Next Article in Journal
Model-Agnostic Method for Thoracic Wall Segmentation in Fetal Ultrasound Videos
Previous Article in Journal
Determination of Major Endogenous FAHFAs in Healthy Human Circulation: The Correlations with Several Circulating Cardiovascular-Related Biomarkers and Anti-Inflammatory Effects on RAW 264.7 Cells
Previous Article in Special Issue
The Biological Function of Extracellular Vesicles during Fertilization, Early Embryo—Maternal Crosstalk and Their Involvement in Reproduction: Review and Overview
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Comparative View on the Oviductal Environment during the Periconception Period

by
Leopoldo González-Brusi
,
Blanca Algarra
,
Carla Moros-Nicolás
,
Mª José Izquierdo-Rico
,
Manuel Avilés
* and
Maria Jiménez-Movilla
*
Department of Cell Biology and Histology, School of Medicine, University of Murcia, Campus Mare Nostrum and IMIB-Arrixaca, 30100 Murcia, Spain
*
Authors to whom correspondence should be addressed.
Equal contribution from the two co-authors.
Biomolecules 2020, 10(12), 1690; https://doi.org/10.3390/biom10121690
Submission received: 4 October 2020 / Revised: 30 November 2020 / Accepted: 9 December 2020 / Published: 17 December 2020
(This article belongs to the Special Issue Biomolecules in Maternal–Embryo Communication in Implantation)

Abstract

:
The oviduct plays important roles in reproductive events: sperm reservoir formation, final gamete maturation, fertilization and early embryo development. It is well known that the oviductal environment affects gametes and embryos and, ultimately, the health of offspring, so that in vivo embryos are better in terms of morphology, cryotolerance, pregnancy rates or epigenetic profile than those obtained in vitro. The deciphering of embryo–maternal interaction in the oviduct may provide a better understanding of the embryo needs during the periconception period to improve reproductive efficiency. Here, we perform a comparative analysis among species of oviductal gene expression related to embryonic development during its journey through the oviduct, as described to date. Cross-talk communication between the oviduct environment and embryo will be studied by analyses of the secreted or exosomal proteins of the oviduct and the presence of receptors in the membrane of the embryo blastomeres. Finally, we review the data that are available to date on the expression and characterization of the most abundant protein in the oviduct, oviductin (OVGP1), highlighting its fundamental role in fertilization and embryonic development.

1. Introduction

The oviduct, a tubular organ that connects the ovaries with the uterus, is composed of four anatomical regions: the infundibulum, the ampulla, the isthmus and the uterine-tubal junction [1]. In female mammals, this passageway is also known as the uterine tube or Fallopian tube. While the oviduct was previously considered a passive channel for the transport of gametes and embryos, it is now widely accepted that it is not just a tract that joins the ovaries to the uterus. Indeed, it plays important roles in reproductive events participating in sperm reservoir formation, final gamete maturation and transport, fertilization and early embryo development; moreover, inside this organ is where the first maternal-embryo cross-talk begins [2,3,4,5,6,7,8,9]. Before implantation, the embryo spends between 1 and 10 days in the oviduct, depending on the species (Table 1). During these days important morphological, molecular and metabolic modifications occur, such as, the first mitotic division and embryonic genome activation [10,11,12,13,14]. In this phase, the conceptus is dependent on oviductal fluid (OF) formed by the nutrients secreted from the oviduct epithelial cells, as well as serum transudate [15,16,17]. The OF is a complex and a dynamic fluid, composed of metabolites, inorganic salts, amino acids, proteins, glycosaminoglycans, lipids and extracellular vesicles, amongst others, which fulfil the preimplantation microenvironment, guaranteeing a conceptus viable for implantation and leading to the creation of healthy offspring [3,7,18,19,20]. Despite the important functions in which it participates, the oviduct has not been the object of a detailed study until very recently, perhaps due to the success of in vitro fertilization (IVF) and other assisted reproductive technologies (ARTs), which bypass the oviduct to produce mammalian embryos in vitro. However, it is well known that the oviductal environment affects gametes and embryos and, ultimately, the offspring health. In fact, in vivo-produced embryos are better in terms of morphology, cryotolerance, pregnancy rates or epigenetic profile than those obtained in vitro [21,22,23,24]. The low efficiency of in vitro embryo production has been widely reported in several species; for instance, in porcine the success rate does not exceed 45% [25], polyspermy and insufficient oocyte cytoplasmic maturation being the main causes. In bovine, the ratio of embryos that reach the transferable stage is around 30% to 40% [26], and, in equine species the blastocyst rate reached by intracytoplasmic sperm injection (ICSI) is also around 40% [27].
In an attempt to improve ARTs the tendency followed by researchers has been to mimic the maternal oviductal environment, culturing the embryos with oviductal epithelial cells [62,63] or by supplementing IVF (for a review see [64]) or in vitro culture (IVC) media with different molecules, EVs or natural fluids. EVs play an important role in cell-to-cell communication [65] by transferring their molecular load from one cell to another [7,66]. In the reproductive field, EVs secreted by the oviduct (oEVs) and embryos (eEVs), are key players in the crucial two-way dialogue between the oviduct and the embryo (for a review see [67]). In porcine, the addition of oEVs to the IVF medium regulates polyspermy [68], while oEVs isolated from the conditioned medium of bovine oviductal epithelial cells [69] or from in vivo-derived OF [7,70] improve bovine embryo quality during IVC, in terms of blastocyst rates, cell number, hatching rates, embryo cryosurvival and gene expression. On the other hand, in pig, the efficiency of IVF has been reported to be higher with the addition of OF [68]. The beneficial effect of OF on bovine embryo development and quality has also been reported in terms of cryotolerance, number of trophectoderm cells, gene expression [71] and DNA methylation [72]. A comparison in pig of in vitro-produced embryos with/without reproductive fluids pointed to a higher quality in terms of cell number and ability to hatch in the ART-derived blastocysts when reproductive fluids were added to the culture medium [24]. In this same work, the authors compared the transcriptome and epigenetic profile of in vitro-produced blastocysts without reproductive fluids (C-IVF) and with reproductive fluids (Natur-IVF) and their in vivo counterparts, finding that, the transcriptomic profile showed greater variability in the C-IVF group. Furthermore, the number of differentially expressed genes was higher between the C-IVF and in vivo, than between Natur-IVF and in vivo. Moreover, C-IVF embryos showed more aberrantly expressed genes, which related with epigenetic reprogramming, better embryo development, cell growth and imprinting, and higher methylation levels. All these studies indicate that, an environment similar to physiological conditions during IVC maximizes embryo development and quality. However, many of the components of the OF remain unknown. In the same way, little information is available about the complex molecular dialogue that occurs between the oviduct and the preimplantated embryo (reviewed in [31,73]. Such knowledge is crucial to better understand embryo development and improvement of ARTs.
The aim of this review was to take a deep look at recent discoveries about the oviduct in different species in order to better understand the importance of this organ in the periconception period. With this in mind, a comparative view of the impact of the embryo on oviductal gene expression and the impact of the oviduct on the embryo are analyzed and discussed, taking into consideration the most recent publications in the field. Finally, a comparative study among mammalian species was made of the molecular structure, synthesis and role of oviductin (OVGP1), the most abundant protein in the OF during fertilization and early embryonic development.

