Acessibilidade / Reportar erro

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

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

Introduction

Semen is a complex biological fluid comprised of spermatozoa, paternal somatic cells, bacteria, and acellular seminal fluid that contains numerous bioactive molecules, substrates, salts, minerals, and various other compounds. The main purpose of semen is to deliver spermatozoa through the male reproductive tract and into the female reproductive tract while supporting the viability of spermatozoa required for fertilization. While the interactions between semen and female reproductive tract tissues have been extensively studied in rodent models, details of a similar response in other animals are lacking (Bromfield, 2018Bromfield 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.
http://doi.org/10.1017/S1751731118000083...
). In this light, it is important to highlight the significant difference in how and where semen is deposited into the female reproductive tract of various species. In rodents the entire uterus is filled with semen at the time of ejaculation, similar to the intracervical insemination that occurs in swine; however, in most other species semen is deposited into the vagina and only a small fraction of spermatozoa traverse the cervix whilst the majority of semen is lost through retrograde transport from the vagina (Marey et al., 2023Marey 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.
http://doi.org/10.1262/jrd.2022-124...
). It is therefore unclear if the type or degree of postcoital inflammation or secondary responses observed in rodents is similar to that of other species and what the consequence of these responses is to pregnancy. This review will focus on what is known regarding the interactions of semen with female tissues and what requires enlightenment if semen has utility in pregnancy outcomes beyond facilitating fertilization in relevant species for agriculture or human health.

What we know about how semen interacts with tissues of the female reproductive tract

In 1929 Donald Yochem performed a series of detailed experiments to evaluate the survival of spermatozoa in the female reproductive tract of the guinea pig after copulation or artificial insemination (Yochem, 1929Yochem 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.
http://doi.org/10.2307/1536900...
). Collectively his work demonstrated a rapid clearance of spermatozoa from the uterus after copulation and he posited that “I have been unable to satisfy myself as to the exact method of their [sperm] elimination, but believe it to be largely due to the phagocytic action of cells within the lumen of the tract.” Since then, a series of experiments have further characterized the inflammatory response of female reproductive tissues to semen, including an elegant series of experiment performed by the reproductive biology pioneers, Ryuzo Yanagimachi and M. C. Chang who describe that postcoital inflammation in hamsters is halved if epididymal sperm were injected into the uterus compared to mating with intact or vasectomized males (Yanagimachi and Chang, 1963Yanagimachi 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.
http://doi.org/10.1530/jrf.0.0050389...
). These men also tested the capacity of mechanical stimulation of the female reproductive tract to induce inflammation and concluded that postcoital inflammation was only induced if females were exposed to an intact male or seminal fluid (Table 1). At this time, it was queried by the authors as to whether there were other functions of postcoital inflammation other than clearance of spermatozoa. Secondary to these observation in various rodent species, the acute postcoital inflammatory response to semen in the first 24 hours after insemination has been characterized in human, cattle, swine, horse and goats (Mattner, 1968Mattner 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.
http://doi.org/10.1530/jrf.0.0170253...
; Fumuso et al., 2003Fumuso 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.
http://doi.org/10.1016/S0165-2427(03)001...
; O'Leary et al., 2004O’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.
http://doi.org/10.1530/rep.1.00160...
; Sharkey et al., 2007Sharkey 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.
http://doi.org/10.1093/molehr/gam028...
; Marey et al., 2023Marey 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.
http://doi.org/10.1262/jrd.2022-124...
).

Table 1
Number of uterine leucocytes recovered from the hamster after mating.

