Animal Reproduction (AR)
https://animal-reproduction.org/article/doi/10.1590/1984-3143-AR2024-0042
Animal Reproduction (AR)
Thematic Section: 37th Annual Meeting of the Brazilian Embryo Technology Society (SBTE)

Interaction of semen with female reproductive tract tissues: what we know, what we guess and what we need to do

John James Bromfield

Downloads: 2
Views: 142

Abstract

For nearly 100 years the postcoital inflammatory response has been described in the female reproductive tract of rodents. Since the 1950’s this observation has been made in a number of animals including humans and domestic species. Yet pregnancy can be initiated and maintained by using embryo transfer which bypasses insemination and the related postcoital inflammatory response. Thus, the role of semen exposure beyond sperm transport and subsequent postcoital inflammatory response in female reproductive tissues has yet to be given a true physiological purpose. Historically the postcoital inflammatory response of female tissues was suggested to remove spermatozoa and male derived pathogens from the female reproductive tract. More recently, semen exposure and the postcoital inflammatory response have been suggested to play a role in long-term preparation of the maternal immune system to the semi-allogeneic pregnancy, ancillary support of the preimplantation embryo, and potentially fetal programing that improves pregnancy outcomes, while the absence or inappropriate postcoital inflammation has been suggested to contribute to pregnancy complications. Although the postcoital inflammatory response has been robustly characterized, the evidence for its role in promoting positive pregnancy outcomes or reducing pregnancy complications remains tenuous. This manuscript is designed to balance the information we know regarding semen exposure and postcoital inflammation in various animal systems, with the information we perceive to be factual but perhaps not yet fully tested, along with the data we have yet to generate if we intend to postulate a physiological purpose of the postcoital inflammatory response to pregnancy outcomes.

Keywords

postcoital inflammation, semen, seminal fluid

References

Aluvihare VR, Kallikourdis M, Betz AG. Regulatory T cells mediate maternal tolerance to the fetus. Nat Immunol. 2004;5(3):266-71. http://doi.org/10.1038/ni1037. PMid:14758358.

Barker DJ. Mothers, babies and health in later life. Edinburgh: Churchill Livingstone; 1998.

Beer AE, Billingham RE. Host responses to intra-uterine tissue, cellular and fetal allografts. J Reprod Fertil Suppl. 1974;21:59-88.

Block J, Hansen PJ, Loureiro B, Bonilla L. Improving post-transfer survival of bovine embryos produced in vitro: actions of insulin-like growth factor-1, colony stimulating factor-2 and hyaluronan. Theriogenology. 2011;76(9):1602-9. http://doi.org/10.1016/j.theriogenology.2011.07.025. PMid:21890189.

Bromfield JJ, Schjenken JE, Chin PY, Care AS, Jasper MJ, Robertson SA. Maternal tract factors contribute to paternal seminal fluid impact on metabolic phenotype in offspring. Proc Natl Acad Sci USA. 2014;111(6):2200-5. http://doi.org/10.1073/pnas.1305609111. PMid:24469827.

Bromfield JJ. A role for seminal plasma in modulating pregnancy outcomes in domestic species. Reproduction. 2016;152(6):R223-32. http://doi.org/10.1530/REP-16-0313. PMid:27601714.

Bromfield JJ. Review: the potential of seminal fluid mediated paternal-maternal communication to optimise pregnancy success. Animal. 2018;12(s1):s104-9. http://doi.org/10.1017/S1751731118000083. PMid:29455706.

Bromfield JJ. Seminal fluid and reproduction: much more than previously thought. J Assist Reprod Genet. 2014;31(6):627-36. http://doi.org/10.1007/s10815-014-0243-y. PMid:24830788.

Coulam CB, Stern JJ. Effect of seminal plasma on implantation rates. Early Pregnancy. 1995;1(1):33-6. PMid:9363233.