2. Materials and Methods

Transcriptomics data concerning the oviduct were taken from the studies done in bovine [74], equine [75] and porcine species [76]. These works were chosen because they use oviducts in vivo and they compare directly the transcriptomic profile of an oviduct with embryos vs. an oviduct in the equivalent stage of the oestrous cycle. Differentially expressed genes (DEGs) from each table were reanalyzed according to the most recent annotation in order to identify former LOC uncharacterized genes (named LOC) whose orthologs have been discovered. Moreover, in the case of the equine species, plenty of mRNAs coding for equine transposases and species-specific endogenous retroviruses were discarded for the cross-species comparison. To compare the DEGs across species, Venn diagrams with all the DEGs and with the down-regulated and up-regulated genes were made.
RNAseq data of in vitro cultured embryos were extracted from the GEO DataSets repository as follows: bovine data from GSE52415 [77], porcine data from GSE139512 [78] and human data from Xue et al., 2013 [79]. Briefly, the fastq files for each dataset were downloaded and transcript expression was quantified with kallisto [80], gene expression was later summarized with an in-home script written in R which makes use of the biomaRt package [81] in order to identify which transcripts belong to a common gene. This approach, using the same workflow for all the datasets, was done in order to minimize differences due to different processing of the data by the original authors. A mean expression in all the available replicates over a detection threshold of 1 TPM (transcript per million) was established in order to filter out non-expressed genes.
Proteomics data involving oviductal fluid samples were obtained from the supplementary files of the studies done in bovine [82,83], equine [84], human [85] and porcine. The study by Canha-Gouveia and coworkers (2019) [85] was to our knowledge the more complete and recent compilation of OF proteins in the human species, including samples corresponding to the secretory phase of the menstrual cycle. When working with bovine data we chose the most recent studies on whole OF by Lamy et al. (2016) [82] and Pillai et al. (2017) [83]. We did not consider discriminating between estrous cycle phases as in the supplementary materials, the data was merged. For the porcine species, we compiled our data from several proteomic assays done with porcine OF from the early luteal stage. We provide a Table S1 with the detected peptide and proteins from each sample. The detailed methods concerning the HPLC Liquid Chromatography-Mass Spectrometry (LC-MS/MS) workflow are described by Luongo et al., 2020 [86].
The database of ligand-receptor interactions in humans compiled by Ramilowski et al., 2015 [87] was used to infer interactions between embryo receptors and oviductal proteins. Selected interactions for each species involved a ligand-receptor pair as described: a ligand present in the oviductal fluid proteome interacting with a receptor present in the blastomere according to the RNAseq analysis.

3. Impact of Embryo and Oviduct Communication

3.1. A Comparative View of the Impact of the Embryo on Oviductal Gene Expression

It was previously reported that a change in the oviductal gene expression pattern occurs in the presence of the embryo in cow, sow and mare [74,75,76], demonstrating the communication between the embryo (or embryos) and the oviduct. We have performed an in silico analysis based on the above mentioned transcriptomic studies in the sow, cow and mare, comparing the gene expression in the oviduct in the presence of an 8-cell embryo (4-cell embryo in the sow) vs. the absence of fertilization. It was observed that more than 250 genes are differentially expressed (DEG) in both conditions in the three species (Figure 1a, Table S2). However, was a surprise to find that very few genes coincided among the three species and only one gene, which corresponds to the MHC-I, is shared among them. When the DEGs were analyzed in more detail, up regulation was observed in 120, 167 and 117 genes in cow, mare and sow, respectively (Figure 1b). In the case of down regulated genes, the data pointed to 147, 90 and 235 in cow, mare and sow, respectively (Figure 1c). These data clearly show that the embryo has an impact on the oviduct, as previously reported in mouse [88], pig [89] and cow [90], but the oviductal response is species-specific. It is not clear if these specific differences are really important but previous studies using heterologous transfer of the embryos showed that the embryos developed well in a foreign oviduct [91]. We are aware of the limitations of our approach: the use of polyovulatory vs. monoovulatory species, different transcriptomics setups (RNAseq vs. microarrays) and different days of sampling after ovulation. Nevertheless, is it also remarkable that all those studies detect a change in genes involved in immune response within the oviduct, suggesting a conserved mechanism of embryo recognition. More research is needed to clarify the impact of these specific genes in each species. Compiling temporal data in those species could be interesting to know more about the evolving scenario across the early embryo development, and to produce the best reproducible results across independent studies.

3.2. Impact of the Oviduct on the Embryo

The embryo changes its gene expression pattern in the oviduct or in co-culture with oviductal epithelial cells [92]. A further analysis was performed to obtain information of potential proteins that have a clear impact in the embryo due to their release into the oviductal lumen and their direct contact with the embryo. It is known that proteins can be secreted extracellularly by different mechanisms [93]. A classical mechanism is exocitosis due to the presence of a signal peptide at the N-terminal region of the protein. This peptide directs the protein towards the secretory pathway due to the participation of two relevant organelles, the endoplasmic reticulum and the Golgi apparatus. An alternative way that has been studied recently in several organs involves the release of the proteins and other components through EVs, including exosomes [94]. It is known that the zona pellucida (ZP) is a porous extracellular matrix that surrounds the oocyte and the early embryos, and is permeable to proteins and also to viruses [95]. Thus, it was reported that one of the most abundant secreted proteins, OVGP1 crosses the ZP, enters the perivitelline space and is endocytosed by the blastomeres [96]. Recently, it was found that exosomes are able to pass through the ZP to reach the embryo [7]. Consequently, both types of secretion have a potential impact on the embryo. For this reason, expressed genes or proteins were investigated using the bioinformatic tool DAVID (v 6.8, [97]) considering “secreted” or “exosome” (Figure 2).
In the cow, it was observed that a total of 100 DEGs are classified as secreted or exosomal proteins (Figure 2a): 16 and 47 are classified as secreted and exosomal proteins, respectively, and 37 as being secreted from both sources. In the mare, it was observed that a total of 62 DEGs are classified as secreted or exosomal proteins (Figure 2b): 19 secreted and 31 exosomal proteins, with 12 coming from both sources.
In the sow, 80 DEGs were classified as secreted or exosomal proteins (Figure 2c): 7 and 57 classified as secreted and exosomal proteins, respectively, and 16 as from both sources. All these proteins had a specific pattern showing up or down regulation (Table S3). However, it is important to take into consideration a technical point that could be responsible for some of the differences, which may be due to the samples analyzed. Thus, the analysis performed in the cow and mare was made using oviductal epithelial cells, while in the sow the whole isthmus region of the oviduct was used.
Additionally, we analyzed the potential receptors on the embryo and the ligands on the oviductal fluid proteins in the cow, pig and human (Table 2). Thus, in the bovine model we detected 99 receptors on the blastomere plasma membrane that may interact with the 58 oviductal proteins (Table S6).
The OF proteomics data are taken from Canha-Gouveia et al., 2019 [85] for woman, Lamy et al., 2016 [82] and Pillai et al., 2017 [83] for cow. The data for sow are based on unpublished research by our group involving 465 proteins (a more modest number compared to the aforementioned studies in human and bovine which detected more than 1200 proteins in each). RNAseq data for 8-cell embryos were taken from Xue et al., 2013 [79] for human samples, from Graf et al., 2014 [77] for bovine embryos, and Kong et al., 2020 [78] for porcine samples. Protein-ligand interactions were extracted from the database generated by Ramilowski et al., 2015 [87].
In human, we detected interactions between 63 receptors on the blastomere surface and 49 oviductal proteins for a total of 119 possible interactions (Table S5), while in porcine there are 32 receptors on the blastomere surface that may interact with 22 oviductal proteins for a total of 56 potential interactions (Table S7). In addition, 45 interactions were shared between human and cow, 21 were shared between pig and cow and 11 were conserved between the three species (Table S4).
The mRNA codifying the receptors was demonstrated using RNAseq analysis expression data from eight-cell embryos. In this stage of pre-implantational development, the embryos have gone through the main events of embryonic genome activation [77,79,103], so it can be considered that they are responding to their environment, as there has been a radical change in their transcriptomic profile compared with the oocyte. A limitation of our approach is that all the embryos collected were obtained in vitro and not in vivo so the reception expression pattern could differ in the in vivo conditions.
The interaction between the OF ligands and their receptors in the blastomeres could be more complex than expected. For example, in the case of the fibronectin present in the OF [104], it can interact with at least 19 receptors of the bovine embryo. Information about the specific pair and ligand interaction is lacking, but it has been observed that fibronectin, a protein typical of the extracellular matrix, is present in the bovine OF [83].
Fibronectin has been shown to induce embryo cleavage but only at an appropriate concentration [105]. In their study, Larson et al. [105] also demonstrated the presence of fibronectin receptors on eight-cells embryos using immunocytochemistry. Fibronectin is present in human and bovine OFs, and its RGD motifs (tripeptide Arg-Gly-Asp, amino acid sequence within the extracellular matrix protein) bind to heterodimeric integrins containing Integrin Subunit Alpha V (ITGAV) [106], both present in human and bovine eight-cell embryos. In contrast, no fibronectin has been detected in porcine species, but as a counterpart, porcine OF has greater quantities of osteopontin (which has also been detected by western blot by Gabler and collaborators in 2003 [107] in bovine OF). This is another protein containing RGD motif, which might work in the same way as fibronectin by stimulating the same receptor.
Among the eight common interactions between the three studied species, the best known ligand present in the OF is lactoferrin (or lactotransferrin, LTF), a glycoprotein secreted by the oviduct to the OF in mouse, human and sheep [104,108,109]. Ward and collaborators [102] detected the expression of this protein by pre-implantational murine embryos, and also the uptake of exogenous protein added to the culture medium, although the receptor present in the blastomeres was not characterized. While LTF is not an essential protein for fertilization, as shown by Ward et al., in a knock-out mouse model [110], it could be interesting to supplement it to an in vitro culture medium at physiological concentrations.
In addition, annexin A1 (ANXA1) is a protein which has been recently identified as an embryo-interacting maternal protein from bovine OF by Banliat, and collaborators in 2020 [98], and has also been immunodetected within the perivitelline space and inside.
Complement component 3 (C3) is another protein present in the OF, the concentration of which is under hormonal regulation in the human, mouse and pig [99,111]. Although Lee, and collaborators in 2004 [99] observed a positive effect of C3 in the size and hatching rates of mouse blastocysts, Georgiou and collaborators in 2011 [112] observed a detrimental effect on porcine blastocysts, suggesting that in this species the C3 concentrations are high during oestrus [111] and decrease in response to the oocyte [100].
Another interesting ligand is the blood protein fibrinogen. Fibrinogen alpha and beta were detected within bovine and porcine OF, while fibrinogen gamma, was also detected in human OF. The role of this protein in reproduction in equine species is known to mediate the implantation of the conceptus in the uterus at about day 40 [101]. The equine conceptus produces fibrinogen on its own (a rare feature, because in adult mammals it is only produced by the liver) and this fibrinogen binds integrins within the mare endometrium.
Furthermore, it may seem surprising that most OF proteins in our list of interactions are actually blood proteins that reach the OF by transudation. It was first thought that this would better explain the above-mentioned successful development of pre-implantational embryos after heterologous transfer, as blood plasma proteins would play a more relevant role than the transcriptomic change in oviductal secreted proteins, which seems to differ greatly between equine, bovine and porcine if we only take into account DEG. However, among the proteins secreted by the oviductal epithelium there are also several ligands with an unknown receptor, so that, they are omitted from our list. Perhaps, one of the most abundant and most intensively studied oviductal proteins is OVGP1, which, after traversing the ZP, is endocytosed by the oocytes or the embryos, in a mechanism that requires the presence of a specific region [113] as explained in more detail below.
Nevertheless, oviductal transcriptomics would also affect oviduct permeability, which explains why the transudate varies during the oestrus cycle and pre-implantational development, as observed by changes in OF volume and total protein concentration [114]. Some authors have also observed changes in concentrations in some plasma proteins in the presence of embryos but not in their absence in the bovine and equine oviduct [84,115].

