Reprod Dom Anim 49 (Suppl. 3), 27–36 (2014); doi: 10.1111/rda.12383 ISSN 0936–6768

Review Article Seminal Fluid and Immune Adaptation for Pregnancy – Comparative Biology in Mammalian Species† JE Schjenken and SA Robertson School of Paediatrics and Reproductive Health, Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia

Contents Seminal fluid delivered to the female reproductive tract at coitus not only promotes the survival and fertilizing capacity of spermatozoa, but also contains potent signalling agents that influence female reproductive physiology to improve the chances of conception and reproductive success. Male to female seminal fluid signalling occurs in rodents, domestic and livestock animals, and all other mammals examined to date. Seminal plasma is instrumental in eliciting the female response, by provision of cytokines and prostaglandins synthesized in the male accessory glands. These agents bind to receptors on target cells in the cervix and uterus, activating changes in gene expression leading to functional adaptations in the female tissues. Sperm also interact with female tract cells, although the molecular basis of this interaction is not yet defined. The consequences are increased sperm survival and fertilization rates, conditioning of the female immune response to tolerate semen and the conceptus, and molecular and cellular changes in the endometrium that facilitate embryo development and implantation. Studies in porcine, equine, bovine, ovine and canine species all show evidence of male–female signalling function for seminal fluid. There are variations between species that relate to their different reproductive strategies and behaviours, particularly the site of seminal fluid deposition and female reproductive tract anatomy. Although the details of the molecular mechanisms require more study, the available data are consistent with both the sperm and plasma fractions of seminal fluid acting in a synergistic fashion to activate inflammation-like responses and downstream female tract changes in each of these species. Insight into the biological function and molecular basis of seminal fluid signalling in the female will inform new interventions and management practices to support optimal reproductive outcomes in domestic, livestock and endangered animal species.

Introduction In addition to sperm, seminal fluid contains a complex mix of bioactive proteins and other agents produced by male accessory sex organs. These soluble factors have important effects after intromission to promote the survival of sperm and their functional capability, by protecting sperm from oxidative stress and providing metabolic support (Poiani 2006). It is less well appreciated that seminal fluid also affects reproductive events independently of sperm, by stimulating the female tissues to increase the chances of conception and progression to pregnancy. There are now a substantial †

The authors acknowledge the support of the NHMRC of Australia. © 2014 Blackwell Verlag GmbH

number of reports from a wide range of mammalian species which show that seminal fluid exerts a considerable influence on female reproductive tract physiology (Robertson 2005; Sharkey et al. 2012b) (Fig. 1). Advances in this field build on decades of research in invertebrates, where effects on female reproductive organs, immune system and behavioural responses are well known to promote fertilization and transmission of the male germ line (Avila et al. 2011). Extensive studies in Drosophila show that seminal fluid proteins, including accessory gland proteins can increase egg laying, ovulation, the production of antimicrobial proteins, remodelling of the female reproductive tract and reduce female receptivity to remating (Chapman and Davies 2004). In this short review, we summarize our current understanding of the physiological significance of seminal fluid signalling in a range of mammalian species, which despite their diverse reproductive tract anatomy and physiology, all share the attribute of female tract responsiveness to seminal fluid after coitus, with biological consequences that impact reproductive success. A better knowledge of this relatively unexplored aspect of reproductive biology will bring new insight into the regulation of reproduction in a range of socially, environmentally and economically important animal species.

Seminal Fluid and the Immune Adaptation to Tolerate Pregnancy The most obvious effect of seminal fluid in the female is the impact on the immune response, particularly the immune changes that are necessary to support the genetically different foetus. For successful pregnancy, a special state of immune ‘tolerance’ must exist from the very earliest time the embryo first contacts the maternal tissues, which for most species, occurs within days of conception. The conceptus is foreign or ‘non-self’ – it expresses antigens, including some encoded by major histocompatibility (MHC) genes, derived from paternal chromosomes. In healthy pregnancy, the conceptus does not experience immunological attack at implantation. A combination of strategies prevents foetal rejection – some aspects of the immune response are circumvented, while others are engaged and activated to skew the immune system towards tolerance. Seminal fluid seems to have a specific role in providing biological signals, specifically paternal antigens and cytokines, which drive the events leading to immune tolerance.

28

JE Schjenken and SA Robertson immune tolerance Treg cells macrophages dendritic cells granulocytes

INFLAMMATORY CYTOKINES

epithelial cells

SEMINAL FLUID embryo development

endometrium

female tract to promote establishment and progression of pregnancy.

phagocytic clearance

uterine lumen

tissue remodeling

Fig. 1. Current understanding of the actions of seminal fluid in the female reproductive tract of species where seminal fluid is deposited in the uterus. Studies in mice and pigs show that after coitus, active moieties in seminal plasma and associated with sperm interact with epithelial cells lining the endometrium, to induce synthesis of proinflammatory cytokines and chemokines. In turn, these cause the recruitment and activation of inflammatory cells in the uterine endometrium, including macrophages, dendritic cells and granulocytes. The macrophages and dendritic cells have roles in inducing endometrial receptivity and in activating regulatory T cells (Treg cells) to mediate maternal immune tolerance of the embryo at implantation. Neutrophils traversing the endometrial epithelium into the luminal cavity act to clear seminal debris and maintain uterine sterility. Epithelial cytokines activated by seminal plasma are also secreted into the luminal fluid where they exert tropic actions on the developing preimplantation embryo [Updated from (Robertson 2005)]

The immediate response to insemination in mammals is a rapid and dramatic influx of inflammatory cells into the site of semen deposition (Robertson 2005). This leucocytic response is evident within an hour of intromission, with firstly neutrophils being released into the luminal space and then other leucocytes – macrophages, dendritic cells and T cells – invading the underlying reproductive tissues. The leucocytes in part respond directly to chemotactic agents in seminal plasma, but the response is amplified as a consequence of seminal fluid-induced production of cytokines and chemokines by epithelial cells lining the reproductive tract surface. The epithelial cell-derived cytokines and chemokines in turn cause leucocytes to extravasate from the blood stream and enter the reproductive tissues. This inflammation-like reaction to semen was first observed in rabbits (McDonald et al. 1952; Taylor 1982), but has since been described in several other species (Robertson 2005). Studies in rodents provide a comprehensive understanding of the molecular and cellular regulation of the inflammatory cascade that has informed experiments in other animals. The inflammatory response to seminal fluid has a central role in the clearance of excess sperm and seminal debris, and promotes recovery of the sterile status of the tract after introduction of micro-organisms at mating. Once this homoeostatic function is accomplished, there is a second and no less important phase in the immune adaptation, which proactively influences the

Seminal Fluid and Regulatory T cells To allow implantation, the female immune response to the conceptus is not switched off or suppressed, but instead immune cells are intimately involved in regulating invasion of conceptus cells at implantation (Trowsdale and Betz 2006). A key element of this active immune tolerance is engagement of special T lymphocytes known as regulatory T cells (Treg cells). Studies in mouse models show that Treg cells are induced to proliferate and then are recruited into the uterus before embryo implantation. Once in place, they permit the immune system to allow the intimate associations between maternal cells and the conceptus-derived placental trophoblasts that are required for adequate placental development and foetal growth. Treg cells operate as potent suppressors of inflammation and cell-mediated immunity (Rudensky 2011). They are critical for the prevention of immune rejection (Sakaguchi 2000; Shevach 2002) via a variety of mechanisms, generally involving suppression of cytokine production and effector function in T cells, B cells, NK cells, dendritic cell and macrophages. Genetic mismatch, particularly in the MHC genes, serves to increase the numbers of Treg cells in the implantation site, and this stimulates the likelihood of healthy pregnancy and benefits foetal growth. In mice, MHC disparate matings result in bigger litters, with heavier foetuses. This is likely to reflect the facilitatory effects of uterine NK cells and Treg cells on transformation of the uterine vasculature, which results in larger diameter blood vessels and increased vascular supply to the placenta (Madeja et al. 2011). A crucial role for seminal fluid in preparation for embryo implantation is through the provision of antigens and cytokines that stimulate expansion of the endometrial Treg-cell population. To ensure enough Treg cells are present in the implantation site, their activation and proliferation during the time between conception and implantation is critical. In mice, the female T-cell response to paternal MHC antigens is initiated immediately after immune cells first contact those antigens transmitted within seminal fluid at coitus, such that the T-cell pool generated during the preimplantation period is expanded by the time of implantation. Soluble MHC is present in seminal plasma, and cell-associated MHC is associated with sperm, seminal leucocytes and/or desquamated genital tract epithelial cells. These and other minor histocompatibility antigens are the same as those later expressed by the conceptus and gestational tissues if pregnancy ensues.