Cox GF, Burger J, Lip V, Mau UA, Sperling K, Wu BL, Horsthemke B. Intracytoplasmic sperm injection may increase the risk of imprinting defects. Am J Hum Genet. 2002;71(1):162-4. http://doi.org/10.1086/341096. PMid:12016591.

Fanchin R, Harmas A, Benaoudia F, Lundkvist U, Olivennes F, Frydman R. Microbial flora of the cervix assessed at the time of embryo transfer adversely affects in vitro fertilization outcome. Fertil Steril. 1998a;70(5):866-70. http://doi.org/10.1016/S0015-0282(98)00277-5. PMid:9806568.

Fanchin R, Righini C, Olivennes F, Taylor S, de Ziegler D, Frydman R. Uterine contractions at the time of embryo transfer alter pregnancy rates after in-vitro fertilization. Hum Reprod. 1998b;13(7):1968-74. http://doi.org/10.1093/humrep/13.7.1968. PMid:9740459.

Fantini MC, Becker C, Tubbe I, Nikolaev A, Lehr HA, Galle PR, Neurath MF. Transforming growth factor beta induced FoxP3+ regulatory T cells suppress Th1 mediated experimental colitis. Gut. 2006;55(5):671-80. http://doi.org/10.1136/gut.2005.072801. PMid:16162681.

Faulkner LC, Hopwood ML, Wiltbank JN. Seminal vesiculectomy in bulls. II. Seminal characteristics and breeding trials. J Reprod Fertil. 1968;16(2):179-82. http://doi.org/10.1530/jrf.0.0160179. PMid:5660277.

Fernandez-Fuertes B, Sanchez JM, Bages-Arnal S, McDonald M, Yeste M, Lonergan P. Species-specific and collection method-dependent differences in endometrial susceptibility to seminal plasma-induced RNA degradation. Sci Rep. 2019;9(1):15072. http://doi.org/10.1038/s41598-019-51413-4. PMid:31636362.

Fumuso E, Giguere S, Wade J, Rogan D, Videla-Dorna I, Bowden RA. Endometrial IL-1beta, IL-6 and TNF-alpha, mRNA expression in mares resistant or susceptible to post-breeding endometritis. Effects of estrous cycle, artificial insemination and immunomodulation. Vet Immunol Immunopathol. 2003;96(1-2):31-41. http://doi.org/10.1016/S0165-2427(03)00137-5. PMid:14522132.

Guerin LR, Moldenhauer LM, Prins JR, Bromfield JJ, Hayball JD, Robertson SA. Seminal fluid regulates accumulation of FOXP3+ regulatory T cells in the preimplantation mouse uterus through expanding the FOXP3+ cell pool and CCL19-mediated recruitment. Biol Reprod. 2011;85(2):397-408. http://doi.org/10.1095/biolreprod.110.088591. PMid:21389340.

Hansen M, Bower C, Milne E, de Klerk N, Kurinczuk JJ. Assisted reproductive technologies and the risk of birth defects--a systematic review. Hum Reprod. 2005;20(2):328-38. http://doi.org/10.1093/humrep/deh593. PMid:15567881.

Hansen M, Kurinczuk JJ, Bower C, Webb S. The risk of major birth defects after intracytoplasmic sperm injection and in vitro fertilization. N Engl J Med. 2002;346(10):725-30. http://doi.org/10.1056/NEJMoa010035. PMid:11882727.

Hunt JS, Robertson SA. Uterine macrophages and environmental programming for pregnancy success. J Reprod Immunol. 1996;32(1):1-25. http://doi.org/10.1016/S0165-0378(96)88352-5. PMid:8953517.

Ibrahim LA, Rizo JA, Fontes PLP, Lamb GC, Bromfield JJ. Seminal plasma modulates expression of endometrial inflammatory meditators in the bovine. Biol Reprod. 2019;100(3):660-71. http://doi.org/10.1093/biolre/ioy226. PMid:30329018.