4. OVGP1

The oviduct-specific glycoprotein OVGP1, also known as oviductin, is the major non-serum glycoprotein present in OF. The components of OF have been studied for decades, and it was Oliphant et al., 1984 [116] who described a protein which was synthesized de novo by the oviduct in rabbit under oestrogen control and was localized in the apical secretory granules, released to the oviductal lumen. In 1986, Brown and Cheng [117] confirmed that oestrus-induced oviductal glycoproteins interact with the porcine egg ZP, and, in 1988 Oikawa and collaborators [118] described a glycoprotein of oviductal origin which altered biochemical properties of the ZP of hamster eggs. Thenceforth different names have been given to this glycoprotein, and through the years more information became available (See [119]). Although, the generation of the null mutation of the oviduct-specific glycoprotein gene indicates that OVGP1 is not essential for the process of in vivo fertilization in mice [120], numerous studies have demonstrated the fundamental role of OVGP1 in reproductive processes and embryonic development in different species of mammals.

4.1. Origin and Localization of OVGP1

OVGP1 is a glycoprotein that has been detected in a number of mammalian species (Table 3), where it is expressed by the non-ciliated epithelial cells of the oviduct. mRNA of OVGP1 has been localized in basal perinuclear compartments and in the apical cytoplasm of fimbria and ampulla epithelial cells in sheep [121], while the protein has been identified in Golgi saccules and in secretory granules of the non-ciliated oviduct cells in hamster [122,123,124] and inside secretory granules in bovine [125,126], porcine [127], baboon [128], human [129], mouse [130] and rabbit [116]. The fact that the protein has not been detected in rat [131] is due to OVGP1 gene is lost/absent in rats. It is consistent with the reported information about OVGP1 gene existing before the divergence of mammals and being from this species [132]. In megabat it is also considered a pseudogene [133].
Traditionally, it has been described that OVGP1 is solely produced by the oviduct [163,177] while the detection of the protein in the lining of the surface epithelia of the uterus and the uterine fluid [104] was attributed to the transfer of components from the OF. The protein was observed in the epithelial cells of the endometrium in mice, but only at the time of embryo implantation [178]. As regards implantation, women suffering recurrent implantation failure showed significant low OVGP1 mRNA in the endometrium [178]. Whatever the case, the oviduct would be the main organ responsible for the synthesis of this glycoprotein as a component of the OF. The oviduct of the pig [164], sheep [179] and cow [143], shows an increased biosynthetic activity at oestrous, suggesting that the production of more abundant glycoprotein is oestrogen-dependent and is at its highest level at the time of fertilization in some species such as human, cow, pig, sheep, baboon and mice [104,131,135,136,138,166,173,175,180,181,182,183,184,185]. At the time of maximum OVGP1 gene transcription in the oviductal tissue, there is an increase in the number of apical secretory granules in the oviductal epithelium containing OVGP1 and also in the amount of OVGP1 in OF [119]. The OVGP1 gene has been reported to be down-regulated in pregnant compared with cyclic heifers [74]. However, the expression of OVGP1 in some species, such as hamster and rabbit is controversial, as some authors describe no differences in mRNA production during the oestrous cycle [186,187]. Others observed that mRNA levels changed significantly between the oestrous and diestrous stages in the ampulla, but not in the isthmus, of a constitutively expressed OVGP1 [188], and also mention stage-specific OVGP1 expression in the secretory granules of the hamster oviductal secretory cells, with a maximum level at oestrus [189]. In addition, the induction of the production of the protein in new born hamsters by oestradiol has been reported [155].
Moreover, there are also located differences in where oviductal secretion take place [115]. The oviductal biosynthetic activity is major in some functional segments like the infundibulum and the ampulla at oestrous in pigs [164,190] so that, the type and distribution of OVGP1 glycoprotein differs between the ampulla and isthmus in pig and sheep [165,179]. Moreover, the levels of OVGP1 expression detected in these regions vary between species. OVGP1 is synthesized and released by the oviduct in a temporally and regionally specific way in the ewe [191]. In rabbit, the ampulla secretes greater amounts of OVGP1 than in the isthmus [192,193]. Similarly, using RNAseq technology, Gonella- Diaza and collaborators [194] showed greater OVGP1 expression in the ampulla than in the isthmus, but no differences were perceived when distinct periovulatory endocrine profiles were analyzed in cattle. On the contrary, the same mRNA levels of OVGP1 were detected in the fimbria, ampulla and isthmus in baboon [182] and no spatial differences between the transcriptome of the isthmus and ampulla in cow was reported [6]. In mouse, cow and sheep, the protein was not localized in the isthmus [126,130,163,174,175], while in cow, sheep, pig and mouse it has been immunolocalized in the infundibulum [119].
The oEVs, recently mentioned as components contained in the OF and described as an important modulator between gametes and embryos and the maternal tract, are released by oviductal epithelial cells at different stages of the oestrous cycle [8]. OVGP1 transcript and protein have been detected inside porcine and bovine oEVs produced in vivo, suggesting that its secretion is, at least in part, by oEVs [7,8,68]. The molecular composition of bovine oviductal oEVs is regulated by steroid hormones [8]. However, no temporal differential mRNA nor OVGP1 protein concentration was detected in oviductal oEVs at any time during the bovine oestrous cycle [8], although this protein was identified as one of the most expressed proteins in the oviduct [8]. MYH9, the OVGP1-protein binding partner in gametes [195] was also detected in oEV samples [7,8,68].