Seminal Fluid and Embryotrophic Cytokines A second key role for seminal fluid signalling is to influence the cytokine and growth factor environment which controls the development of the pre-implantation embryo. As it traverses the female reproductive tract, the embryo is exposed to cytokines and growth factors secreted into the luminal compartment from oviduct © 2014 Blackwell Verlag GmbH

Seminal Plasma Signalling in the Female Tract

and uterine epithelial cells in precise spatial and temporal patterns. Embryos express cytokine receptors from conception until implantation, and several cytokines exert different effects on blastocyst cell number and viability, gene expression and developmental competence (Robertson et al. 1994; Sharkey et al. 1995). The identity and biological effects of several growth factors and cytokines targeting the pre-implantation embryo have been reviewed previously (Kane et al. 1997; Hardy and Spanos 2002). Factors including granulocyte macrophage colony-stimulating factor (GM-CSF), macrophage colony-stimulating factor (M-CSF), and leukemia inhibitory factor (LIF) promote blastocyst development, while others including tumor necrosis factor (TNF), TNF-related apoptosis-inducing ligand (TRAIL), and interferon-c (IFNc) exert potent inhibitory effects. The secretion profile of these cytokines is regulated at the mRNA level principally by ovarian steroid hormones and, in species where seminal fluid reaches the higher tract, by the signalling factors present in seminal fluid. The importance of seminal fluid signalling as a regulator of female tract cytokines is indicated by experiments in mice and other rodents. When the effect of seminal plasma is evaluated by comparing expression of oviduct cytokines in females mated to intact males and females mated with males from which the seminal vesicles are surgically removed to ablate the seminal stimulus, there are substantial effects on oviduct cytokine expression. In the absence of seminal plasma stimulation, oviduct expression of LIF, GM-CSF and interleukin-6 (IL6) was significantly decreased, while the inhibitory cytokine TRAIL was elevated (Bromfield et al. 2014). This was associated with a substantial reduction in the proportion of females achieving pregnancy.

Male to Female Signalling Factors in Seminal Fluid Several studies have aimed to identify the active factors present in seminal fluid. Early experiments in mice using surgical ablation to remove the male accessory glands demonstrated that many active factors in seminal fluid are derived from the seminal plasma fraction (Robertson et al. 1996). These seminal plasma factors include cytokines, sex hormones and prostaglandins (Aumuller and Riva 1992; Maegawa et al. 2002). Using protein chromatography, the cytokine transforming growth factor-b (TGFb) was identified as the principal active factor in mouse seminal plasma that induces the uterine leucocytic response (Tremellen et al. 1998). TGFb has also been identified as an important seminal fluid signalling factor in women (Sharkey et al. 2012a), pigs (O’Leary et al. 2011) and sheep (Scott et al. 2006b). In the mouse, the TGFb1 concentration is 70 ng/ml (Robertson et al. 2002), in humans the average TGFb concentration ranges from 219 ng/ml for TGFb1, 5 ng/ml for TGFb2 and 172 ng/ml for TGFb3 (Sharkey et al. 2012a), while in pigs, the TGFb1 and TGFb2 concentrations are approximately 150 ng/ml each (O’Leary et al. 2011). Studies to evaluate TGFb in seminal plasma of other species are yet to be undertaken, but are clearly important © 2014 Blackwell Verlag GmbH

29

in understanding the importance of this active factor in seminal fluid signalling in other mammals. In addition to TGFb, prostaglandins have also been identified as putative signalling agents present in seminal plasma. In particular, prostaglandin E (PGE) is abundant in human seminal plasma, notably in the 19-hydroxy (19-OH) form (Templeton et al. 1978). In vitro experiments using human cervical explants have shown that 19-OH PGE induces the expression of IL8 and suppresses the anti-inflammatory molecule, secretory leucocyte protease inhibitor (Denison et al. 1999). While PGE is undetectable in rodent and porcine seminal fluid, prostaglandins are present in horse seminal plasma supporting the concept that this factor may play a role in seminal fluid signalling in this species (Claus et al. 1992). There are other active factors identified as potentially playing a role in seminal fluid signalling. Experiments in human tissues show effects of IL8 (Gutsche et al. 2003) and soluble human leukocyte antigen-g (HLA-G) (Larsen et al. 2011), while cysteine-rich secretory protein-3 (CRISP3) (Doty et al. 2011) and porcine sperm adhesion (PSP)-I and PSP-II (Rodriguez-Martinez et al. 2010) have been implicated as signalling agents in equine and porcine species, respectively. In addition to these factors, bacterial products including lipopolysaccharide (Schaefer et al. 2004) and potentially other ligands for Toll-like receptors (TLRs) also contribute to the capacity of seminal fluid to interact with the female reproductive tract. Not all agents act to promote gene expression changes in female tract cells; the type 1 cytokine IFNc is a potent inhibitor of seminal fluid signalling mediated by TGFb (Glynn et al. 2004). Not all seminal fluid signalling can be accounted for by soluble factors in seminal plasma. Studies in several species implicate sperm as also contributing, although the molecular basis of any direct sperm-mediated signalling remains unclear. This array of seminal fluid signalling components presumably acts in concert to regulate the female immune response to coitus. It seems likely that different species will utilize a range of factors in different proportions, depending on the species and the anatomical site of semen deposition. Substantial research will be required to identify the full range of active factors present in seminal fluid of different species, to define how the balance of different agents is regulated in the male, and to understand how alterations to the balance of signalling agents affect the female tract response after coitus.

Evidence for Seminal Fluid Signalling in Various Mammalian Species Seminal fluid signalling in mice The most extensive studies to define the role of seminal fluid in the female immune response to pregnancy are in mice. In mice, the immune response to seminal fluid is initiated when seminal proteins interact with oestrogenprimed cervical and uterine epithelial cells to activate synthesis of pro-inflammatory cytokines including GMCSF, IL6, macrophage chemotactic protein-1 (MCP-1), IL8 (KC) and an array of other chemokines (Sanford