It wasn’t until the 1990’s that the interest in postcoital inflammation was reenergized, and a physiological role for insemination beyond delivery of spermatozoa was investigated. Comprehensive studies in the rodent demonstrate numerous effects in maternal tissues after semen exposure. Initially the same acute, transient postcoital inflammation of the endometrium was observed, characterized by an influx of neutrophils, macrophages and dendritic cells within 12 to 24 hours of mating (Hunt and Robertson, 1996Hunt 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.
http://doi.org/10.1016/S0165-0378(96)883...
; Robertson et al., 1996Robertson 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.
http://doi.org/10.1530/jrf.0.1070265...
). Simultaneously it was demonstrated that semen induced cellular inflammation was due to increased expression of proinflammatory cytokines by the estrogen primed endometrial epithelium, including expression of Csf2, Rantes, Mip1a and Mcp1 (Robertson and Seamark, 1992Robertson 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.
http://doi.org/10.1071/RD9920435...
; Robertson et al., 1997Robertson 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.
http://doi.org/10.1111/j.1600-0897.1997....
). Furthermore, the active component of semen to elicit this response was identified as seminal fluid and not spermatozoa (Robertson et al., 1996Robertson 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.
http://doi.org/10.1530/jrf.0.1070265...
), while additional studies showed seminal fluid transforming growth factor beta (TGFβ) as the bioactive molecule in semen of mice responsible for postcoital inflammation (Tremellen et al., 1998Tremellen 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.
http://doi.org/10.1095/biolreprod58.5.12...
). Similar effects were characterized in the human cervix, where by semen exposure could induce cellular inflammation 24 hours after coitus, while seminal fluid or TGFβ would modulate the gene expression of proinflammatory cytokines in cultured cervical cells (Pandya and Cohen, 1985Pandya 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.
http://doi.org/10.1016/S0015-0282(16)484...
; Thompson et al., 1992Thompson 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.
http://doi.org/10.1111/j.1600-0897.1992....
; Sharkey et al., 2007Sharkey 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.
http://doi.org/10.1093/molehr/gam028...
; Sharkey et al., 2012aSharkey 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.
http://doi.org/10.4049/jimmunol.1200005...
; Sharkey et al., 2012bSharkey 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.
http://doi.org/10.4049/jimmunol.1102736...
). Exposure of cultured bovine endometrial epithelial cells, stromal cells or explants to seminal fluid or TGFβ increases the expression of IL1B, TNF and IL6 expression, while intrauterine infusion of seminal fluid had very little effect on endometrial gene expression at 12 or 24 h post infusion (Ibrahim et al., 2019Ibrahim 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.
http://doi.org/10.1093/biolre/ioy226...
; Rizo et al., 2019Rizo 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.
http://doi.org/10.1530/REP-19-0421...
). Interestingly, bovine seminal fluid has been shown to possess significant RNAse activity which could be determinantal to cell viability (Fernandez-Fuertes et al., 2019Fernandez-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.
http://doi.org/10.1038/s41598-019-51413-...
). Regardless of the mixed results from species, the physiological function of postcoital inflammation is still not clear.

Following allogeneic mating in the rodent the lymph nodes that drain the uterine tissues and the spleen transiently enlarge due to an influx of lymphocytes and antigen presenting cells that had phagocytosed spermatozoa (Beer and Billingham, 1974Beer AE, Billingham RE. Host responses to intra-uterine tissue, cellular and fetal allografts. J Reprod Fertil Suppl. 1974;21:59-88.; Watson et al., 1983Watson 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.
http://doi.org/10.1002/mrd.1120070107...
; Parr and Parr, 1990Parr 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.
http://doi.org/10.1016/0165-0378(90)9002...
). When the phenotype of lymphocytes in uterine draining lymph nodes were characterized, it was revealed that exposure to semen 4 days prior, or specifically seminal fluid increased the development of regulatory T cells with tolerogenic function directed to paternal antigens found in semen (Robertson et al., 2009Robertson 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.
http://doi.org/10.1095/biolreprod.108.07...
; Guerin et al., 2011Guerin 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.
http://doi.org/10.1095/biolreprod.110.08...
; Robertson et al., 2013Robertson 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.
http://doi.org/10.1111/aji.12107...
). The consequence of maternal tolerance to paternal antigen dates back to the postulates of Sir Peter Medawar who suggested that specific mechanisms of maternal tolerance must exist to allow the survival of the semi-allogeneic fetus to survive gestation (Medawar, 1953Medawar P. Some immunological and endocrinological problems raised by the evolution of viviparity in vertebrates. Symp Soc Exp Biol. 1953;7:320-38.). More recently, it was shown in the mouse that maternal tolerance to paternal antigens via regulatory T cells is critical in the survival of the fetus (Aluvihare et al., 2004Aluvihare 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.
http://doi.org/10.1038/ni1037...
). It is plausible that maternal tolerance is initiated at the time of insemination when the maternal immune system is first exposed to paternal antigen and that postcoital inflammation is responsible for the trafficking of paternal antigen in the form of spermatozoa to the uterine draining lymph nodes and drive the expansion of T regulatory cells in preparation for embryo implantation. However, no or little evidence for these mechanisms exist in species other than the mouse.