Jiwakanon J, Berg M, Persson E, Fossum C, Dalin AM. Cytokine expression in the gilt oviduct: effects of seminal plasma, spermatozoa and extender after insemination. Anim Reprod Sci. 2010;119(3-4):244-57. http://doi.org/10.1016/j.anireprosci.2010.01.005. PMid:20171026.

Kannampuzha-Francis J, Denicol AC, Loureiro B, Kaniyamattam K, Ortega MS, Hansen PJ. Exposure to colony stimulating factor 2 during preimplantation development increases postnatal growth in cattle. Mol Reprod Dev. 2015;82(11):892-7. http://doi.org/10.1002/mrd.22533. PMid:26227079.

Klonoff-Cohen HS, Savitz DA, Celafo RC, McCann MF. An epidemiologic study of contraception and preeclampsia. JAMA. 1989;262(22):3143-7. http://doi.org/10.1001/jama.1989.03430220066032. PMid:2810672.

Konkel JE, Zhang D, Zanvit P, Chia C, Zangarle-Murray T, Jin W, Wang S, Chen W. Transforming growth factor-beta signaling in regulatory T cells controls T helper-17 cells and tissue-specific immune responses. Immunity. 2017;46(4):660-74. http://doi.org/10.1016/j.immuni.2017.03.015. PMid:28423340.

Kurinczuk JJ, Hansen M, Bower C. The risk of birth defects in children born after assisted reproductive technologies. Curr Opin Obstet Gynecol. 2004;16(3):201-9. http://doi.org/10.1097/00001703-200406000-00002. PMid:15129049.

Lafontaine S, Labrecque R, Blondin P, Cue RI, Sirard MA. Comparison of cattle derived from in vitro fertilization, multiple ovulation embryo transfer, and artificial insemination for milk production and fertility traits. J Dairy Sci. 2023;106(6):4380-96. http://doi.org/10.3168/jds.2022-22736. PMid:37028966.

Lima FS, Risco CA, Thatcher MJ, Benzaquen ME, Archbald LF, Santos JE, Thatcher WW. Comparison of reproductive performance in lactating dairy cows bred by natural service or timed artificial insemination. J Dairy Sci. 2009;92(11):5456-66. http://doi.org/10.3168/jds.2009-2197. PMid:19841208.

Lonergan P. Review: historical and futuristic developments in bovine semen technology. Animal. 2018;12(s1):s4-18. http://doi.org/10.1017/S175173111800071X. PMid:29642968.

Loras B, Vetele F, El Malki A, Rollet J, Soufir JC, Benahmed M. Seminal transforming growth factor-beta in normal and infertile men. Hum Reprod. 1999;14(6):1534-9. http://doi.org/10.1093/humrep/14.6.1534. PMid:10357971.

Ludwig M, Katalinic A, Gross S, Sutcliffe A, Varon R, Horsthemke B. Increased prevalence of imprinting defects in patients with Angelman syndrome born to subfertile couples. J Med Genet. 2005;42(4):289-91. http://doi.org/10.1136/jmg.2004.026930. PMid:15805153.

Marey MA, Ma D, Yoshino H, Elesh IF, Zinnah MA, Fiorenza MF, Moriyasu S, Miyamoto A. Sperm induce proinflammatory responses in the uterus and peripheral blood immune cells of artificially inseminated cows. J Reprod Dev. 2023;69(2):95-102. http://doi.org/10.1262/jrd.2022-124. PMid:36775285.

Mateo-Otero Y, Sanchez JM, Recuero S, Bages-Arnal S, McDonald M, Kenny DA, Yeste M, Lonergan P, Fernandez-Fuertes B. Effect of exposure to seminal plasma through natural mating in cattle on conceptus length and gene expression. Front Cell Dev Biol. 2020;8:341. http://doi.org/10.3389/fcell.2020.00341. PMid:32478076.

Mattner PE. The distribution of spermatozoa and leucocytes in the female genital tract in goats and cattle. J Reprod Fertil. 1968;17(2):253-61. http://doi.org/10.1530/jrf.0.0170253. PMid:5749378.