30

et al. 1992; Robertson et al. 1996, 1998). The presence of these inflammatory cytokines elicits recruitment from the blood of inflammatory leucocytes including macrophages, dendritic cells and granulocytes, resulting in the accumulation of these cells in the subepithelial stromal tissue. During this initial acute phase, large populations of neutrophils are also observed to migrate through the epithelial surface and efflux into the luminal cavity (De et al. 1991; McMaster et al. 1992; Robertson et al. 1996). This is a transient response and is resolved prior to embryo implantation, in parallel with the decline in inflammatory cytokine release as rising progesterone levels inhibit synthesis. Detailed studies in different surgical and genetic models in mice have revealed that the female immune response to seminal fluid contributes to the establishment of pregnancy via various pathways. In addition to clearance from the uterine cavity of micro-organisms introduced at mating (Robertson 2005) and induction of Treg cells to confer tolerance at implantation (Guerin et al. 2011), seminal fluid induces the oviduct and uterus to provide trophic factors that promote the development of the pre-implantation embryo (Robertson et al. 2011), and induces expression of embryo attachment ligands and angiogenic factors that promote embryo implantation (Jasper et al. 2011). Another, less well-explored pathway through which seminal fluid may support progression to pregnancy is through effects on the ovary. Experiments in mice show that macrophage populations in corpora lutea are augmented by exposure of the female tract to seminal plasma constituents (Gangnuss et al. 2004). These cells are now understood to have central functions in the development of the luteal vasculature and the progesterone secretion required for endometrial receptivity (Care et al. 2013). The pathway for establishment of tolerance towards paternal antigens is well defined in mice. Using T-cell transgenic mice that have large populations of clonal T cells reactive with male antigens, it can be clearly demonstrated that priming of the female immune response commences when the female is exposed to male transplantation antigens in the seminal fluid (Moldenhauer et al. 2009). This is a crucial first step in eliciting the expansion of antigen-specific populations of Treg cells that prevent immune destruction of the embryo (Robertson et al. 2009). In addition to providing antigen and immune regulatory cytokines to stimulate Treg-cell proliferation, seminal fluid promotes recruitment of Treg cells into the implantation site through stimulating expression of the Treg-cell chemokine, CCL19 (Guerin et al. 2011). Within 2 days of conception, an expanded population of Treg cells can be detected within the uterine lymph nodes (Aluvihare et al. 2004; Zhao et al. 2007; Robertson et al. 2009). By day 3.5 post-coitum, when embryos implant in mice, Treg cells also accumulate in the uterus and are increased >two-fold compared with oestrous numbers (Robertson et al. 2009; Guerin et al. 2011). In mice, this increase in Treg cells clearly depends on seminal plasma, as when seminal vesicles are excised from males before mating, there is no expansion in Tregcell numbers (Robertson et al. 2009; Guerin et al. 2011). TGFb is the prime candidate for this seminal plasma

JE Schjenken and SA Robertson

factor, as TGFb is clearly implicated in Treg-cell generation from precursor T cells (Chen et al. 2003), and furthermore, exogenous TGFb increased vaginal Treg-cell numbers and prevented spontaneous foetal loss in abortion-prone mice when delivered at coitus (Clark et al. 2008). Interestingly, sperm may also provide Treg-inducing and Treg-stabilizing factors, as females mated with vasectomized males show reduced expression of the signature Treg-cell transcription factor forkhead Box P-3 (Foxp3) (Guerin et al. 2011). The importance of the seminal plasma fraction in pregnancy success by pathways independent of Treg cells can be examined by studying syngeneic pregnancies, where there is no MHC disparity between sire and dam. Even in this case, where Treg cells are not required for pregnancy success, the success and quality of the outcome are compromised if females are not exposed to seminal plasma (Bromfield et al. 2014). Experiments in which the seminal vesicle, prostate or coagulating glands are surgically removed from mice, rats and hamsters prior to mating each show that seminal vesicle fluid is the most vital non-sperm component of the ejaculate, and support the interpretation that seminal plasma is essential for optimal sperm survival and fertilization potential (Pang et al. 1979; Queen et al. 1981; Peitz and Olds Clarke 1986; O et al. 1988). In mice, embryo transfer protocols generally employ recipients exposed to seminal plasma by mating to vasectomized males, but foetal loss and abnormality is considerably greater when embryos are transferred after pseudopregnancy achieved without exposure to male fluids (Watson et al. 1983). Confirmation that the effects of seminal plasma are mediated, at least partly, through promoting embryotrophic cytokines in the female tract is provided by studies showing that when recipient females are mated with seminal vesicle-deficient males, transferred embryos give rise to foetuses with impaired growth trajectories and placental development (Bromfield et al. 2014). In other rodent species, implantation rates and foetal growth are similarly impaired unless females are inseminated prior to embryo transfer (Carp et al. 1984). Seminal fluid signalling in human There are a limited number of studies on the role of seminal fluid in the peri-conceptional period in humans. The cervix is the site for seminal fluid deposition following coitus in women and is a major effector site for immune responses in the female genital tract (Pudney et al. 2005). Early studies documented that artificial insemination (AI) of human sperm into the cervix results in an influx of leucocytes, predominantly neutrophils into the surrounding tissue (Pandya and Cohen 1985; Thompson et al. 1992). The seminal plasma fraction is demonstrated to be important in this process, as in vitro and in vivo experiments show that addition of seminal plasma results in the induction of GM-CSF, IL6, IL8, MCP-1, Macrophage inflammatory protein-3a (Mip-3a) and IL1a (Sharkey et al. 2007, 2012b). In addition to the increase in cytokines and chemokines observed in vivo, leucocyte recruitment of predominantly macrophages and dendritic cells and also memory T cells was demonstrated (Sharkey et al. 2012b). © 2014 Blackwell Verlag GmbH

Seminal Plasma Signalling in the Female Tract

Interestingly, there is some evidence consistent with a role for seminal plasma in induction of Treg cells in women, although the definitive experiments have not yet been conducted. In in vitro experiments, seminal plasma induces the differentiation of tolerogenic dendritic cells, potentially through effects mediated by E-series prostaglandins (Remes Lenicov et al. 2012). TGFb within seminal plasma appears to play a major role in this process; however, TGFb alone does not completely explain the signalling properties of seminal fluid (Sharkey et al. 2012a). Seminal plasma also contains substantial concentrations of other immune-deviating cytokines and molecules involved in Treg-cell production from na€ıve T cells, including HLA-G5, TGFb and PGE2 (Kelly and Critchley 1997; Hutter and Dohr 1998; Robertson et al. 2002). Further studies are required to identify the full range of active factors present in seminal fluid, to increase our understanding of seminal fluid signalling in humans. Clinical studies are consistent with an effect of seminal fluid exposure on the outcomes of pregnancy. Immunological conditions of pregnancy such as pre-eclampsia are more common in the first conception, after a short period of cohabitation, after a long period of cohabitation when barrier contraceptives are used or when pregnancy is conceived with a new male partner in multiparous mothers (Klonoff-Cohen et al. 1989; Robillard et al. 1993, 1994; Trupin et al. 1996; Dekker et al. 1998; Saftlas et al. 2014). Data from IVF and related reproductive technologies support a benefit of seminal fluid signalling at conception, with the absence of seminal plasma in the in vitro setting potentially contributing to higher rates of implantation failure and a decrease in embryo quality. IVF success is limited for some subsets of patients with potential consequences for embryos, seen through subtle changes in birthweight and health outcomes of IVF children (Schieve et al. 2002; Maher et al. 2003; Ceelen et al. 2007, 2008). Taking these findings into account, strategies have been devised to utilize factors induced by seminal plasma to improve IVF outcomes, with addition of GM-CSF to IVF culture media significantly improving the survival of transferred embryos to week 12 and live birth (Ziebe et al. 2013). Seminal fluid signalling in pigs In pigs, seminal fluid induces a well-described inflammatory cascade regulated by similar processes to mouse and humans. Like mice, pigs are intrauterine inseminators resulting in direct contact between seminal fluid and epithelial cells lining the endometrium. The most immediate response to seminal fluid is rapid recruitment of neutrophils into the uterine lumen (Lovell and Getty 1968; Claus 1990; Rozeboom et al. 1998), followed by neutrophil binding to viable, but not non-viable spermatozoa (Taylor et al. 2008). The influx of neutrophils is regulated at least partly by seminal plasma proteins PSP-I and PSP-II, which are major proteins in boar seminal plasma (Garcia et al. 2008). In vivo studies show that a heterodimer of PSP-I and PSP-II acts as a postmating pro-inflammatory mediator targeting neutrophils in pigs (Rodriguez-Martinez et al. 2010). Exposing sperm to this complex preserves sperm viability, motility © 2014 Blackwell Verlag GmbH