Embryo transfer to unmated recipients (with no exposure to semen) routinely establishes healthy pregnancies in many species including rodents, humans, swine, cattle, horses etc indicating that female exposure to semen is not a necessity for pregnancy. Indeed, if seminal fluid is supplemented at the time of artificial insemination using semen that contains little to no seminal fluid, pregnancy rates are not increased in cattle or swine (Murray et al., 1983Murray 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.
http://doi.org/10.2527/jas1983.564895x...
; Odhiambo et al., 2009Odhiambo 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.
http://doi.org/10.1016/j.theriogenology....
; Ortiz et al., 2019Ortiz 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.
http://doi.org/10.3168/jds.2019-16251...
). However, while seminal fluid supplementation at the time of insemination does not increase pregnancy rates in swine, evidence does suggest that additional seminal fluid exposure increases litter size and embryo development (Murray et al., 1983Murray 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.
http://doi.org/10.2527/jas1983.564895x...
; O'Leary et al., 2004O’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.
http://doi.org/10.1530/rep.1.00160...
). Two large studies in cattle using over 3,500 cattle subjected to timed artificial insemination supplemented with seminal fluid or saline saw no effect on conception rate, pregnancy loss, calving rate or gestation length (Odhiambo et al., 2009Odhiambo 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.
http://doi.org/10.1016/j.theriogenology....
; Ortiz et al., 2019Ortiz 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.
http://doi.org/10.3168/jds.2019-16251...
). However, it was observed that primiparous dams bred with X-sorted semen and supplemented with seminal fluid at the time of insemination produced heifer calves that were heavier at birth compared to dams supplemented with saline (Ortiz et al., 2019Ortiz 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.
http://doi.org/10.3168/jds.2019-16251...
). Separately, transfer of blastocysts to heifers seven days after mating to vasectomized bulls increased the length of recovered filamentous embryos and altered trophectoderm gene expression compared to heifers that were not mated (Mateo-Otero et al., 2020Mateo-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.
http://doi.org/10.3389/fcell.2020.00341...
). Collectively, these findings suggest that seminal fluid exposure at the time of insemination may not be critical for pregnancy establishment but could alter pregnancy outcomes by increasing litter size or programing fetal growth. In mice, the surgical removal of the seminal vesicle glands that produce the vast majority of seminal fluid does decreases pregnancy rates by 65% and the number of implantations by 37% when sires are used for natural mating. Altered pregnancy rates in these mice is likely due to alterations in sperm transport and viability, as fertility can be restored if 2-cell or blastocyst stage embryos are transferred to pseudopregnant recipients mated with vasectomized males that have also had the seminal vesicles removed (Bromfield et al., 2014Bromfield 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.
http://doi.org/10.1073/pnas.1305609111...
). Interestingly, offspring produced by seminal vesicle excised sires have increased placental weight, grow heavier in adulthood, have increased adiposity and altered metabolic activity, suggesting that the seminal vesicle gland secretions alter postnatal phenotype of offspring (Bromfield et al., 2014Bromfield 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.
http://doi.org/10.1073/pnas.1305609111...
). The authors also demonstrate that the surgical removal of the seminal vesicle gland reduces early embryo development due to altered expression of embryotropic factors in the oviduct, which they conclude is partially responsible for the altered adult phenotype observed in offspring, concordant with the fetal programing hypothesis (Barker, 1998Barker DJ. Mothers, babies and health in later life. Edinburgh: Churchill Livingstone; 1998.). Interestingly, the same increased adiposity is observed in male offspring produced by the transfer of 2-cell embryos into pseudopregnant recipients mated with vasectomized males that have also had the seminal vesicles removed, but not if blastocyst embryos are transferred, further providing evidence that the oviductal response to seminal fluid is altering pregnancy outcomes in the mouse. Conversely, bulls that have had the seminal vesicle glands surgically excised have reduced ejaculate volume but no reduction in conception rates when used for breeding (Faulkner et al., 1968Faulkner 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.
http://doi.org/10.1530/jrf.0.0160179...
).

While numerous studies have demonstrated specific alterations to maternal tissues in response to semen of various species, only three studies exist that show that maternal exposure to seminal fluid alters pregnancy outcomes – one in the mouse in which adult offspring are altered in the absence of seminal fluid, one in swine in which seminal fluid supplementation increases litter size, and one in cattle in which seminal fluid supplementation increased birth weight of heifer calves (Murray et al., 1983Murray 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.
http://doi.org/10.2527/jas1983.564895x...
; Bromfield et al., 2014Bromfield 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.
http://doi.org/10.1073/pnas.1305609111...
; Ortiz et al., 2019Ortiz 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.
http://doi.org/10.3168/jds.2019-16251...
). However, it must be clarified that the intrauterine infusion of seminal fluid performed in cattle is a pharmacological application as it does not mirror the physiological deposition of semen into the vagina during live cover with a bull. In parallel, pregnancies are routinely initiated in the absence of seminal fluid in multiple species, but it is unclear if the outcomes of these pregnancies are optimal or could be improved if seminal fluid exposure of maternal tissues was to occur at the time of conception.