4.2. Characterization of OVGP1

OVGP1 is a mucin belonging to a protein family named glycoside hydrolase 18, which shares a chitinase catalytic domain that is not active in OVGP1 [182,196], due to the lack of an essential glutamic acid residue in the N-terminal domain of the protein [119,197]. The protein contains a mucin-type tandem repeat, a signal peptide and several post-translational modification sites involved in secretion [119], as well as a clatrhin box associated with endocytosis. In human, baboon, porcine and bonnet a Class III PDZ-binding domain has been reported, which suggests that OVGP1 could be part of a multi-protein complex [198].
Analysis of the amino acid sequence of OVGP1 exhibited that the N-terminal region of mature OVGP1 shares a high degree of identity (77–84%) and similarity (86–90%) with other species. In contrast, the C-terminal region has a low degree of identity (37–63%) and similarity (50–75%), as well as several insertions/deletions in its sequence [113]. A comparative analysis of the alignment of deduced aminoacidic sequences of several mammalian OVGP1 proteins revealed the existence of five differentiated regions (A–E). Region A, which corresponds to the N-terminus, has a high degree of identity in monotremes, marsupials and placental mammals. Region B shows low identity among different mammals and contains multiple insertions/deletions. Region C is an insertion present only in the mouse, and region E is typical of human, chimpanzee and orang-utan [3]. Analysis, at both the molecular and physiological level, of the role of the C-terminal region of OVGP1 in fertilization provided a model in which OVGP1 binds to ZP via its highly conserved A region, which may also provide an anchor for additional oviduct proteins. The C-terminal regions of OVGP1 modulate its binding to the ZP, regulate OVGP1 activity and account for the reproductive role of OVGP1 in different mammalian orders [113].
The apparent molecular mass of OVGP1 glycoprotein on reducing SDS-PAGE ranges from 66 to 350 kDa in different species [177]. Carbohydrates make a large contribution to the weight of the protein. At least two isoforms of the protein [119], as well as polymorphism in the gene sequence [3,186,187], have also been reported. Oddly, a 95 kDa bovine OVGP1 isoform has been identified inside EVs, which is more than the 75 kDa found in the OF [68].

4.3. Effect of OVGP1 Localized in Eggs and Embryos

OVGP1 has been associated with the ZP of embryos and oocytes from different species, both in homologous and heterologous systems, as well as with blastomere plasma membranes, inside blastomeres, endosomes, lysosomes, multivesicular bodies and with the perivitelline space, as shown in Table 4, suggesting the protein role in the regulation of fertilization and early embryo development. The association of OVGP1 to the ZP is stable and uniform and is maintained until day 7 in bovine uterine embryos [199] and was also detected in 7–day old hatched porcine uterine embryos [127]. The protein complex formed by OVGP1 and heparin-like glycosaminoglycans is an important regulator of OVGP1 binding to the ZP since their union is considered reversible [200]. Recombinant porcine OVGP1 was detected through the whole thickness of the ZP of porcine and bovine oocytes when it was present during the IVF [113,201]. It was also found bound to the ZP in 9-day old bovine embryos, but only if the in vitro culture medium contained OVGP1 until day 3.5 post-insemination [201]. Using OF during the incubation of in vitro-produced bovine embryos, OVGP1 was localized in the perivitelline space and in blastomeres of both 4–6 cell and morula-stage embryos, but not bound to the ZP [98]. This agrees with the previously described reversible nature of OVGP1-ZP binding [200], whereby only two bovine OVGP1 proteins of 75 and 95 kDa were reported to bind porcine ZP [200]. In mouse, OVGP1 was associated with the perivitelline space of oocytes and embryos [130,202,203] but only a peanut agglutinin-binding glycoform of the protein was associated with the ZP [203].
As regards the role of OVGP1 in fertilization, ZP exposure to OVGP1 generally results in modification of the ZP and an increase in resistance to digestion by proteolytic enzymes (enzymatic hardening of the ZP) [149,167,210,211,212,213,214,215,216,217], which, in turn, contributes to the control of polyspermy [200] and improves the efficiency of in vitro fertilization [113,218]. Introducing specific antibodies against the protein blocked the observed biological effect [146,167]. Although one publication reported no ZP hardening after the treatment of porcine oocytes with OVGP1 protein purified from pig OF [167], the ZP of porcine oocytes exposed to both, purified recombinant porcine and rabbit OVGP1 increased ZP resistance to enzymatic proteolysis in a dose-dependent manner [113] but only homologous recombinant protein increased the efficiency of fertilization. Thus, the correlation between the induction of hardening and improved IVF rates only occurs in homologous systems. Indeed, human OVGP1 enhances sperm binding to the ZP, whereas heterologous OVGP1 (baboon) inhibits this effect even though the two proteins are 94% identical [137] the same being observed in hamster [219]. The presence or absence of specific regions in the C-terminus of OVGP1 affects its association with the ZP, as well as its ability to remodel the matrix and so its effect on fertilization [113]. The pre-treatment of oocytes with OVGP1 increases sperm-egg binding [207] and zona penetration rates in hamster [220] and human [137], but the opposite effect occurs in pig [200].
Moreover, the presence of the glycoprotein inside the embryo suggests it plays role in embryo development. Notably, antibodies against a C-terminal peptide of OVGP1 inhibit early mouse development, so that embryos do not progress from the 2-cell stage [221]. Indeed, purified porcine OVGP1 enhanced cleavage and the blastocyst formation rate in in vitro-produced porcine embryos when they were cultured in a medium supplemented with the protein for 48 or 144 h [218]. Using purified porcine OVGP1 during preincubation and IVF increased their post-cleavage development to blastocyst [167]. Similarly, the supplementation of the IVF medium with an OVGP1-enriched fraction of OF increased cleavage rates in ovine [222]. By contrast, using ovine OVGP1 in IVC decreased the proportion of zygotes undergoing the first cleavage, increased the time needed for blastocyst formation, and also the mean number of nuclei per blastocyst, resulting in blastocysts developing more similarly to the those produced in vivo [222,223]. Moreover, supplementing the IVM, IVF and IVC medium with purified OVGP1 from goat oviductal tissue increased the cleavage rate, and morula and blastocyst yield at the lowest used concentration (10 µg/mL), but had an inhibitory effect at higher concentrations (50 and 100 µg/mL) [149]. A feline recombinant OVGP1 expressed in a bacterial expression system did not affect cleavage, morula or blastocyst rates in cat, but increased the relative mRNA level of the GJA1 gen, an embryo quality marker [141]. In addition, the use of recombinant porcine OVGP1 during bovine IVF or embryo IVC did not affect cleavage or blastocyst yield but resulted in embryos with an increased relative abundance of mRNA of embryo quality marker genes as: aquaporins (AQP3), transcription factor (ATF4), cell adhesion proteins (DSC2) and methyltransferase (DNMT3A) [201].
The reported effects of OVGP1 glycoprotein differs from species to species. It is also important to bear in mind the great variability in the experimental design used: different OVGP1 sources, protein concentrations, times of OVGP1 exposure, among others. Furthermore, the recent discovery of OVGP1 in EVs could determine the protein activity controlled by this system, which has not been described to date. In addition to the species-specific effect, all the above could explain the varied effects described for this oviductal glycoprotein. New gene editing techniques used in species other than murine, might help clarify in determining role of this protein in different species of mammals during periconception period.