Medawar P. Some immunological and endocrinological problems raised by the evolution of viviparity in vertebrates. Symp Soc Exp Biol. 1953;7:320-38.

Mitchell MH, Swanson RJ, Oehninger S. In vivo effect of leukemia inhibitory factor (LIF) and an anti-LIF polyclonal antibody on murine embryo and fetal development following exposure at the time of transcervical blastocyst transfer. Biol Reprod. 2002;67(2):460-4. http://doi.org/10.1095/biolreprod67.2.460. PMid:12135882.

Murray FA, Grifo AP Jr, Parker CF. Increased litter size in gilts by intrauterine infusion of seminal and sperm antigens before breeding. J Anim Sci. 1983;56(4):895-900. http://doi.org/10.2527/jas1983.564895x. PMid:6682857.

National Center for Chronic Disease Prevention and Health Promotion, Division of Reproductive Health [homepage on the Internet]. USA: CDC; 2024. 2020 National ART Summary; 2020 [cited 2024 Mar 30]. Available from: https://www.cdc.gov/art/reports/2020/summary.html

Nocera M, Chu TM. Characterization of latent transforming growth factor-beta from human seminal plasma. Am J Reprod Immunol. 1995;33(4):282-91. http://doi.org/10.1111/j.1600-0897.1995.tb00897.x. PMid:7546247.

O’Leary S, Armstrong DT, Robertson SA. Transforming growth factor-beta (TGFbeta) in porcine seminal plasma. Reprod Fertil Dev. 2011;23(6):748-58. http://doi.org/10.1071/RD11001. PMid:21791176.

O’Leary S, Jasper MJ, Warnes GM, Armstrong DT, Robertson SA. Seminal plasma regulates endometrial cytokine expression, leukocyte recruitment and embryo development in the pig. Reproduction. 2004;128(2):237-47. http://doi.org/10.1530/rep.1.00160. PMid:15280563.

Odhiambo JF, Poole DH, Hughes L, Dejarnette JM, Inskeep EK, Dailey RA. Pregnancy outcome in dairy and beef cattle after artificial insemination and treatment with seminal plasma or transforming growth factor beta-1. Theriogenology. 2009;72(4):566-71. http://doi.org/10.1016/j.theriogenology.2009.04.013. PMid:19501392.

Ortiz WG, Rizo JA, Carvalheira LR, Ahmed BMS, Estrada-Cortes E, Harstine BR, Bromfield JJ, Hansen PJ. Effects of intrauterine infusion of seminal plasma at artificial insemination on fertility of lactating Holstein cows. J Dairy Sci. 2019;102(7):6587-94. http://doi.org/10.3168/jds.2019-16251. PMid:31103294.

Pandya IJ, Cohen J. The leukocytic reaction of the human uterine cervix to spermatozoa. Fertil Steril. 1985;43(3):417-21. http://doi.org/10.1016/S0015-0282(16)48442-6. PMid:3979580.

Parr MB, Parr EL. Antigen recognition in the female reproductive tract: I. Uptake of intraluminal protein tracers in the mouse vagina. J Reprod Immunol. 1990;17(2):101-14. http://doi.org/10.1016/0165-0378(90)90029-6. PMid:2338672.

Recuero S, Sanchez JM, Mateo-Otero Y, Bages-Arnal S, McDonald M, Behura SK, Spencer TE, Kenny DA, Yeste M, Lonergan P, Fernandez-Fuertes B. Mating to intact, but not vasectomized, males elicits changes in the endometrial transcriptome: insights from the bovine model. Front Cell Dev Biol. 2020;8:547. http://doi.org/10.3389/fcell.2020.00547. PMid:32766237.

Rizo JA, Ibrahim LA, Molinari PCC, Harstine B, Piersanti RL, Bromfield JJ. Effect of seminal plasma or transforming growth factor on bovine endometrial cells. Reproduction. 2019;158(6):529-41. http://doi.org/10.1530/REP-19-0421. PMid:31645014.