What we think we know about the consequences of female tissue responses to semen

Achieving pregnancies in the absence of semen and postcoital inflammation is commonplace in many species including rodents, humans and many domestic species. In 2020, 203,164 human IVF cycles were initiated in the United States with the intent of embryo transfer, of which 165,041 transfers occurred, resulting in 91,453 pregnancies (55%) and 75,023 live birth pregnancies (45%) (National Center for Chronic Disease Prevention and Health Promotion, Division of Reproductive Health, 2020National 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
https://www.cdc.gov/art/reports/2020/sum...
). According to data obtained by the International Embryo Technology Society, in 2022 a total of 1,189,699 embryos were transferred globally into cattle, 41,316 into sheep, 20,381 into goats, and 28,996 into horses (Viana, 2023Viana JH. 2022 Statistics of embryo production and transfer in domestic farm animals. Embryo Technology Newsletter. 2023;41:4.). The numbers of embryo transfers for domestic species pales into insignificance compared to the number of bovine semen units sold by the United States for artificial insemination, with over 69 million units sold in 2022 (Weiker, 2023Weiker JL. 2022 Semen sales report reflects changing global trends. Madison: National Association of Animal Breeders; 2023. National Association of Animal Breeders newsletter.). The processing of semen for artificial insemination varies amongst companies, but all entail the significant dilution of semen in extenders that contain various compounds including antibiotics, lipids sourced from animal products like hen eggs or proprietary synthetic compounds, effectively reducing the volume of semen and seminal fluid encountered by the female reproductive tract (Lonergan, 2018Lonergan P. Review: historical and futuristic developments in bovine semen technology. Animal. 2018;12(s1):s4-18. http://doi.org/10.1017/S175173111800071X. PMid:29642968.
http://doi.org/10.1017/S175173111800071X...
). Additionally, sexed semen contains only spermatozoa diluted in artificial semen extender, effectively eliminating exposure of the female to seminal fluid. Collectively it is important to consider the sheer numbers of cattle born by embryo transfer or artificial insemination where postcoital inflammation is either absent or significantly reduced. And while conception rates are marginally increased in cows bred by natural service with a bull compared to artificial insemination, this is due to the increased opportunities for breeding when live cover is used in place of artificial insemination (Lima et al., 2009Lima 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.
http://doi.org/10.3168/jds.2009-2197...
). The simple realization that millions of pregnancies have been achieved in cattle and humans by using embryo transfer whereby female tissues are never exposed to semen, one may ask, what is the potential physiological consequence of maternal responses to semen and the postcoital inflammatory response?

It has been proposed that maternal exposure to semen improves pregnancy outcomes and diminishes pregnancy complications by facilitating the improved development of the preimplantation embryo due to increased expression of embryotropic molecules in the female tract, modulating maternal immune tolerance to the semiallogeneic conceptus and altering tissue remodeling of the endometrium to allow for improved implantation and placentation (Robertson and Sharkey, 2001Robertson 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.
http://doi.org/10.1006/smim.2000.0320...
; Robertson, 2005Robertson 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.
http://doi.org/10.1007/s00441-005-1127-3...
; Bromfield, 2014Bromfield 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.
http://doi.org/10.1007/s10815-014-0243-y...
, 2016Bromfield 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.
http://doi.org/10.1530/REP-16-0313...
, 2018Bromfield 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.
http://doi.org/10.1017/S1751731118000083...
). However, very little direct experimental evidence exists to support these theories. As described above, one study has demonstrated a reduced preimplantation embryo quality when seminal vesicle excised males are used to generate pregnancies in the mouse, and two studies (one in the pig and one in the bovine) have suggested improved embryo quality when females are exposed to seminal fluid – neither of which demonstrated altered pregnancy outcomes (O'Leary et al., 2004O’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.
http://doi.org/10.1530/rep.1.00160...
; Bromfield et al., 2014Bromfield 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.
http://doi.org/10.1073/pnas.1305609111...
; Mateo-Otero et al., 2020Mateo-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.
http://doi.org/10.3389/fcell.2020.00341...
). While semen does induce the expression of embryotropic factors such as CSF-2, IL-6 and LIF in the female reproductive tract of many species, in vitro culture of embryos demonstrates that these factors are not an absolute requirement to embryo development or pregnancy success (Schluns et al., 1997Schluns 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.
http://doi.org/10.4049/jimmunol.158.6.27...
; Bromfield et al., 2014Bromfield 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.
http://doi.org/10.1073/pnas.1305609111...
). However, the supplementation of embryo culture medium with CSF-2, IL-6 or LIF has been shown to improve embryo development and potentially even program offspring development after parturition in mice and cattle (Mitchell et al., 2002Mitchell 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.
http://doi.org/10.1095/biolreprod67.2.46...
; Sjoblom et al., 2005Sjöblom 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.
http://doi.org/10.1210/en.2004-1260...
; Block et al., 2011Block 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.
http://doi.org/10.1016/j.theriogenology....
; Kannampuzha-Francis et al., 2015Kannampuzha-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.
http://doi.org/10.1002/mrd.22533...
; Wooldridge and Ealy, 2019Wooldridge 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.
http://doi.org/10.1186/s12861-019-0182-z...
).