Supplementary Materials

The following are available online at https://www.mdpi.com/2218-273X/10/12/1690/s1, Table S1: Deletes peptides Sus scrofa, Table S2: DEGs oviduct cow + mare + so, Table S3: Exosomal vs secreted, Table S4: Conserved interactions, Table S5: Human interactions, Table S6: Cow interactions, Table S7: Pig interactions.

Funding

Supported by Fundación Seneca-Agencia de Ciencia y Tecnología de la Región de Murcia “Ayudas a la realización de proyectos para el desarrollo de investigación científica y técnica por grupos competitivos 2018” (20887/PI/18) and PGC2018-094781-B-I00 ((MCINN/AEI/FEDER, UE) from the Ministry of Economy and Competitiveness (Spain).

Acknowledgments

The authors thanks Alejandro Torrecillas Sánchez from the Molecular Biology Service (ACTI, University of Murcia, Spain) for his technical assistance and advice.

Conflicts of Interest

The authors declare no conflict of interest.

Date Availability

All data generated or analyzed during this study are included in this published article (and its supplementary information files).

References

  1. Woodruff, J.D.; Pauerstein, C.J. The Fallopian Tube: Structure, Function, Pathology, and Management; Williams & Wilkins Co.: Washington, DC, USA, 1969. [Google Scholar]
  2. Hunter, R.H.F. Have the Falopian Tubes a Vital Role in Promoting Fertility? Acta Obs. Gynecol. Scand. 1998, 77, 475–486. [Google Scholar]
  3. Avilés, M.; Gutiérrez-Adán, A.; Coy, P. Oviductal Secretions: Will They Be Key Factors for the Future ARTs? Mol. Hum. Reprod. 2010, 16, 896–906. [Google Scholar] [CrossRef]
  4. Holt, W.V.; Fazeli, A. The Oviduct as a Complex Mediator of Mammalian Sperm Function and Selection. Mol. Reprod. Dev. 2010, 77, 934–943. [Google Scholar] [CrossRef] [PubMed]
  5. Coy, P.; Garcia-Vasquez, F.A.; Visconti, P.E.; Aviles, M. Roles of the Oviduct in Mammalian Fertilization. Reproduction 2012, 144, 649–660. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Maillo, V.; De Frutos, C.; O’Gaora, P.; Forde, N.; Burns, G.W.; Spencer, T.E.; Gutierrez-Adan, A.; Lonergan, P.; Rizos, D. Spatial Differences in Gene Expression in the Bovine Oviduct. Reproduction 2016, 152, 37–46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Alminana-Brines, C.; Corbin, E.; Tsikis, G.; Neto, A.S.D.A.; Labas, V.; Reynaud, K.; Galio, L.; Uzbekov, R.; Garanina, A.S.; Druart, X.; et al. Oviduct Extracellular Vesicles Protein Content and Their Role During oviduct–embryo Cross-Talk. Reproduction 2017, 154, 253–268. [Google Scholar] [CrossRef] [PubMed]
  8. Almiñana, C.; Tsikis, G.; Labas, V.; Uzbekov, R.E.; Da Silveira, J.C.; Bauersachs, S.; Mermillod, P. Deciphering the Oviductal Extracellular Vesicles Content across the Estrous Cycle: Implications for the Gametes-Oviduct Interactions and the Environment of the Potential Embryo. BMC Genom. 2018, 19, 1–27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Pérez-Cerezales, S.; Ramos-Ibeas, P.; Acuña, O.S.; Avilés, M.; Coy, P.; Rizos, D.; Gutiérrez-Adán, A. The Oviduct: From Sperm Selection to the Epigenetic Landscape of the embryo†. Biol. Reprod. 2018, 98, 262–276. [Google Scholar] [CrossRef] [Green Version]
  10. Duranthon, V.; Watson, A.J.; Lonergan, P. Preimplantation Embryo Programming: Transcription, Epigenetics, and Culture Environment. Reproduction 2008, 135, 141–150. [Google Scholar] [CrossRef] [Green Version]
  11. Lee, M.T.; Bonneau, A.R.; Takacs, C.M.; Bazzini, A.A.; DiVito, K.R.; Fleming, E.S.; Giraldez, A.J. Nanog, Pou5f1 and SoxB1 Activate Zygotic Gene Expression During the Maternal-to-Zygotic Transition. Nat. Cell Biol. 2013, 503, 360–364. [Google Scholar] [CrossRef] [Green Version]
  12. Ko, M.S.H. Zygotic Genome Activation Revisited: Looking through the Expression and Function of Zscan4. In Current Topics in Developmental Biology; Academic Press: Cambridge, MA, USA, 2016; Volume 120, pp. 103–124. [Google Scholar]
  13. Wu, E.; Vastenhouw, N.L. From Mother to Embryo: A Molecular Perspective on Zygotic Genome Activation. Curr. Top. Dev. Biol. 2020, 209–254. [Google Scholar] [CrossRef]
  14. Salilew-Wondim, D.; Saeed-Zidane, M.; Hoelker, M.; Gebremedhn, S.; Poirier, M.; Pandey, H.O.; Tholen, E.; Neuhoff, C.; Held, E.; Besenfelder, U.; et al. Genome-Wide DNA Methylation Patterns of Bovine Blastocysts Derived from in Vivo Embryos Subjected to in Vitro Culture Before, During or After Embryonic Genome Activation. BMC Genom. 2018, 19, 1–19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Hugentobler, S.A.; Diskin, M.G.; Leese, H.J.; Humpherson, P.G.; Watson, T.; Sreenan, J.M.; Morris, D.G. Amino Acids in Oviduct and Uterine Fluid and Blood Plasma During the Estrous Cycle in the Bovine. Mol. Reprod. Dev. 2007, 74, 445–454. [Google Scholar] [CrossRef] [PubMed]
  16. Simintiras, C.A.; Fröhlich, T.; Sathyapalan, T.; Arnold, G.J.; Ulbrich, S.; Leese, H.J.; Sturmey, R.G. Modelling Aspects of Oviduct Fluid Formation in Vitro. Reproduction 2017, 153, 23–33. [Google Scholar] [CrossRef] [Green Version]
  17. Saint-Dizier, M.; Schoen, J.; Chen, S.; Banliat, C.; Mermillod, P. Composing the Early Embryonic Microenvironment: Physiology and Regulation of Oviductal Secretions. Int. J. Mol. Sci. 2019, 21, 223. [Google Scholar] [CrossRef] [Green Version]
  18. Leese, H.J.; Tay, J.I.; Reischl, J.; Downing, S.J. Formation of Fallopian Tubal Fluid: Role of a Neglected Epithelium. Reproduction 2001, 121, 339–346. [Google Scholar] [CrossRef]
  19. Leese, H.J.; Hugentobler, S.A.; Gray, S.M.; Morris, D.G.; Sturmey, R.G.; Whitear, S.-L.; Sreenan, J.M. Female Reproductive Tract Fluids: Composition, Mechanism of Formation and Potential Role in the Developmental Origins of Health and Disease. Reprod. Fertil. Dev. 2008, 20, 1–8. [Google Scholar] [CrossRef]
