al. Inhibitors of apoptotic proteins: new targets for anticancer therapy. Chem Biol Drug Des 2013;82:243-51. https://doi.org/10.1111/cbdd.12176
  18. Silke J, Meier P. Inhibitor of apoptosis (IAP) proteins-modulators of cell death and inflammation. Cold Spring Harb Perspect Biol 2013;5:a008730. https://doi.org/10.1101/cshperspect.a008730
  19. Hu S, Yang X. Cellular inhibitor of apoptosis 1 and 2 are ubiquitin ligases for the apoptosis inducer Smac/DIABLO. J Biol Chem 2003;278:10055-60. https://doi.org/10.1074/jbc.M207197200
  20. Eckelman BP, Salvesen GS, Scott FL. Human inhibitor of apoptosis proteins: why XIAP is the black sheep of the family. EMBO Rep 2006;7:988-94. https://doi.org/10.1038/sj.embor.7400795
  21. Shiozaki EN, Shi Y. Caspases, IAPs and Smac/DIABLO: mechanisms from structural biology. Trends Biochem Sci 2004;29:486-94. https://doi.org/10.1016/j.tibs.2004.07.003
  22. Deveraux QL, Takahashi R, Salvesen GS, Reed JC. X-linked IAP is a direct inhibitor of cell-death proteases. Nature 1997;388:300-4. https://doi.org/10.1038/40901
  23. Watanabe A, Taniguchi F, Izawa M, et al. The role of survivin in the resistance of endometriotic stromal cells to drug-induced apoptosis. Hum Reprod 2009;24:3172-9. https://doi.org/10.1093/humrep/dep305
  24. Taniguchi F, Higaki H, Izawa M, et al. The cellular inhibitor of apoptosis protein-2 is a possible target of novel treatment for endometriosis. Am J Reprod Immunol 2014;71:278-85. https://doi.org/10.1111/aji.12193
  25. Neubauer NL, Ward EC, Patel P, et al. Progesterone receptor-B induction of BIRC3 protects endometrial cancer cells from AP1-59-mediated apoptosis. Horm Cancer 2011;2:170-81. https://doi.org/10.1007/s12672-011-0065-7
  26. Ka H, Hunt JS. Temporal and spatial patterns of expression of inhibitors of apoptosis in human placentas. Am J Pathol 2003;163:413-22. https://doi.org/10.1016/S0002-9440(10)63671-1
  27. Gill RM, Hunt JS. Soluble receptor (DcR3) and cellular inhibitor of apoptosis-2 (cIAP-2) protect human cytotrophoblast cells against LIGHT-mediated apoptosis. Am J Pathol 2004;165:309-17. https://doi.org/10.1016/S0002-9440(10)63298-1
  28. Shooner C, Caron PL, Frechette-Frigon G, Leblanc V, Dery MC, Asselin E. TGF-beta expression during rat pregnancy and activity on decidual cell survival. Reprod Biol Endocrinol 2005;3:20. https://doi.org/10.1186/1477-7827-3-20
  29. Straszewski-Chavez SL, Abrahams VM, Aldo PB, Romero R, Mor G. AKT controls human first trimester trophoblast cell sensitivity to FAS-mediated apoptosis by regulating XIAP expression. Biol Reprod 2010;82:146-52. https://doi.org/10.1095/biolreprod.109.078972
  30. Fest S, Brachwitz N, Schumacher A, et al. Supporting the hypothesis of pregnancy as a tumor: survivin is upregulated in normal pregnant mice and participates in human trophoblast proliferation. Am J Reprod Immunol 2008;59:75-83. https://doi.org/10.1111/j.1600-0897.2007.00557.x
  31. Lee S, Yoo I, Han J, Ka H. Antimicrobial peptides cathelicidin, PMAP23, and PMAP37: Expression in the endometrium throughout the estrous cycle and at the maternal-conceptus interface during pregnancy and regulation by steroid hormones and calcitriol in pigs. Theriogenology 2021;160:1-9. https://doi.org/10.1016/j.theriogenology.2020.10.034
  32. Seo H, Kim M, Choi Y, Lee CK, Ka H. Analysis of lysophosphatidic acid (LPA) receptor and LPA-induced endometrial prostaglandin-endoperoxide synthase 2 expression in the porcine uterus. Endocrinology 2008;149:6166-75. https://doi.org/10.1210/en.2008-0354
  33. Ka H, Seo H, Choi Y, Yoo I, Han J. Endometrial response to conceptus-derived estrogen and interleukin-1beta at the time of implantation in pigs. J Anim Sci Biotechnol 2018;9:44. https://doi.org/10.1186/s40104-018-0259-8
  34. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods 2001;25:402-8. https://doi.org/10.1006/meth.2001.1262
  35. Braissant O, Wahli W. Differential expression of peroxisome proliferator-activated receptor-alpha, -beta, and -gamma during rat embryonic development. Endocrinology 1998;139:2748-54. https://doi.org/10.1210/endo.139.6.6049
  36. Gruslin A, Qiu Q, Tsang BK. X-linked inhibitor of apoptosis protein expression and the regulation of apoptosis during human placental development. Biol Reprod 2001;64:1264-72. https://doi.org/10.1095/biolreprod64.4.1264
  37. Graber TE, Holcik M. Distinct roles for the cellular inhibitors of apoptosis proteins 1 and 2. Cell Death Dis 2011;2:e135. https://doi.org/10.1038/cddis.2011.20
  38. Tamm I, Wang Y, Sausville E, et al. IAP-family protein survivin inhibits caspase activity and apoptosis induced by Fas (CD95), Bax, caspases, and anticancer drugs. Cancer Res 1998;58:5315-20.
  39. Garcia MG, Tirado-Gonzalez I, Handjiski B, et al. High expression of survivin and down-regulation of Stat-3 characterize the feto-maternal interface in failing murine pregnancies during the implantation period. Placenta 2007;28:650-7. https://doi.org/10.1016/j.placenta.2006.09.010
  40. Shalini S, Dorstyn L, Dawar S, Kumar S. Old, new and emerging functions of caspases. Cell Death Differ 2015;22:526-39. https://doi.org/10.1038/cdd.2014.216
  41. Burghardt RC, Johnson GA, Jaeger LA, et al. Integrins and extracellular matrix proteins at the maternal-fetal interface in domestic animals. Cells Tissues Organs 2002;172:202-17. https://doi.org/10.1159/000066969
  42. Leblanc V, Dery MC, Shooner C, Asselin E. Opposite regulation of XIAP and Smac/DIABLO in the rat endometrium in response to 17beta-estradiol at estrus. Reprod Biol Endocrinol 2003;1:59. https://doi.org/10.1186/1477-7827-1-59
  43. Caron PL, Frechette-Frigon G, Shooner C, Leblanc V, Asselin E. Transforming growth factor beta isoforms regulation of Akt activity and XIAP levels in rat endometrium during estrous cycle, in a model of pseudopregnancy and in cultured decidual cells. Reprod Biol Endocrinol 2009;7:80. https://doi.org/10.1186/1477-7827-7-80
  44. Nabilsi NH, Broaddus RR, McCampbell AS, et al. Sex hormone regulation of survivin gene expression. J Endocrinol 2010;207:237-43. https://doi.org/10.1677/JOE-10-0128