Maternal immune tolerance to the semiallogeneic conceptus during pregnancy is considered an absolute requirement for successful pregnancy outcomes (Medawar, 1953Medawar P. Some immunological and endocrinological problems raised by the evolution of viviparity in vertebrates. Symp Soc Exp Biol. 1953;7:320-38.). The mechanisms by which maternal tolerance to paternal antigen is achieved has yet to be fully elucidated; however, exposure to semen at conception has been proposed as one mechanism in the rodent. The true nature for requirements of maternal immune tolerance are evident in the mouse, as depletion of peripheral CD25 positive T regulatory cells renders females infertile (Aluvihare et al., 2004Aluvihare 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.
http://doi.org/10.1038/ni1037...
). In parallel, the expansion of T regulatory cells in the mouse and pig oviduct, endometrium, and draining lymph nodes of the uterus is in part facilitated by exposure to seminal fluid at the time of mating (Robertson et al., 2009Robertson 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.
http://doi.org/10.1095/biolreprod.108.07...
; Jiwakanon et al., 2010Jiwakanon 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.
http://doi.org/10.1016/j.anireprosci.201...
; Guerin et al., 2011Guerin 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.
http://doi.org/10.1095/biolreprod.110.08...
). However, alternative mechanisms for the expansion of these important cell types must exist to allow for pregnancy establishment after embryo transfer.

Secondary associations of pregnancy success and semen exposure exist as part of epidemiological studies. In women receiving embryo transfer, it is routine to abstain from coitus around the time of transfer to avoid uterine contraction and infection that could terminate a pregnancy (Fanchin et al., 1998aFanchin 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.
http://doi.org/10.1016/S0015-0282(98)002...
; Fanchin et al., 1998bFanchin 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.
http://doi.org/10.1093/humrep/13.7.1968...
). However, a single study where patients were asked to engage in intercourse around the time of embryo transfer demonstrated a small yet significant increase in the proportion of women with a biochemical pregnancy compared to patients that abstained from intercourse (11.0% vs. 7.7%) (Tremellen et al., 2000Tremellen 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.
http://doi.org/10.1093/humrep/15.12.2653...
). Similarly, the maternal response to semen has been suggested to reduce pregnancy complications including preeclampsia, recurrent miscarriage and fetal growth restriction. A positive association between the use of barrier contraception or short periods of cohabitation and preeclampsia has been established, whereby the authors suggest that increased exposure of maternal tissues to semen reduces the incidence of preeclampsia (Klonoff-Cohen et al., 1989Klonoff-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.
http://doi.org/10.1001/jama.1989.0343022...
; Robillard et al., 1994Robillard 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.
http://doi.org/10.1016/S0140-6736(94)916...
). Even more compelling is a study that utilized vaginal suppositories of seminal fluid to demonstrate a decrease in recurrent miscarriage (Coulam and Stern, 1995Coulam CB, Stern JJ. Effect of seminal plasma on implantation rates. Early Pregnancy. 1995;1(1):33-6. PMid:9363233.). While these studies suggest a positive association of semen exposure and pregnancy outcomes, to date no mechanism has been able to directly link pregnancy outcomes and the postcoital response to semen in humans or animals other than the mouse.