  20. Almiñana, C.; Bauersachs, S. Extracellular Vesicles in the Oviduct: Progress, Challenges and Implications for the Reproductive Success. Bioengineering 2019, 6, 32. [Google Scholar] [CrossRef] [Green Version]
  21. Kikuchi, K.; Kashiwazaki, N.; Noguchi, J.; Shimada, A.; Takahashi, R.; Hirabayashi, M.; Shino, M.; Ueda, M.; Kaneko, H. Developmental Competence, After Transfer to Recipients, of Porcine Oocytes Matured, Fertilized, and Cultured in Vitro. Biol. Reprod. 1999, 60, 336–340. [Google Scholar] [CrossRef] [Green Version]
  22. Lonergan, P. State-of-the-Art Embryo Technologies in Cattle. Biosci. Proc. 2019, 64. [Google Scholar] [CrossRef]
  23. Urrego, R.; Rodríguez-Osorio, N.; Niemann, H. Epigenetic Disorders and Altered Gene Expression After Use of Assisted Reproductive Technologies in Domestic Cattle. Epigenetics 2014, 9, 803–815. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Canovas, S.; Ivanova, E.; Romar, R.; García-Martínez, S.; Soriano-Úbeda, C.; García-Vázquez, F.A.; Saadeh, H.; Andrews, S.; Kelsey, G.; Coy, P. Author Response: DNA Methylation and Gene Expression Changes Derived from Assisted Reproductive Technologies Can Be Decreased by Reproductive Fluids. eLife 2017, 6, e23670. [Google Scholar] [CrossRef] [PubMed]
  25. Romar, R.; Funahashi, H.; Coy, P. In Vitro Fertilization in Pigs: New Molecules and Protocols to Consider in the Forthcoming Years. Theriogenology 2016, 85, 125–134. [Google Scholar] [CrossRef]
  26. Lonergan, P.; Fair, T.; Forde, N.; Rizos, D. Embryo Development in Dairy Cattle. Theriogenology 2016, 86, 270–277. [Google Scholar] [CrossRef] [PubMed]
  27. Salgado, R.M.; Brom-De-Luna, J.G.; Resende, H.L.; Canesin, H.S.; Hinrichs, K. Lower Blastocyst Quality After Conventional Vs. Piezo ICSI in the Horse Reflects Delayed Sperm Component Remodeling and Oocyte Activation. J. Assist. Reprod. Genet. 2018, 35, 825–840. [Google Scholar] [CrossRef] [PubMed]
  28. Harada, Y.; Maeda, T.; Fukunaga, E.; Shiba, R.; Okano, S.; Kinutani, M.; Horiuchi, T. Selection of High-Quality and Viable Blastocysts Based on Timing of Morula Compaction and Blastocyst Formation. Reprod. Med. Biol. 2019, 19, 58–64. [Google Scholar] [CrossRef]
  29. Lewis, W. On the Early Development of the Mouse Egg. Carnegie Inst. Contrib. Embryol. 1935, 25, 113–143. [Google Scholar]
  30. Hunter, R.H.F. Chronological and Cytological Details of Fertilization and Early Embryonic Development in the Domestic pig, Sus Scrofa. Anat. Rec. Adv. Integr. Anat. Evol. Biol. 1974, 178, 169–185. [Google Scholar] [CrossRef]
  31. Rizos, D.; Maillo, V.; Sánchez-Calabuig, M.J.; Lonergan, P. The Consequences of Maternal-Embryonic Cross Talk During the Periconception Period on Subsequent Embryonic Development. Adv. Exp. Med. Biol. 2017, 1014, 69–86. [Google Scholar]
  32. Clark, R.T. Studies on the Physiology of Reproduction in the Sheep II. The Cleavage Stages of the Ovum. Anat. Rec. Adv. Integr. Anat. Evol. Biol. 1934, 60, 135–159. [Google Scholar] [CrossRef]
  33. Webel, S.K.; Franklin, V.; Harland, B.; Dziuk, P.J. Fertility, Ovulation and Maturation of Eggs in Mares Injected With HCG. Reproduction 1977, 51, 337–341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Swanson, W.F.; Roth, T.L.; Wildt, D.E. In Vivo Embryogenesis, Embryo Migration, and Embryonic Mortality in the Domestic Cat1. Biol. Reprod. 1994, 51, 452–464. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Reynaud, K.; Fontbonne, A.; Marseloo, N.; Thoumire, S.; Chebrout, M.; De Lesegno, C.V.; Chastant-Maillard, S. In Vivo Meiotic Resumption, Fertilization and Early Embryonic Development in the Bitch. Reproduction 2005, 130, 193–201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Suzuki, O.; Ogura, A.; Asano, T.; Noguchi, Y.; Yamamoto, Y.; Oike, M. Development of Preimplantation Guinea-Pig Embryos in Serum-Free Media. Reprod. Fertil. Dev. 1993, 5, 425–432. [Google Scholar] [CrossRef]
  37. Sultana, F.; Hatori, M.; Shimozawa, N.; Ebisawa, T.; Sankai, T. Continuous Observation of Rabbit Preimplantation Embryos In Vitro by Using a Culture Device Connected to a Microscope. J. Am. Assoc. Lab. Anim. Sci. 2009, 48, 52–56. [Google Scholar]
  38. Hlinka, D.; Kaľatová, B.; Uhrinová, I.; Dolinská, S.; Rutarová, J.; Řezáčová, J.; Lazarovská, S.; Dudáš, M. Time-Lapse Cleavage Rating Predicts Human Embryo Viability. Physiol. Res. 2012, 61, 513–525. [Google Scholar] [CrossRef]
  39. Hamilton, W.; Laing, J. Development of the Egg of the Cow up to the Stage of Blastocyst Formation. J. Anat. 1946, 80, 194. [Google Scholar]
  40. Betteridge, K.J.; Eaglesome, M.D.; Mitchell, D.; Flood, P.F.; Beriault, R. Development of Horse Embryos up to Twenty Two Days After Ovulation: Observations on Fresh Specimens. J. Anat. 1982, 135, 191–209. [Google Scholar]
  41. Renton, J.P.; Boyd, J.S.; Eckersall, P.D.; Ferguson, J.M.; Harvey, M.J.A.; Mullaney, J.; Perry, B. Ovulation, Fertilization and Early Embryonic Development in the Bitch (Canis Familiaris). Reproduction 1991, 93, 221–231. [Google Scholar] [CrossRef] [Green Version]
  42. Denker, H.-W.; Gerdes, H.-J. The Dynamic Structure of Rabbit Blastocyst Coverings. Anat. Embryol. 1979, 157, 15–34. [Google Scholar] [CrossRef]
  43. Fischer, B.; Chavatte-Palmer, P.; Viebahn, C.; Santos, A.N.; Duranthon, V. Rabbit as a Reproductive Model for Human Health. Reproduction 2012, 144, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Spencer, T.E.; Johnson, G.A.; Bazer, F.W.; Burghardt, R.C. Implantation Mechanisms: Insights from the Sheep. Reproduction 2004, 128, 657–668. [Google Scholar] [CrossRef] [PubMed]
  45. Sathananthan, H.; Gunasheela, S.; Menezes, J. Mechanics of Human Blastocyst Hatching in Vitro. Reprod. Biomed. Online 2003, 7, 228–234. [Google Scholar] [CrossRef]