31

and mitochondrial activity compared with vehicletreated sperm. This preservation may be due to the PSP-I/PSP-II heterodimer adhering to the acrosomal area of sperm (Caballero et al. 2006). In addition to neutrophils, seminal fluid elicits accumulation of macrophages, dendritic cells, granulocytes and lymphocytes within the endometrial stroma (Bischof et al. 1994, 1995; Engelhardt et al. 1997). Experiments with vasectomized boars show that seminal plasma constituents contribute to this phase of the response (Bischof et al. 1994). Studies in gilts have shown that specific factors present in seminal plasma interact with uterine cells to induce expression of GMCSF, IL6, MCP-1 and IL10 and that neuroendocrine stimulation resulting from the physical act of mating is not necessary for this response. GM-CSF, IL6 and MCP-1 have chemotactic properties and are implicated in regulating macrophage and dendritic cell recruitment into the endometrial stroma. Interestingly, exposure to sperm in addition to seminal plasma alters the seminal plasma response (O’Leary et al. 2004; Taylor et al. 2009). While the luminal neutrophil response is resolved within 24 h (Rozeboom et al. 1998), the inflammatory cells infiltrating the endometrium appear to persist and undergo differentiation in the tissue for several days, expanding the endometrial MHC class II+ dendritic cell pool for the duration of the pre-implantation period (Bischof et al. 1994; O’Leary et al. 2004). This transition in leucocyte phenotype is likely to be driven by the more rapid termination of inflammatory cytokine synthesis that occurs in gilts exposed to seminal plasma (O’Leary et al. 2004). Studies in pigs have been instrumental for exploring the effects of seminal fluid on ovarian function. In oestrous pigs, there is a reduction in the interval between the LH surge and ovulation in response to natural mating (Signoret et al. 1972) or seminal plasma administered transcervically (Waberski et al. 1997). The effects appear to be mediated by local transport of seminal constituents or semen-induced effector molecules originating in proximal parts of the tract, because the effect is seen only in ovaries ipsilateral to uterine horns receiving the seminal stimulus. Seminal plasma also promotes the development of the corpus luteum and steroidogenic capacity (O’Leary et al. 2006), with infiltrating macrophages implicated as mediating cells in this process. Seminal fluid signalling in rabbit The first reported evidence of an inflammatory reaction to semen was observed in rabbits in 1952 (McDonald et al. 1952), when pseudopregnant female rabbits or oestrous rabbits were inseminated with semen and the resulting leucocyte influx was examined. The study demonstrated that a leucocytic influx in response to seminal fluid was evident under both hormonal conditions, but that a stronger response occurred in the uterus of pseudopregnant rabbits. Interestingly, administration of sterile epididymal sperm failed to induce a significant reaction, leading the authors to suggest that bacterial contamination was the reason for the leucocytic influx (McDonald et al. 1952). Subsequent studies showed

32

that both seminal plasma and sperm may contribute to the initiation of the leucocytic response in rabbits (Phillips and Mahler 1977a,b). In the presence of seminal plasma, rabbit sperm show similar viability and potentially increased motility, but fertility was not improved (Castellini et al. 2000). Rabbits have also been utilized to provide evidence for rapid transport of seminal fluid components to distal parts of the female reproductive tract. Using bucks immunized with fd-tet, male-derived immunoglobulin could be detected in the uterus within 20 min of intromission (Jappel 1992). Seminal fluid signalling in horse and donkey In equine species, seminal fluid accesses the uterus after coitus and the endometrial response is an active area of investigation because of its relevance to a condition known as mating-induced endometritis, a major issue in these species, particularly horses (Watson 2000; Katila 2012). Endometritis is generally considered a pathological condition, but recent studies have advanced the notion that an appropriate form of endometrial inflammation is a physiologically normal process which has evolved to benefit the establishment of pregnancy, as observed for human and mice. Early studies into endometritis in the horse have focused on the role of sperm in initiating the inflammation-like response and have identified that sperm alone is chemotactic for equine neutrophils. Neutrophils are rapidly recruited into the uterus following exposure to sperm where they peak between 4 and 8 h and are mostly removed by 48 h (Katila 1995). After recruitment into the uterus, neutrophils bind to and then phagocytize sperm (Troedsson et al. 2001; Alghamdi et al. 2004). The presence of seminal plasma inhibits this process in viable but not non-viable sperm (Troedsson et al. 2005) potentially through formation of neutrophil extracellular traps (NETs), which reduce sperm ability to bind to epithelial cells (Alghamdi and Foster 2005). Furthermore, a role for CRISP3 in seminal plasma has been indicated in mediating the interaction between sperm and neutrophils (Doty et al. 2011). Examination of T-cell populations by immunohistochemistry has shown that an increase in CD4+ but not CD8+ T cells in the uterine body can be observed at 6 and 48 h following insemination (Tunon et al. 2000). In addition, as observed in human and mouse, uterine seminal plasma treatment induces the expression of IL1b, IL6, TNF and cyclooxygenase-2 (COX2) genes in the endometrial tissue, suggesting that the actions of seminal plasma in the horse are similar to those observed in mouse and human. Fertility studies examining the impact of seminal plasma in the mare reveal contrasting results. An effect of seminal plasma can be observed when inflammation is induced in the uterus by the prior administration of dead spermatozoa. In this case, delivery of sperm in the presence of seminal plasma results in a 77% conception rate in mares, compared with only 4.5% conception in mares inseminated with sperm in the absence of seminal plasma (Troedsson et al. 2002). In contrast, when there is no prior induction of uterine inflammation, conception rates do not differ (Rigby et al. 2001; Portus et al. 2005). This has led to suggestions that seminal plasma is

JE Schjenken and SA Robertson

required for protection of sperm against phagocytosis by neutrophils in an inflamed uterus, but is less important for conception in a normal quiescent uterine environment (Katila 2005). Studies evaluating the effectiveness of AI in the donkey have demonstrated that commonly used equine cryoprotectants are not suitable, possibly due to toxic effects on the female reproductive tract (Vidament et al. 2009; Rota et al. 2012). In an effort to improve AI rates in donkeys, a number of semen extenders have been compared with seminal plasma in glycerol, or ethylene glycol cryoprotected sperm (Rota et al. 2012). While the use of semen extenders improved total and progressive motility of sperm, there was an improved pregnancy rate when seminal plasma was present. Seminal plasma also increased the female neutrophil response; however, the neutrophil-mediated clearance of sperm was reduced due to suppressed sperm–neutrophil attachment (Rota et al. 2012; Miro et al. 2013). Together, these observations are consistent with a regulatory role for seminal plasma during the peri-conception period in equine species. Seminal fluid signalling in cows In the bovine, little is known about the interactions between seminal fluid and the female reproductive tract. Bovine semen, unlike equine semen, is naturally deposited in the vagina and sperm migrate through the cervix into the uterus leaving the bulk of seminal plasma behind in the lower tract (Alghamdi et al. 2009). However, ejaculated sperm are coated in major seminal plasma proteins which may allow seminal plasma to indirectly function in the bovine uterus (Gwathmey et al. 2006; Alghamdi et al. 2010). Despite this, the potential effects of seminal plasma in the uterus are pertinent, given the common practice of intrauterine AI to yield a higher reproductive efficiency. In studies utilizing intrauterine AI, there is evidence for neutrophil migration but not sperm binding following delivery of sperm alone (Alghamdi et al. 2009). The addition of bovine seminal plasma to diluted or washed sperm has been demonstrated to increase sperm viability, motility and protection (Baas et al. 1983; Garner et al. 2001; Gwathmey et al. 2006). Further, in the presence of seminal plasma, bovine neutrophils have more cell membrane damage, produce less reactive oxygen species following mitogenic stimulation and have a lower transmigration in response to IL8, compared with control (Aloe et al. 2012). However, an increase in sperm–neutrophil binding and NET formation is also be observed in the presence of seminal plasma (Alghamdi et al. 2009). Overall, these studies suggest that bovine seminal plasma has a regulatory effect on inflammatory processes in the uterus following insemination and that neutrophil–sperm interactions may be a physiologically relevant pathway in bovine fertility. Seminal fluid signalling in sheep As observed in other species, mating in sheep induces a transient inflammatory response that draws macrophages and neutrophils into the cervical and uterine tissues © 2014 Blackwell Verlag GmbH