In the mouse, TGFβ derived from seminal fluid has been shown to be the most potent bioactive molecule to elicit the postcoital inflammatory response in maternal tissues and drive the expansion of T regulator cells (Tremellen et al., 1998Tremellen 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.
http://doi.org/10.1095/biolreprod58.5.12...
; Robertson et al., 2002Robertson 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.
http://doi.org/10.1016/S0165-0378(02)000...
; Fantini et al., 2006Fantini 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.
http://doi.org/10.1136/gut.2005.072801...
; Konkel et al., 2017Konkel 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.
http://doi.org/10.1016/j.immuni.2017.03....
). Similarly, TGFβ is present at high concentrations in human, boar and bull semen and can partially mimic the actions of semen or seminal fluid in cultured cells (Nocera and Chu, 1995Nocera 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.
http://doi.org/10.1111/j.1600-0897.1995....
; Robertson et al., 2002Robertson 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.
http://doi.org/10.1016/S0165-0378(02)000...
; Sharkey et al., 2002Sharkey DJ, Tremellen KP, Dekker GA, Robertson SA. Seminal cytokine concentration and human reproductive outcome. Adelaide, Australia: Society for Reproductive Biology; 2002., 2012aSharkey 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.
http://doi.org/10.4049/jimmunol.1200005...
; O'Leary et al., 2011O’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.
http://doi.org/10.1071/RD11001...
; Rizo et al., 2019Rizo 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.
http://doi.org/10.1530/REP-19-0421...
). However, the supplementation of TGFβ at the time of artificial insemination in cattle does not improve pregnancy rates and no correlation between semen TGFβ and fertility exists in boars or humans (Loras et al., 1999Loras 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.
http://doi.org/10.1093/humrep/14.6.1534...
; von Wolff et al., 2007von 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.
http://doi.org/10.1016/j.ejogrb.2007.01....
; Odhiambo et al., 2009Odhiambo 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.
http://doi.org/10.1016/j.theriogenology....
; O'Leary et al., 2011O’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.
http://doi.org/10.1071/RD11001...
).

What we need to do if we envisage using semen components to modulate pregnancy outcomes

Assisted reproductive technologies like embryo transfer and artificial insemination reduce or eliminate exposure of the maternal reproductive tract to semen and/or seminal fluid. The utility of advanced technologies including in vitro fertilization, embryo culture and intracytoplasmic sperm injection in the human have been associated with increased pregnancy failures, increased pregnancy complications and increased birth defects in the human (Cox et al., 2002Cox 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.
http://doi.org/10.1086/341096...
; Hansen et al., 2002Hansen 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.
http://doi.org/10.1056/NEJMoa010035...
; Sutcliffe et al., 2002Sutcliffe 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.
http://doi.org/10.1056/NEJM2002103134718...
; Kurinczuk et al., 2004Kurinczuk 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.
http://doi.org/10.1097/00001703-20040600...
; Hansen et al., 2005Hansen 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.
http://doi.org/10.1093/humrep/deh593...
; Ludwig et al., 2005Ludwig 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.
http://doi.org/10.1136/jmg.2004.026930...
). The vast majority of these studies conclude that the extreme manipulation of gametes during these procedures and suboptimal culture of embryos results in imprinting and genetic defects responsible for the increased complications observed in human patients. In cattle however, two unique studies exist to suggest that advanced reproductive technologies also diminish the genetic potential of offspring (Siqueira et al., 2017Siqueira 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.
http://doi.org/10.3168/jds.2016-12539...
; Lafontaine et al., 2023Lafontaine 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.
http://doi.org/10.3168/jds.2022-22736...
). These studies utilized the genomic potential of dairy cows and compared it to the actual phenotypic outputs of milk production in a total of 319,140 animals conceived by artificial insemination using conventional semen or X-sorted semen, multiple ovulation embryo transfer or in vitro fertilization with embryo transfer. Collectively these studies suggest that as the complexity of technology for conception increased the cumulative mortality of offspring increased and their productive phenotype did not reach their genomic potential (Siqueira et al., 2017Siqueira 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.
http://doi.org/10.3168/jds.2016-12539...
; Lafontaine et al., 2023Lafontaine 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.
http://doi.org/10.3168/jds.2022-22736...
). The question from these impressive studies remains, what is the mechanisms by which the phenotype of these offspring is altered following assisted reproductive technologies, and could semen exposure play a role?