  46. Pratt, H.P. Marking Time and Making Space: Chronology and Topography in the Early Mouse Embryo. Adv. Clin. Chem. 1989, 117, 99–130. [Google Scholar]
  47. Pope, C. Aspects of in Vivo Oocyte Production, Blastocyst Development, and Embryo Transfer in the Cat. Theriogenology 2014, 81, 126–137. [Google Scholar] [CrossRef]
  48. Concannon, P.; Tsutsui, T.; Shille, V. Embryo Development, Hormonal Requirements and Maternal Responses During Canine Pregnancy. J. Reprod. Fertil. 2001, 57, 169–179. [Google Scholar]
  49. Blandau, R.J. Observations on Implantation of the Guinea Pig Ovum. Anat. Rec. Adv. Integr. Anat. Evol. Biol. 1949, 103, 19–47. [Google Scholar] [CrossRef]
  50. Adams, C.E. Egg Transfer in The Rabbit. Mamm. Egg Transf. 2018, 1982, 29–48. [Google Scholar]
  51. Croxatto, H.B. Physiology of Gamete and Embryo Transport through the Fallopian Tube. Reprod. Biomed. Online 2002, 4, 160–169. [Google Scholar] [CrossRef]
  52. Battut, I.; Colchen, S.; Fiéni, F.; Tainturier, D.; Bruyas, J. Success Rates When Attempting to Nonsurgically Collect Equine Embryos at 144, 156 or 168 Hours After Ovulation. Equine Veter J. 2010, 29, 60–62. [Google Scholar] [CrossRef]
  53. Tsutsui, T. Studies on the Reproduction in the Dog. V. On Cleavage and Transport of Fertilized Ova in the Oviduct. Jpn. J. Anim. Reprod. 1975, 21, 70–75. [Google Scholar] [CrossRef] [Green Version]
  54. Harper, M. Gamete and Zygote Transport. In The Physiology of Reproduction; Knobil, E., Neill, J.D., Eds.; Raven Press: New York, NY, USA, 1988; pp. 103–134. [Google Scholar]
  55. Wang, H.; Dey, S.K. Roadmap to Embryo Implantation: Clues from Mouse Models. Nat. Rev. Genet. 2006, 7, 185–199. [Google Scholar] [CrossRef] [PubMed]
  56. Valtonen, M.; Jalkanen, L. Species-Specific Features of Oestrous Development and Blastogenesis in Domestic Canine Species. J. Reprod. Fertil. 1993, 47, 133–137. [Google Scholar]
  57. Hunter, R.H.F.; Hunt, D.M.; Chang, M.C. Temporal and Cytological Aspects of Fertilization and Early Development in the Guinea pig, Cavia Porcellus. Anat. Rec. Adv. Integr. Anat. Evol. Biol. 1969, 165, 411–429. [Google Scholar] [CrossRef]
  58. Cha, J.; Sun, X.; Dey, S.K. Mechanisms of Implantation: Strategies for Successful Pregnancy. Nat. Med. 2012, 18, 1754–1767. [Google Scholar] [CrossRef]
  59. Senger, P.L. Pathways to Pregnancy and Parturition, 2nd ed.; Current Conceptions, Inc.: Redmond, OR, USA, 2003. [Google Scholar]
  60. Denker, H.-W.; Eng, L.A.; Hamner, C.E. Studies on the Early Development and Implantation in the Cat. Brain Struct. Funct. 1978, 154, 39–54. [Google Scholar] [CrossRef]
  61. Holst, P.A.; Phemister, R.D. The Prenatal Development of the Dog: Preimplantation Events1. Biol. Reprod. 1971, 5, 194–206. [Google Scholar] [CrossRef] [Green Version]
  62. Cordova, A.; Perreau, C.; Schmaltz-Panneau, B.; Locatelli, Y.; Ponsart, C.; Mermillod, P. Use of an in Vitro Model in Bovine to Evidence a Functional and Molecular Dialogue Between Preimplantation Embryo and Oviduct Epithelial Cells. Gynecol. Obs. Fertil. 2013, 41, 537–539. [Google Scholar] [CrossRef]
  63. Hamdi, M.; Lopera, R.; Maillo, V.; Núñez, C.; Gutierrez-Adan, A.; Lonergan, P.; Bermejo-Alvarez, P.; Rizos, D. Bovine Oviduct Epithelial Cells: An in Vitro Model to Study Early Embryo-Maternal Communication. Anim. Reprod. 2015, 12, 798. [Google Scholar]
  64. Romar, R.; Cánovas, S.; Matás, C.; Gadea, J.; Coy, P. Pig in Vitro Fertilization: Where Are We and Where Do We Go? Theriogenology 2019, 137, 113–121. [Google Scholar] [CrossRef]
  65. Valadi, H.; Ekström, K.; Bossios, A.; Sjöstrand, M.; Lee, J.J.; Lötvall, J.O. Exosome-Mediated Transfer of MRNAs and MicroRNAs Is a Novel Mechanism of Genetic Exchange Between Cells. Nat. Cell Biol. 2007, 9, 654–659. [Google Scholar] [CrossRef] [Green Version]
  66. Raposo, G.; Stoorvogel, W. Extracellular vesicles: Exosomes, microvesicles, and friends. J. Cell Biol. 2013, 200, 373–383. [Google Scholar] [CrossRef] [Green Version]
  67. Almiñana, C.; Bauersachs, S. Extracellular Vesicles: Multi-Signal Messengers in the gametes/Embryo-Oviduct Cross-Talk. Theriogenology 2020, 150, 59–69. [Google Scholar] [CrossRef]
  68. Alcântara-Neto, A.S.; Fernandez-Rufete, M.; Corbin, E.; Tsikis, G.; Uzbekov, R.; Garanina, A.S.; Coy, P.; Almiñana, C.; Mermillod, P. Oviduct Fluid Extracellular Vesicles Regulate Polyspermy During Porcine in Vitro Fertilisation. Reprod. Fertil. Dev. 2020, 32, 409–418. [Google Scholar] [CrossRef]
  69. Lopera-Vasquez, R.; Hamdi, M.; Fernandez-Fuertes, B.; Maillo, V.; Beltrán-Breña, P.; Calle, A.; Redruello, A.; López-Martín, S.; Gutiérrez-Adán, A.; Yáñez-Mó, M.; et al. Extracellular Vesicles from BOEC in In Vitro Embryo Development and Quality. PLoS ONE 2016, 11, e0148083. [Google Scholar] [CrossRef]
  70. Lopera-Vasquez, R.; Hamdi, M.; Maillo, V.; Gutierrez-Adan, A.; Bermejo-Alvarez, P.; Ramírez, M.Á.; Yáñez-Mó, M.; Rizos, D. Effect of Bovine Oviductal Extracellular Vesicles on Embryo Development and Quality in Vitro. Reproduction 2017, 153, 461–470. [Google Scholar] [CrossRef] [Green Version]
  71. Lopera-Vasquez, R.; Hamdi, M.; Maillo, V.; Lloreda, V.; Coy, P.; Gutiérrez-Adán, A.; Bermejo-Álvarez, P.; Rizos, D. Effect of Bovine Oviductal Fluid on Development and Quality of Bovine Embryos Produced in Vitro. Reprod. Fertil. Dev. 2017, 29, 621. [Google Scholar] [CrossRef]
  72. Hamdi, M.; Lopera-Vasquez, R.; Maillo, V.; Sánchez-Calabuig, M.J.; Núnez, C.; Gutiérrez-Adán, A.; Rizos, D. Bovine Oviductal and Uterine Fluid Support In Vitro Embryo Development. Reprod. Fertil. Dev. 2018, 30, 935. [Google Scholar] [CrossRef]
  73. Fazeli, A.; Holt, W.V. Cross Talk During the Periconception Period. Theriogenology 2016, 86, 438–442. [Google Scholar] [CrossRef]