Seminal Plasma Signalling in the Female Tract

(Mattner 1968; Scott et al. 2006a). This is despite the fact that semen is deposited into the cranial vagina and around the cervical os of ewes, rather than directly into the uterus. As observed in humans and mice, ovine seminal plasma contains the active factor TGFb (Scott et al. 2006b). Studies of the uterine inflammatory response at oestrus have shown that both sperm and seminal plasma contribute to the macrophage and neutrophil infiltration as well as increased endometrial IL8 secretion. Exposure to whole semen or washed sperm but not seminal plasma alone induced secretion of GM-CSF. However, a limitation of this study was the possible contribution of bacterial contamination, as antibiotic addition partially reduced the neutrophil response to seminal fluid (Scott et al. 2009). Seminal fluid signalling in dogs There are a limited number of studies examining the role of seminal fluid in the immune response to coitus in bitches. Following coitus, canine sperm are deposited in the cranial vagina and distributed throughout the female reproductive tract via vaginal and uterine contractions. Due to the potential length between coitus and fertilization in canine (up to 9 days), studies have focused on sperm storage in the reproductive tract and it has been established that epithelial cells of the uterine gland and uterine tube junction tether sperm and act to provide a sperm reservoir (Rijsselaere et al. 2004; England et al. 2012, 2013). This interaction is thought to promote sperm survival and fertility and reduce polyspermia (Suarez 2008). In the canine, studies have shown that seminal plasma has beneficial effects on fertility. The addition of seminal plasma to intravaginally administered sperm was shown to increase the odds of conception when compared to sperm combined with Tyrode’s albumin lactate pyruvate media (a medium used for canine sperm during computer-assisted motility assessment) (Nothling et al. 2005). In the first study to document the canine response to seminal fluid, the role of seminal fluid in the canine uterus was examined following AI and it was demonstrated that seminal plasma induces vasodilation (England et al. 2012). Further, sperm attachment to uterine epithelium was shown to be hindered without the presence of seminal plasma, potentially due to sperm interaction with neutrophils. Seminal fluid signalling in other species In a number of other species, there is evidence for seminal fluid function during the peri-conception period of early pregnancy. In cats, assessment of Foxp3, cytotoxic T-lymphocyte-associated protein-4 (Ctla4) and CD25 mRNA levels supports the conclusion that Treg cells are higher in early pregnancy compared with late pregnancy. This study did not examine the role of seminal fluid, but one interpretation is that seminal fluid induces Treg cells following coitus as it does in other species (Lockett et al. 2010). Studies in camels have shown that factors in seminal plasma are required for the induction of ovulation (Li and Zhao 2004), consistent with reports in pigs that seminal fluid influences © 2014 Blackwell Verlag GmbH

33

ovarian function including ovulation and corpus luteum development (O’Leary et al. 2006). Studies in birds provide evidence for immunological responses in the presence of components of seminal fluid. Sperm within the fowl oviduct are phagocytized by immune cells, likely to be macrophages (Koyanagi and Nishiyama 1981).

Conclusions In all species examined so far, seminal fluid appears to play an integral role in eliciting inflammatory changes in the female reproductive tract following coitus. Utilizing rodents and also pigs as experimental models has allowed considerable insight into the molecular and cellular basis for this response and its role in the physiology of embryo implantation and establishing successful pregnancy. There are remarkable similarities in the female response in mice and pigs, where the uterus is the site of seminal fluid deposition. In both species, factors in seminal plasma account for the majority of the endometrial changes with induction of proinflammatory cytokines and chemokines, which combined with endogenous chemokines in seminal plasma cause recruitment of macrophages and dendritic cells. Sperm appear to be particularly important for inducing the first phase of neutrophil recruitment into the luminal cavity of the tract, and additionally contribute by interacting with seminal plasma factors to further modulate cytokine expression and regulate leucocyte influx. NET formation induced by neutrophil– sperm interaction has the potential to further attenuate the immunological environment and may contribute to influencing the female response to sperm, perhaps ultimately impacting which sperm are available to fertilize oocytes. The cascade of changes in local leucocyte populations and cytokine synthesis persists through the pre-implantation period and has sets the course of the female immune response to implantation. In both species, exposure to seminal fluid alters the dynamics of pre-implantation embryo development with an increase in the number of fertilized oocytes attaining viable blastocyst stage. Both also provide evidence that seminal factors promote corpus luteum development and progesterone synthesis. The commonality between these species is consistent with an evolutionarily conserved pathway that has substantial influence on the reproductive process. Furthermore, there is emerging understanding of the similarities and differences in the seminal fluid response of women, where the cervix is the site of seminal fluid deposition. Here, it seems that the cervical response at least to some extent mirrors that seen in the uterus of intrauterine ejaculators. There is less knowledge on how seminal fluid functions in other mammalian species. In sheep and cows where seminal fluid is deposited in the cervix, there is preliminary evidence that signalling factors associated with sperm can influence immune events in the higher tract. Clearly, there is still much to learn about how and why seminal fluid signalling varies between species, the evolutionary value of this event in reproductive biology, and the potential benefit that seminal fluid may impart

34

JE Schjenken and SA Robertson

during the establishment of pregnancy in many animals. This is particularly relevant in species which are commonly bred using AI, where purified sperm is combined with artificial semen extenders for fertilization and the normal process of seminal fluid signalling is averted. Future studies should focus on defining the effects of seminal fluid exposure on conception and implantation, as well as foetal development and postnatal growth trajectory, viability and health of offspring. A better understanding of seminal fluid signalling in a range of animal species will no doubt lead to a better understanding of this important area of biology

References Alghamdi AS, Foster DN, 2005: Seminal DNase frees spermatozoa entangled in neutrophil extracellular traps. Biol Reprod 73, 1174–1181. Alghamdi AS, Foster DN, Troedsson MH, 2004: Equine seminal plasma reduces sperm binding to polymorphonuclear neutrophils (PMNs) and improves the fertility of fresh semen inseminated into inflamed uteri. Reproduction 127, 593–600. Alghamdi AS, Lovaas BJ, Bird SL, Lamb GC, Rendahl AK, Taube PC, Foster DN, 2009: Species-specific interaction of seminal plasma on sperm-neutrophil binding. Anim Reprod Sci 114, 331–344. Alghamdi AS, Funnell BJ, Bird SL, Lamb GC, Rendahl AK, Taube PC, Foster DN, 2010: Comparative studies on bull and stallion seminal DNase activity and interaction with semen extender and spermatozoa. Anim Reprod Sci 121, 249–258. Aloe S, Weber F, Behr B, Sauter-Louis C, Zerbe H, 2012: Modulatory effects of bovine seminal plasma on uterine inflammatory processes. Reprod Domest Anim 47, 12–19. Aluvihare VR, Kallikourdis M, Betz AG, 2004: Regulatory T cells mediate maternal tolerance to the fetus. Nat Immunol 5, 266–271. Aumuller G, Riva A, 1992: Morphology and functions of the human seminal vesicle. Andrologia 24, 183–196. Avila FW, Sirot LK, LaFlamme BA, Rubinstein CD, Wolfner MF, 2011: Insect seminal fluid proteins: identification and function. Annu Rev Entomol 56, 21–40. Baas JW, Molan PC, Shannon P, 1983: Factors in seminal plasma of bulls that affect the viability and motility of spermatozoa. J Reprod Fertil 68, 275–280. Bischof RJ, Lee CS, Brandon MR, Meeusen E, 1994: Inflammatory response in the pig uterus induced by seminal plasma. J Reprod Immunol 26, 131–146. Bischof RJ, Brandon MR, Lee CS, 1995: Cellular immune responses in the pig uterus during pregnancy. J Reprod Immunol 29, 161–178. Bromfield JJ, Schjenken JE, Chin PY, Care AS, Jasper MJ, Robertson SA, 2014: Maternal tract factors contribute to paternal seminal fluid impact on metabolic phenotype in offspring. Proc Natl Acad Sci USA 111, 2200–2205. Caballero I, Vazquez JM, Garcia EM, Roca J, Martinez EA, Calvete JJ, Sanz L, Ekwall H, Rodriguez-Martinez H, 2006:

and may also reveal strategies to improve breeding management and ensure maximal reproductive efficiency. Conflict of Interest None of the authors have any conflict of interest to declare.

Author Contribution JES and SAR both critically reviewed the literature and drafted the manuscript.