While we have discussed three experiments (one each in mouse, cattle and swine) that demonstrate a definitive link between improved pregnancy outcomes and semen exposure at the time of conception, many unanswered questions pertaining to the relevance of physiological semen exposure and pregnancy outcomes remain. Various experimental paradigms have shown the influence of semen, seminal fluid or bioactive semen components on maternal responses that vary from postcoital cellular inflammation, altered endometrial or oviductal gene expression, modulation of lymphocyte populations and even embryo quality in different species. However, key questions remain that would allow us to draw conclusive, mechanistic effects of how semen exposure modulates pregnancy outcomes. The first and most important question pertains to physiological semen exposure; while many studies focus on the rodent where the uterus is exposed to large volumes of semen, fewer studies have focused on the effects of vaginal semen exposure on promoting pregnancy outcomes in species like the human or cattle. The singular study in cattle that demonstrated an effect of seminal fluid exposure on birth weight utilized intrauterine infusion of pooled seminal fluid. It is unclear if any seminal fluid reaches the upper reproductive tract in cattle (or humans), and as such this experiment was indeed a pharmacological intervention. Indeed, when heifers were mated with intact bulls a total of 22 differentially expressed genes were identified in the endometrium 24 h later when compared to heifers that were not mated; whereas no differentially expressed genes were identified in the endometrium when heifers were mated with vasectomized bulls (compared to heifers that were not mated), suggesting that vaginal exposure to semen or seminal fluid by copulation has very little effect on the endometrium (Recuero et al., 2020Recuero 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.
http://doi.org/10.3389/fcell.2020.00547...
). It will be imperative to determine in all species how much, if any, seminal fluid reaches the upper reproductive tract if we hope to understand the physiological significance of semen exposure. Similarly, only a single study in each of the pig, cow and mouse have shown that semen exposure alters development of the preimplantation embryo. It is unclear how the increased birth weight of calves is driven by seminal fluid exposure, is this due to altered embryo development, interaction of spermatozoa with seminal fluid, altered placentation, and are there further consequences to the offspring that are born slightly heavier than their herd mates? Similarly, do semen induced changes to peripheral immune cell populations have any effect on pregnancy success or are these simply alterations driven by semen exposure that do not have a specific physiological function associated with pregnancy outcome, and do these alterations exist in animals other than mice? Beyond the physiological responses to semen, if the intent is to develop pharmacological interventions or semen derived supplements to improve pregnancy outcomes it will be important to identify the bioactive compounds of semen that elicit the maternal responses involved in modulating pregnancy outcomes. Semen is a complex biological fluid, and its composition varies dramatically from one species to another, within an individual over time and is susceptible to exogenous influence by diet and environment. It is likely that all of these types of questions are pertinent to understanding the true physiological role of semen exposure in the female, more over it will be important to understand these in relevant species in which we are trying to improve pregnancy outcomes like the human or domestic species and less so in the mouse.

Conclusions

There is indisputable evidence that semen interacts with maternal tissues at the time of insemination to elicit distinctive cellular and molecular alterations to reproductive and peripheral tissues. The consequence of semen exposure on pregnancy outcomes is less established, especially beyond the mouse, with minimal evidence that seminal fluid supplementation increases litter size in swine and heifer birth weight in cattle. While these studies are of great interest to animal agriculture, they do not directly speak to physiological responses due to the use of artificial insemination with diluted semen and intrauterine application of additional seminal fluid that would normally be found only in the vagina. While I and others will continue to explore how semen interacts with maternal tissues, we have a long journey ahead to determine a physiological role and potentially develop therapeutics to improve pregnancy outcomes in domestic species and humans.

  • Financial support: None .
  • How to cite: Bromfield JJ. Interaction of semen with female reproductive tract tissues: what we know, what we guess and what we need to do. Anim Reprod. 2024;21(3):e20240042. https://doi.org/10.1590/1984-3143-AR2024-0042