Robertson SA, Guerin LR, Bromfield JJ, Branson KM, Ahlstrom AC, Care AS. Seminal fluid drives expansion of the CD4+CD25+ T regulatory cell pool and induces tolerance to paternal alloantigens in mice. Biol Reprod. 2009;80(5):1036-45. http://doi.org/10.1095/biolreprod.108.074658. PMid:19164169.

Robertson SA, Ingman WV, O’Leary S, Sharkey DJ, Tremellen KP. Transforming growth factor beta--a mediator of immune deviation in seminal plasma. J Reprod Immunol. 2002;57(1-2):109-28. http://doi.org/10.1016/S0165-0378(02)00015-3. PMid:12385837.

Robertson SA, Mau VJ, Hudson SN, Tremellen KP. Cytokine-leukocyte networks and the establishment of pregnancy. Am J Reprod Immunol. 1997;37(6):438-42. http://doi.org/10.1111/j.1600-0897.1997.tb00257.x. PMid:9228299.

Robertson SA, Mau VJ, Tremellen KP, Seamark RF. Role of high molecular weight seminal vesicle proteins in eliciting the uterine inflammatory response to semen in mice. J Reprod Fertil. 1996;107(2):265-77. http://doi.org/10.1530/jrf.0.1070265. PMid:8882294.

Robertson SA, Prins JR, Sharkey DJ, Moldenhauer LM. Seminal fluid and the generation of regulatory T cells for embryo implantation. Am J Reprod Immunol. 2013;69(4):315-30. http://doi.org/10.1111/aji.12107. PMid:23480148.

Robertson SA, Seamark RF. Granulocyte-macrophage colony stimulating factor (GM-CSF): one of a family of epithelial cell-derived cytokines in the preimplantation uterus. Reprod Fertil Dev. 1992;4(4):435-48. http://doi.org/10.1071/RD9920435. PMid:1461994.

Robertson SA, Sharkey DJ. The role of semen in induction of maternal immune tolerance to pregnancy. Semin Immunol. 2001;13(4):243-54. http://doi.org/10.1006/smim.2000.0320. PMid:11437632.

Robertson SA. Seminal plasma and male factor signalling in the female reproductive tract. Cell Tissue Res. 2005;322(1):43-52. http://doi.org/10.1007/s00441-005-1127-3. PMid:15909166.

Robillard PY, Hulsey TC, Perianin J, Janky E, Miri EH, Papiernik E. Association of pregnancy-induced hypertension with duration of sexual cohabitation before conception. Lancet. 1994;344(8928):973-5. http://doi.org/10.1016/S0140-6736(94)91638-1. PMid:7934427.

Schluns KS, Cook JE, Le PT. TGF-beta differentially modulates epidermal growth factor-mediated increases in leukemia-inhibitory factor, IL-6, IL-1 alpha, and IL-1 beta in human thymic epithelial cells. J Immunol. 1997;158(6):2704-12. http://doi.org/10.4049/jimmunol.158.6.2704. PMid:9058804.

Sharkey DJ, Tremellen KP, Dekker GA, Robertson SA. Seminal cytokine concentration and human reproductive outcome. Adelaide, Australia: Society for Reproductive Biology; 2002.

Sharkey DJ, Macpherson AM, Tremellen KP, Robertson SA. Seminal plasma differentially regulates inflammatory cytokine gene expression in human cervical and vaginal epithelial cells. Mol Hum Reprod. 2007;13(7):491-501. http://doi.org/10.1093/molehr/gam028. PMid:17483528.

Sharkey DJ, Macpherson AM, Tremellen KP, Mottershead DG, Gilchrist RB, Robertson SA. TGF-beta mediates proinflammatory seminal fluid signaling in human cervical epithelial cells. J Immunol. 2012a;189(2):1024-35. http://doi.org/10.4049/jimmunol.1200005. PMid:22706080.