Immunolocalization and possible functional role of PSP-I/PSP-II heterodimer in highly extended boar spermatozoa. J Androl 27, 766–773. Care AS, Diener KR, Jasper MJ, Brown HM, Ingman WV, Robertson SA, 2013: Macrophages regulate corpus luteum development during embryo implantation in mice. J Clin Investig 123, 3472–3487. Carp HJ, Serr DM, Mashiach S, Nebel L, 1984: Influence of insemination on the implantation of transferred rat blastocysts. Gynecol Obstet Invest 18, 194–198. Castellini C, Lattaioli P, Moroni M, Minelli A, 2000: Effect of seminal plasma on the characteristics and fertility of rabbit spermatozoa. Anim Reprod Sci 63, 275–282. Ceelen M, van Weissenbruch MM, Roos JC, Vermeiden JP, van Leeuwen FE, Delemarre-van de Waal HA, 2007: Body composition in children and adolescents born after in vitro fertilization or spontaneous conception. J Clin Endocrinol Metab 92, 3417–3423. Ceelen M, van Weissenbruch MM, Vermeiden JP, van Leeuwen FE, Delemarre-van de Waal HA, 2008: Cardiometabolic differences in children born after in vitro fertilization: follow-up study. J Clin Endocrinol Metab 93, 1682–1688. Chapman T, Davies SJ, 2004: Functions and analysis of the seminal fluid proteins of male Drosophila melanogaster fruit flies. Peptides 25, 1477–1490. Chen W, Jin W, Hardegen N, Lei KJ, Li L, Marinos N, McGrady G, Wahl SM, 2003: Conversion of peripheral CD4+CD25naive T cells to CD4+CD25+ regulatory T cells by TGF-beta induction of transcription factor Foxp3. J Exp Med 198, 1875–1886. Clark DA, Fernandes J, Banwatt D, 2008: Prevention of spontaneous abortion in the CBA x DBA/2 mouse model by intravaginal TGF-beta and local recruitment of CD4+8+ FOXP3+ cells. Am J Reprod Immunol 59, 525–534. Claus R, 1990: Physiological role of seminal components in the reproductive tract of the female pig. J Reprod Fertil Suppl 40, 117–131. Claus R, Dimmick MA, Gimenez T, Hudson LW, 1992: Estrogens and prostaglandin F2alpha in the semen and blood plasma of stallions. Theriogenology 38, 687–693. De M, Choudhuri R, Wood GW, 1991: Determination of the number and distribution of macrophages, lymphocytes, and granulocytes in the mouse uterus from

mating through implantation. J Leukoc Biol 50, 252–262. Dekker GA, Robillard PY, Hulsey TC, 1998: Immune maladaptation in the etiology of preeclampsia: a review of corroborative epidemiologic studies. Obstet Gynecol Surv 53, 377–382. Denison FC, Calder AA, Kelly RW, 1999: The action of prostaglandin E2 on the human cervix: stimulation of interleukin 8 and inhibition of secretory leukocyte protease inhibitor. Am J Obstet Gynecol 180, 614–620. Doty A, Buhi WC, Benson S, Scoggin KE, Pozor M, Macpherson M, Mutz M, Troedsson MH, 2011: Equine CRISP3 modulates interaction between spermatozoa and polymorphonuclear neutrophils. Biol Reprod 85, 157–164. Engelhardt H, Croy BA, King GJ, 1997: Role of uterine immune cells in early pregnancy in pigs. J Reprod Fertil Suppl 52, 115–131. England GC, Russo M, Freeman SL, 2012: The bitch uterine response to semen deposition and its modification by male accessory gland secretions. Vet J 195, 179–184. England GC, Burgess CM, Clutterbuck AL, Freeman SL, 2013: Epithelial surface changes and spermatozoa storage in the reproductive tract of the bitch. Vet J 195, 185–191. Gangnuss S, Sutton-McDowall ML, Robertson SA, Armstrong DT, 2004: Seminal plasma regulates corpora lutea macrophage populations during early pregnancy in mice. Biol Reprod 71, 1135–1141. Garcia EM, Vazquez JM, Parrilla I, Ortega MD, Calvete JJ, Sanz L, Martinez EA, Roca J, Rodriguez-Martinez H, 2008: Localization and expression of spermadhesin PSP-I/PSP-II subunits in the reproductive organs of the boar. Int J Androl 31, 408–417. Garner DL, Thomas CA, Gravance CG, Marshall CE, DeJarnette JM, Allen CH, 2001: Seminal plasma addition attenuates the dilution effect in bovine sperm. Theriogenology 56, 31–40. Glynn DJ, Sharkey DJ, Robertson SA, 2004: Interferon-gamma inhibits female reproductive tract responsiveness to seminal plasma. Biology of reproduction, 37th Annual Meeting of the Society for the Study of Reproduction 242. Guerin LR, Moldenhauer LM, Prins JR, Bromfield JJ, Hayball JD, Robertson SA, 2011: Seminal fluid regulates accumulation of FOXP3+ regulatory T Cells in the

© 2014 Blackwell Verlag GmbH

Seminal Plasma Signalling in the Female Tract preimplantation mouse uterus through expanding the FOXP3+ cell pool and CCL19-mediated recruitment. Biol Reprod 85, 397–408. Gutsche S, von Wolff M, Strowitzki T, Thaler CJ, 2003: Seminal plasma induces mRNA expression of IL-1beta, IL-6 and LIF in endometrial epithelial cells in vitro. Mol Hum Reprod 9, 785–791. Gwathmey TM, Ignotz GG, Mueller JL, Manjunath P, Suarez SS, 2006: Bovine seminal plasma proteins PDC-109, BSP-A3, and BSP-30-kDa share functional roles in storing sperm in the oviduct. Biol Reprod 75, 501–507. Hardy K, Spanos S, 2002: Growth factor expression and function in the human and mouse preimplantation embryo. J Endocrinol 172, 221–236. Hutter H, Dohr G, 1998: HLA expression on immature and mature human germ cells. J Reprod Immunol 38, 101–122. Jappel D, 1992: Rapid transport of seminal immunoglobulin to distal parts of the female reproductive tract in rabbits. J Reprod Fertil 96, 135–139. Jasper MJ, Care AS, Sullivan B, Ingman WV, Aplin JD, Robertson SA, 2011: Macrophage-derived LIF and IL1B regulate alpha(1,2)fucosyltransferase 2 (Fut2) expression in mouse uterine epithelial cells during early pregnancy. Biol Reprod 84, 179–188. Kane MT, Morgan PM, Coonan C, 1997: Peptide growth factors and preimplantation development. Hum Reprod Update 3, 137–157. Katila T, 1995: Onset and duration of uterine inflammatory response of mares after insemination with fresh semen. Biol Reprod Mono 1, 515–517. Katila T, 2005: Effect of the inseminate and the site of insemination on the uterus and pregnancy rates of mares. Anim Reprod Sci 89, 31–38. Katila T, 2012: Post-mating inflammatory responses of the uterus. Reprod Domest Anim 47(Suppl. 5), 31–41. Kelly RW, Critchley HO, 1997: Immunomodulation by human seminal plasma: a benefit for spermatozoon and pathogen? Human Reprod (Oxford, England) 12, 2200–2207. Klonoff-Cohen HS, Savitz DA, Celafo RC, McCann MF, 1989: An epidemiologic study of contraception and preeclampsia. JAMA 262, 3143–3147. Koyanagi F, Nishiyama H, 1981: Fate of spermatozoa that do not participate in fertilization in the domestic fowl. Cell Tissue Res 214, 89–95. Larsen MH, Bzorek M, Pass MB, Larsen LG, Nielsen MW, Svendsen SG, Lindhard A, Hviid TV, 2011: Human leukocyte antigen-G in the male reproductive system and in seminal plasma. Mol Hum Reprod 17, 727–738. Li X, Zhao X, 2004: Separation and purification of ovulation-inducing factors in the seminal plasma of the Bactrian camel (Camelus bactrianus). Vet Res Commun 28, 235–245. Lockett NN, Scott VL, Boudreaux CE, Clay BT, Pruett SB, Ryan PL, Coats KS, 2010: Expression of regulatory T cell (Treg) activation markers in endometrial tissues