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.
    » http://doi.org/10.1038/ni1037
  • 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.
    » http://doi.org/10.1016/j.theriogenology.2011.07.025
  • 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.
    » http://doi.org/10.1073/pnas.1305609111
  • 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.
    » http://doi.org/10.1530/REP-16-0313
  • 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.
    » http://doi.org/10.1017/S1751731118000083
  • 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.
    » http://doi.org/10.1007/s10815-014-0243-y
  • 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.
    » http://doi.org/10.1086/341096
  • 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.
    » http://doi.org/10.1016/S0015-0282(98)00277-5
  • 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.
    » http://doi.org/10.1093/humrep/13.7.1968
  • 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.
    » http://doi.org/10.1136/gut.2005.072801
  • 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.
    » http://doi.org/10.1530/jrf.0.0160179
  • 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.
    » http://doi.org/10.1038/s41598-019-51413-4
  • 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.
    » http://doi.org/10.1016/S0165-2427(03)00137-5
  • 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.
    » http://doi.org/10.1095/biolreprod.110.088591
  • 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.
    » http://doi.org/10.1093/humrep/deh593
  • 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.
    » http://doi.org/10.1056/NEJMoa010035
  • 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.
    » http://doi.org/10.1016/S0165-0378(96)88352-5
  • 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.
    » http://doi.org/10.1093/biolre/ioy226
  • 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.
    » http://doi.org/10.1016/j.anireprosci.2010.01.005
  • 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.
    » http://doi.org/10.1002/mrd.22533
  • 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.
    » http://doi.org/10.1001/jama.1989.03430220066032
  • 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.
    » http://doi.org/10.1016/j.immuni.2017.03.015
  • 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.
    » http://doi.org/10.1097/00001703-200406000-00002
  • 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.
    » http://doi.org/10.3168/jds.2022-22736
  • 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.
    » http://doi.org/10.3168/jds.2009-2197
  • 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.
    » http://doi.org/10.1017/S175173111800071X
  • 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.
    » http://doi.org/10.1093/humrep/14.6.1534
  • 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.
    » http://doi.org/10.1136/jmg.2004.026930
  • 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.
    » http://doi.org/10.1262/jrd.2022-124
  • 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.
    » http://doi.org/10.3389/fcell.2020.00341
  • 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.
    » http://doi.org/10.1530/jrf.0.0170253
  • 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.
    » http://doi.org/10.1095/biolreprod67.2.460
  • 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.
    » http://doi.org/10.2527/jas1983.564895x
  • 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
    » 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.
    » http://doi.org/10.1111/j.1600-0897.1995.tb00897.x
  • 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.
    » http://doi.org/10.1071/RD11001
  • 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.
    » http://doi.org/10.1530/rep.1.00160
  • 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.
    » http://doi.org/10.1016/j.theriogenology.2009.04.013
  • 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.
    » http://doi.org/10.3168/jds.2019-16251
  • 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.
    » http://doi.org/10.1016/S0015-0282(16)48442-6
  • 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.
    » http://doi.org/10.1016/0165-0378(90)90029-6
  • 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.
    » http://doi.org/10.3389/fcell.2020.00547
  • 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.
    » http://doi.org/10.1530/REP-19-0421
  • 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.
    » http://doi.org/10.1095/biolreprod.108.074658
  • 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.
    » http://doi.org/10.1016/S0165-0378(02)00015-3
  • 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.
    » http://doi.org/10.1111/j.1600-0897.1997.tb00257.x
  • 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.
    » http://doi.org/10.1530/jrf.0.1070265
  • 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.
    » http://doi.org/10.1111/aji.12107
  • 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.
    » http://doi.org/10.1071/RD9920435
  • 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.
    » http://doi.org/10.1006/smim.2000.0320
  • 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.
    » http://doi.org/10.1007/s00441-005-1127-3
  • 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.
    » http://doi.org/10.1016/S0140-6736(94)91638-1
  • 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.
    » http://doi.org/10.4049/jimmunol.158.6.2704
  • 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.
    » http://doi.org/10.1093/molehr/gam028
  • 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.
    » http://doi.org/10.4049/jimmunol.1200005
  • 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.
    » http://doi.org/10.4049/jimmunol.1102736
  • 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.
    » http://doi.org/10.3168/jds.2016-12539
  • 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.
    » http://doi.org/10.1210/en.2004-1260
  • 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.
    » http://doi.org/10.1056/NEJM200210313471814
  • 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.
    » http://doi.org/10.1111/j.1600-0897.1992.tb00765.x
  • 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.
    » http://doi.org/10.1095/biolreprod58.5.1217
  • 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.
    » http://doi.org/10.1093/humrep/15.12.2653
  • 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.
    » http://doi.org/10.1016/j.ejogrb.2007.01.009
  • 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
    » 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.
    » http://doi.org/10.1186/s12861-019-0182-z
  • 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.
    » http://doi.org/10.1530/jrf.0.0050389
  • 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
    » http://doi.org/10.2307/1536900

Publication Dates

  • Publication in this collection
    05 Aug 2024
  • Date of issue
    2024

History

  • Received
    30 Mar 2024
  • Accepted
    20 May 2024
Colégio Brasileiro de Reprodução Animal Coronel José dias Bicalho, 1224, CEP: , 31275-050, Belo Horizonte, MG - Brasil, Tel.: 55-31-3491 7122 - Belo Horizonte - MG - Brazil
E-mail: animreprod.journal@gmail.com