Sharkey DJ, Tremellen KP, Jasper MJ, Gemzell-Danielsson K, Robertson SA. Seminal fluid induces leukocyte recruitment and cytokine and chemokine mRNA expression in the human cervix after coitus. J Immunol. 2012b;188(5):2445-54. http://doi.org/10.4049/jimmunol.1102736. PMid:22271649.

Siqueira LGB, Dikmen S, Ortega MS, Hansen PJ. Postnatal phenotype of dairy cows is altered by in vitro embryo production using reverse X-sorted semen. J Dairy Sci. 2017;100(7):5899-908. http://doi.org/10.3168/jds.2016-12539. PMid:28456408.

Sjöblom C, Roberts CT, Wikland M, Robertson SA. Granulocyte-macrophage colony-stimulating factor alleviates adverse consequences of embryo culture on fetal growth trajectory and placental morphogenesis. Endocrinology. 2005;146(5):2142-53. http://doi.org/10.1210/en.2004-1260. PMid:15705781.

Sutcliffe AG, Bonduelle M, Taylor BW. Major birth defects after assisted reproduction. N Engl J Med. 2002;347(18):1449-51, author reply 1449-51. http://doi.org/10.1056/NEJM200210313471814. PMid:12418038.

Thompson LA, Barratt CL, Bolton AE, Cooke ID. The leukocytic reaction of the human uterine cervix. Am J Reprod Immunol. 1992;28(2):85-9. http://doi.org/10.1111/j.1600-0897.1992.tb00765.x. PMid:1285856.

Tremellen KP, Seamark RF, Robertson SA. Seminal transforming growth factor beta1 stimulates granulocyte-macrophage colony-stimulating factor production and inflammatory cell recruitment in the murine uterus. Biol Reprod. 1998;58(5):1217-25. http://doi.org/10.1095/biolreprod58.5.1217. PMid:9603256.

Tremellen KP, Valbuena D, Landeras J, Ballesteros A, Martinez J, Mendoza S, Norman RJ, Robertson SA, Simon C. The effect of intercourse on pregnancy rates during assisted human reproduction. Hum Reprod. 2000;15(12):2653-8. http://doi.org/10.1093/humrep/15.12.2653. PMid:11098040.

Viana JH. 2022 Statistics of embryo production and transfer in domestic farm animals. Embryo Technology Newsletter. 2023;41:4.

von Wolff M, Nowak O, Pinheiro RM, Strowitzki T. Seminal plasma--immunomodulatory potential in men with normal and abnormal sperm count. Eur J Obstet Gynecol Reprod Biol. 2007;134(1):73-8. http://doi.org/10.1016/j.ejogrb.2007.01.009. PMid:17341438.

Watson JG, Chaykin S, Carroll J. Repoduction in mice: the fate of sprematozoa not involved in fertilisation. Gamete Res. 1983;7(1):75-84. http://doi.org/10.1002/mrd.1120070107.

Weiker JL. 2022 Semen sales report reflects changing global trends. Madison: National Association of Animal Breeders; 2023. National Association of Animal Breeders newsletter.

Wooldridge LK, Ealy AD. Interleukin-6 increases inner cell mass numbers in bovine embryos. BMC Dev Biol. 2019;19(1):2. http://doi.org/10.1186/s12861-019-0182-z. PMid:30709330.

Yanagimachi R, Chang MC. Infiltration of leucocytes into the uterine lumen of the golden hamster during the oestrous cycle and following mating. J Reprod Fertil. 1963;5(3):389-96. http://doi.org/10.1530/jrf.0.0050389. PMid:14002428.

Yochem DE. Spermatozoon life in the female reproductive tract of the guinea pig and rat. Biol Bull. 1929;56(4):274-97. http://doi.org/10.2307/1536900.
 


Submitted date:
03/30/2024

Accepted date:
05/20/2024

66ace928a953950982718d15 animreprod Articles
Links & Downloads

Anim Reprod

Share this page
Page Sections