© 2014 Blackwell Verlag GmbH

from early and late pregnancy in the feline immunodeficiency virus (FIV)-infected cat. Placenta 31, 796–802. Lovell JW, Getty R, 1968: Fate of semen in the uterus of the sow: histologic study of endometrium during the 27 hours after natural service. Am J Vet Res 29, 609–625. Madeja Z, Yadi H, Apps R, Boulenouar S, Roper SJ, Gardner L, Moffett A, Colucci F, Hemberger M, 2011: Paternal MHC expression on mouse trophoblast affects uterine vascularization and fetal growth. Proc Natl Acad Sci USA 108, 4012–4017. Maegawa M, Kamada M, Irahara M, Yamamoto S, Yoshikawa S, Kasai Y, Ohmoto Y, Gima H, Thaler CJ, Aono T, 2002: A repertoire of cytokines in human seminal plasma. J Reprod Immunol 54, 33–42. Maher ER, Afnan M, Barratt CL, 2003: Epigenetic risks related to assisted reproductive technologies: epigenetics, imprinting, ART and icebergs? Human Reprod (Oxford, England) 18, 2508–2511. Mattner PE, 1968: The distribution of spermatozoa and leucocytes in the female genital tract in goats and cattle. J Reprod Fertil 17, 253–261. McDonald LE, Black WG, McNutt SH, Casida LE, 1952: The response of the rabbit uterus to instillation of semen at different phases of the estrous cycle. Am J Vet Res 13, 419–424. McMaster MT, Newton RC, Dey SK, Andrews GK, 1992: Activation and distribution of inflammatory cells in the mouse uterus during the preimplantation period. J Immunol 148, 1699–1705. Miro J, Viles K, Garcia W, Jordana J, Yeste M, 2013: Effect of donkey seminal plasma on sperm movement and sperm-polymorphonuclear neutrophils attachment in vitro. Anim Reprod Sci 140, 164–172. Moldenhauer LM, Diener KR, Thring DM, Brown MP, Hayball JD, Robertson SA, 2009: Cross-presentation of male seminal fluid antigens elicits T cell activation to initiate the female immune response to pregnancy. J Immunol 182, 8080–8093. Nothling JO, Shuttleworth R, de Haas K, Thompson PN, 2005: Homologous prostatic fluid added to frozen-thawed dog spermatozoa prior to intravaginal insemination of bitches resulted in better fertility than albumin-free TALP. Theriogenology 64, 975–991. O WS, Chen HQ, Chow PH, 1988: Effects of male accessory sex gland secretions on early embryonic development in the golden hamster. J Reprod Fertil, 84, 341–344. O’Leary S, Jasper MJ, Warnes GM, Armstrong DT, Robertson SA, 2004: Seminal plasma regulates endometrial cytokine expression, leukocyte recruitment and embryo development in the pig. Reproduction 128, 237–247. O’Leary S, Jasper MJ, Robertson SA, Armstrong DT, 2006: Seminal plasma regulates ovarian progesterone production, leukocyte recruitment and follicular cell responses in the pig. Reproduction 25, 1–12. O’Leary S, Armstrong DT, Robertson SA, 2011: Transforming growth factor-beta

35 (TGFbeta) in porcine seminal plasma. Reprod Fertil Dev 23, 748–758. Pandya IJ, Cohen J, 1985: The leukocytic reaction of the human uterine cervix to spermatozoa. Fertil Steril 43, 417–421. Pang SF, Chow PH, Wong TM, 1979: The role of the seminal vesicle, coagulating glands and prostate glands on the fertility and fecundity of mice. J Reprod Fertil 56, 129–132. Peitz B, Olds Clarke P, 1986: Effects of seminal vesicle removal on fertility and uterine sperm motility in the house mouse. Biol Reprod 35, 608–617. Phillips DM, Mahler S, 1977a: Leukocyte emigration and migration in the vagina following mating in the rabbit. Anat Rec 189, 45–59. Phillips DM, Mahler S, 1977b: Phagocytosis of spermatozoa by the rabbit vagina. Anat Rec 189, 61–71. Poiani A, 2006: Complexity of seminal fluid: a review. Behav Ecol Sociobiol 60, 289– 310. Portus BJ, Reilas T, Katila T, 2005: Effect of seminal plasma on uterine inflammation, contractility and pregnancy rates in mares. Equine Vet J 37, 515–519. Pudney J, Quayle AJ, Anderson DJ, 2005: Immunological microenvironments in the human vagina and cervix: mediators of cellular immunity are concentrated in the cervical transformation zone. Biol Reprod 73, 1253–1263. Queen F, Dhabuwala CB, Pierrepoint CG, 1981: The effect of removal of the various accessory sex glands on the fertility of male rats. J Reprod Fertil 62, 423–436. Remes Lenicov F, Rodriguez RC, Sabatte J, Cabrini M, Jancic C, Ostrowski M, Merlotti A, Gonzalez H, Alonso A, Pasqualini RA, Davio C, Geffner J, Ceballos A, 2012: Semen promotes the differentiation of tolerogenic dendritic cells. J Immunol 189, 4777–4786. Rigby SL, Brinsko SP, Cochran M, Blanchard TL, Love CC, Varner DD, 2001: Advances in cooled semen technologies: seminal plasma and semen extender. Anim Reprod Sci 68, 171–180. Rijsselaere T, Van Soom A, Van Cruchten S, Coryn M, Gortz K, Maes D, de Kruif A, 2004: Sperm distribution in the genital tract of the bitch following artificial insemination in relation to the time of ovulation. Reproduction 128, 801–811. Robertson SA, 2005: Seminal plasma and male factor signalling in the female reproductive tract. Cell Tissue Res 322, 43–52. Robertson SA, Seamark RF, Guilbert LJ, Wegmann TG, 1994: The role of cytokines in gestation. Crit Rev Immunol 14, 239–292. Robertson SA, Mau VJ, Tremellen KP, Seamark RF, 1996: Role of high molecular weight seminal vesicle proteins in eliciting the uterine inflammatory response to semen in mice. J Reprod Fertil 107, 265–277. Robertson SA, Allanson M, Mau VJ, 1998: Molecular regulation of uterine leukocyte recruitment during early pregnancy in the mouse. Trop Res 11, 101–120. Robertson SA, Ingman WV, O’Leary S, Sharkey DJ, Tremellen KP, 2002: Transforming growth factor beta–a mediator of

36 immune deviation in seminal plasma. J Reprod Immunol 57, 109–128. Robertson SA, Guerin LR, Bromfield JJ, Branson KM, Ahlstrom AC, Care AS, 2009: Seminal fluid drives expansion of the CD4+CD25+ T regulatory cell pool and induces tolerance to paternal alloantigens in mice. Biol Reprod 80, 1036–1045. Robertson SA, Chin PY, Glynn DJ, Thompson JG, 2011: Peri-conceptual cytokines– setting the trajectory for embryo implantation, pregnancy and beyond. Am J Reprod Immunol 66(Suppl. 1), 2–10. Robillard PY, Hulsey TC, Alexander GR, Keenan A, de Caunes F, Papiernik E, 1993: Paternity patterns and risk of preeclampsia in the last pregnancy in multiparae. J Reprod Immunol 24, 1–12. Robillard PY, Hulsey TC, Perianin J, Janky E, Miri EH, Papiernik E, 1994: Association of pregnancy-induced hypertension with duration of sexual cohabitation before conception. Lancet 344, 973–975. Rodriguez-Martinez H, Saravia F, Wallgren M, Martinez EA, Sanz L, Roca J, Vazquez JM, Calvete JJ, 2010: Spermadhesin PSP-I/PSP-II heterodimer induces migration of polymorphonuclear neutrophils into the uterine cavity of the sow. J Reprod Immunol 84, 57–65. Rota A, Panzani D, Sabatini C, Camillo F, 2012: Donkey jack (Equus asinus) semen cryopreservation: studies of seminal parameters, post breeding inflammatory response, and fertility in donkey jennies. Theriogenology 78, 1846–1854. Rozeboom KJ, Troedsson MH, Crabo BG, 1998: Characterization of uterine leukocyte infiltration in gilts after artificial insemination. J Reprod Fertil 114, 195– 199. Rudensky AY, 2011: Regulatory T cells and Foxp3. Immunol Rev 241, 260–268. Saftlas AF, Rubenstein L, Prater K, Harland KK, Field E, Triche EW, 2014: Cumulative exposure to paternal seminal fluid prior to conception and subsequent risk of preeclampsia. J Reprod Immunol 101–102, 104–110. Sakaguchi S, 2000: Regulatory T cells: Key controllers of immunologic self-tolerance. Cell 101, 455–458. Sanford TR, De M, Wood GW, 1992: Expression of colony-stimulating factors and inflammatory cytokines in the uterus of CD1 mice during days to 3 of pregnancy. J Reprod Fertil 94, 213–220. Schaefer TM, Desouza K, Fahey JV, Beagley KW, Wira CR, 2004: Toll-like receptor (TLR) expression and TLR-mediated cytokine/chemokine production by human uterine epithelial cells. Immunology 112, 428–436. Schieve LA, Meikle SF, Ferre C, Peterson HB, Jeng G, Wilcox LS, 2002: Low and very low birth weight in infants conceived with use of assisted reproductive technology. N Engl J Med 346, 731–737. Scott JL, Ketheesan N, Summers PM, 2006a: Leucocyte population changes in the reproductive tract of the ewe in response to insemination. Reprod Fertil Dev 18, 627–634. Scott JL, Ketheesan N, Summers PM, 2006b: Presence of TGFbeta but not

JE Schjenken and SA Robertson IL-8 or GM-CSF in Ram Seminal Plasma. Society of Reproductive Biology, Gold Coast, Queensland, p. 49. Scott JL, Ketheesan N, Summers PM, 2009: Spermatozoa and seminal plasma induce a greater inflammatory response in the ovine uterus at oestrus than dioestrus. Reprod Fertil Dev 21, 817–826. Sharkey AM, Dellow K, Blayney M, Macnamee M, Charnock JS, Smith SK, 1995: Stage-specific expression of cytokine and receptor messenger ribonucleic acids in human preimplantation embryos. Biol Reprod 53, 974–981. Sharkey DJ, Macpherson AM, Tremellen KP, Robertson SA, 2007: Seminal plasma differentially regulates inflammatory cytokine gene expression in human cervical and vaginal epithelial cells. Mol Hum Reprod 13, 491–501. Sharkey DJ, Macpherson AM, Tremellen KP, Mottershead DG, Gilchrist RB, Robertson SA, 2012a: TGF-beta mediates proinflammatory seminal fluid signaling in human cervical epithelial cells. J Immunol 189, 1024–1035. Sharkey DJ, Tremellen KP, Jasper MJ, Gemzell-Danielsson K, Robertson SA, 2012b: Seminal fluid induces leukocyte recruitment and cytokine and chemokine mRNA expression in the human cervix after coitus. J Immunol 188, 2445–2454. Shevach EM, 2002: CD4+ CD25+ suppressor T cells: more questions than answers. Nat Rev Immunol 2, 389–400. Signoret JP, Mesnil D, du Buisson F, Mauleon P, 1972: Effect of mating on the onset and duration of ovulation in the sow. J Reprod Fertil 31, 327–330. Suarez SS, 2008: Regulation of sperm storage and movement in the mammalian oviduct. Int J Dev Biol 52, 455–462. Taylor NJ, 1982: Investigation of sperm-induced cervical leucocytosis by a double mating study in rabbits. J Reprod Fertil 66, 157–160. Taylor U, Rath D, Zerbe H, Schuberth HJ, 2008: Interaction of intact porcine spermatozoa with epithelial cells and neutrophilic granulocytes during uterine passage. Reprod Domest Anim 43, 166–175. Taylor U, Zerbe H, Seyfert HM, Rath D, Baulain U, Langner KF, Schuberth HJ, 2009: Porcine spermatozoa inhibit post-breeding cytokine induction in uterine epithelial cells in vivo. Anim Reprod Sci 115, 279–289. Templeton AA, Cooper I, Kelly RW, 1978: Prostaglandin concentrations in the semen of fertile men. J Reprod Fertil 52, 147–150. Thompson LA, Barratt CL, Bolton AE, Cooke ID, 1992: The leukocytic reaction of the human uterine cervix. Am J Reprod Immunol 28, 85–89. Tremellen KP, Seamark RF, Robertson SA, 1998: Seminal transforming growth factor beta1 stimulates granulocyte-macrophage colony-stimulating factor production and inflammatory cell recruitment in the murine uterus. Biol Reprod 58, 1217– 1225. Troedsson MH, Loset K, Alghamdi AM, Dahms B, Crabo BG, 2001: Interaction between equine semen and the endome-

trium: the inflammatory response to semen. Anim Reprod Sci 68, 273–278. Troedsson MH, Alghamdi AS, Mattison J, 2002: Equine seminal plasma protects the fertility of spermatozoa in an inflamed uterine environment. Theriogenology 58, 453–456. Troedsson MH, Desvousges A, Alghamdi AS, Dahms B, Dow CA, Hayna J, Valesco R, Collahan PT, Macpherson ML, Pozor M, Buhi WC, 2005: Components in seminal plasma regulating sperm transport and elimination. Anim Reprod Sci 89, 171–186. Trowsdale J, Betz AG, 2006: Mother’s little helpers: mechanisms of maternal-fetal tolerance. Nat Immunol 7, 241–246. Trupin LS, Simon LP, Eskenazi B, 1996: Change in paternity: a risk factor for preeclampsia in multiparas Epidemiology, 7, 240–244. Tunon AM, Katila T, Magnusson U, Nummijarvi A, Rodriguez-Martinez H, 2000: T-cell distribution in two different segments of the equine endometrium 6 and 48 hours after insemination. Theriogenology 54, 835–841. Vidament M, Vincent P, Martin FX, Magistrini M, Blesbois E, 2009: Differences in ability of jennies and mares to conceive with cooled and frozen semen containing glycerol or not. Anim Reprod Sci 112, 22–35. Waberski D, Claassen R, Hahn T, Jungblut PW, Parvizi N, Kallweit E, Weitze KF, 1997: LH profile and advancement of ovulation after transcervical infusion of seminal plasma at different stages of oestrus in gilts. J Reprod Fertil 109, 29– 34. Watson ED, 2000: Post-breeding endometritis in the mare. Anim Reprod Sci 60–61, 221–232. Watson JG, Carroll J, Chaykin S, 1983: Reproduction in mice: the fate of spermatozoa not involved in fertilization. Gamete Res 7, 75–84. Zhao JX, Zeng YY, Liu Y, 2007: Fetal alloantigen is responsible for the expansion of the CD4(+)CD25(+) regulatory T cell pool during pregnancy. J Reprod Immunol 75, 71–81. Ziebe S, Loft A, Povlsen BB, Erb K, Agerholm I, Aasted M, Gabrielsen A, Hnida C, Zobel DP, Munding B, Bendz SH, Robertson SA, 2013: A randomized clinical trial to evaluate the effect of granulocyte-macrophage colony-stimulating factor (GM-CSF) in embryo culture medium for in vitro fertilization. Fertil Steril 99, 1600–1609.

Submitted: 0 0000; Accepted: 21 Jun 2014 Author’s address (for correspondence): SA Robertson, School of Paediatrics and Reproductive Health, Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia. E-mail: sarah.robertson@ adelaide.edu.au

© 2014 Blackwell Verlag GmbH

Seminal fluid and immune adaptation for pregnancy--comparative biology in mammalian species.

Seminal fluid delivered to the female reproductive tract at coitus not only promotes the survival and fertilizing capacity of spermatozoa, but also co...
471KB Sizes 4 Downloads 3 Views