  74. Maillo, V.; Gaora, P.Ó.; Forde, N.; Besenfelder, U.; Havlicek, V.; Burns, G.W.; Spencer, T.E.; Gutiérrez-Adán, A.; Lonergan, P.; Rizos, D. Oviduct-Embryo Interactions in Cattle: Two-Way Traffic or a One-Way Street? Biol. Reprod. 2015, 92, 144. [Google Scholar] [CrossRef]
  75. Smits, K.; De Coninck, D.I.; Van Nieuwerburgh, F.; Govaere, J.; Van Poucke, M.; Peelman, L.; Deforce, D.; Van Soom, A. The Equine Embryo Influences Immune-Related Gene Expression in the Oviduct1. Biol. Reprod. 2016, 94, 36. [Google Scholar] [CrossRef] [Green Version]
  76. Martyniak, M.; Zglejc-Waszak, K.; Franczak, A.; Kotwica, G. Transcriptomic Analysis of the Oviduct of Pigs During the Peri-Conceptional Period. Anim. Reprod. Sci. 2018, 197, 278–289. [Google Scholar] [CrossRef]
  77. Graf, A.; Krebs, S.; Zakhartchenko, V.; Schwalb, B.; Blum, H.; Wolf, E. Fine Mapping of Genome Activation in Bovine Embryos by RNA Sequencing. Proc. Natl. Acad. Sci. USA 2014, 111, 4139–4144. [Google Scholar] [CrossRef] [Green Version]
  78. Kong, Q.; Yang, X.; Zhang, H.; Liu, S.; Zhao, J.; Zhang, J.; Weng, X.; Jin, J.; Liu, Z. Lineage Specification and Pluripotency Revealed by Transcriptome Analysis from Oocyte to Blastocyst in Pig. FASEB J. 2019, 34, 691–705. [Google Scholar] [CrossRef] [Green Version]
  79. Xue, Z.; Huang, K.; Cai, C.; Cai, L.; Jiang, C.-Y.; Feng, Y.; Liu, Z.; Zeng, Q.; Cheng, L.; Sun, Y.E.; et al. Genetic Programs in Human and Mouse Early Embryos Revealed by Single-Cell RNA Sequencing. Nat. Cell Biol. 2013, 500, 593–597. [Google Scholar] [CrossRef] [Green Version]
  80. Bray, N.L.; Pimentel, H.; Melsted, P.; Pachter, L. Near-Optimal Probabilistic RNA-Seq Quantification. Nat. Biotechnol. 2016, 34, 525–527. [Google Scholar] [CrossRef]
  81. Durinck, S.; Spellman, P.T.; Birney, E.; Huber, W. Mapping Identifiers for the Integration of Genomic Datasets With the R/Bioconductor Package BiomaRt. Nat. Protoc. 2009, 4, 1184–1191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Lamy, J.; Labas, V.; Harichaux, G.; Tsikis, G.; Mermillod, P.; Saint-Dizier, M. Regulation of the Bovine Oviductal Fluid Proteome. Reproduction 2016, 152, 629–644. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Pillai, V.V.; Weber, D.M.; Phinney, B.S.; Selvaraj, V. Profiling of Proteins Secreted in the Bovine Oviduct Reveals Diverse Functions of This Luminal Microenvironment. PLoS ONE 2017, 12, e0188105. [Google Scholar] [CrossRef] [Green Version]
  84. Smits, K.; Nelis, H.; Van Steendam, K.; Govaere, J.; Roels, K.; Ververs, C.; Leemans, B.; Wydooghe, E.; Deforce, D.; Van Soom, A. Proteome of Equine Oviducal Fluid: Effects of Ovulation and Pregnancy. Reprod. Fertil. Dev. 2017, 29, 1085. [Google Scholar] [CrossRef]
  85. Canha-Gouveia, A.; Paradela, A.; Ramos-Fernández, A.; Prieto-Sánchez, M.T.; Sánchez-Ferrer, M.L.; Corrales, F.; Coy, P. Which Low-Abundance Proteins are Present in the Human Milieu of Gamete/Embryo Maternal Interaction? Int. J. Mol. Sci. 2019, 20, 5305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Luongo, C.; González-Brusi, L.; Cots-Rodríguez, P.; Izquierdo-Rico, M.J.; Avilés, M.; García-Vázquez, F.A. Sperm Proteome After Interaction with Reproductive Fluids in Porcine: From the Ejaculation to the Fertilization Site. Int. J. Mol. Sci. 2020, 21, 6060. [Google Scholar] [CrossRef] [PubMed]
  87. Ramilowski, J.A.; Goldberg, T.; Harshbarger, J.; Kloppmann, E.; Lizio, M.; Satagopam, V.P.; Itoh, M.; Kawaji, H.; Carninci, P.; Rost, B.; et al. A Draft Network of ligand–receptor-Mediated Multicellular Signalling in Human. Nat. Commun. 2015, 6, 7866. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Lee, K.-F.; Yao, Y.; Kwok, K.-L.; Xu, J.-S.; Yeung, W.S.B. Early Developing Embryos Affect the Gene Expression Patterns in the Mouse Oviduct. Biochem. Biophys. Res. Commun. 2002, 292, 564–570. [Google Scholar] [CrossRef]
  89. Almiñana, C.; Heath, P.R.; Wilkinson, S.; Sanchez-Osorio, J.; Cuello, C.; Parrilla, I.; Gil, M.A.; Vazquez, J.L.; Vazquez, J.M.; Roca, J.; et al. Early Developing Pig Embryos Mediate Their Own Environment in the Maternal Tract. PLoS ONE 2012, 7, e33625. [Google Scholar] [CrossRef] [Green Version]
  90. Schmaltzpanneau, B.; Cordova, A.; Dhorne-Pollet, S.; Hennequet-Antier, C.; Uzbekova, S.; Martinot, E.; Doret, S.; Martin, P.; Mermillod, P.; Locatelli, Y. Early Bovine Embryos Regulate Oviduct Epithelial Cell Gene Expression During In Vitro Co-Culture. Anim. Reprod. Sci. 2014, 149, 103–116. [Google Scholar] [CrossRef]
  91. Rizos, D.; Ramírez, M.; Pintado, B.; Lonergan, P.; Gutiérrez-Adán, A. Culture of Bovine Embryos in Intermediate Host Oviducts with Emphasis on the Isolated Mouse Oviduct. Theriogenology 2010, 73, 777–785. [Google Scholar] [CrossRef]
  92. Talukder, A.K.; Rashid, M.B.; Yousef, M.S.; Kusama, K.; Shimizu, T.; Shimada, M.; Suarez, S.S.; Imakawa, K.; Miyamoto, A. Oviduct Epithelium Induces Interferon-Tau in Bovine Day-4 Embryos, Which Generates an Anti-Inflammatory Response in Immune Cells. Sci. Rep. 2018, 8, 7850. [Google Scholar] [CrossRef]
  93. Dimou, E.; Nickel, W. Unconventional Mechanisms of Eukaryotic Protein Secretion. Curr. Biol. 2018, 28, R406–R410. [Google Scholar] [CrossRef] [Green Version]
  94. Raposo, G.; Stahl, P.D. Extracellular Vesicles: A New Communication Paradigm? Nat. Rev. Mol. Cell Biol. 2019, 20, 509–510. [Google Scholar] [CrossRef]
  95. Gwatkin, R. Effect of Viruses on Early Mammalian Development: III. Further Studies Concerning the Interaction of Mengo Encephalitis Virus with Mouse Ova. Fertil. Steril. 1966, 17, 411–420. [Google Scholar] [CrossRef]
  96. Kan, F.W.; Roux, E.; Bleau, G. Immunolocalization of Oviductin in Endocytic Compartments in the Blastomeres of Developing Embryos in the Golden Hamster1. Biol. Reprod. 1993, 48, 77–88. [Google Scholar] [CrossRef] [PubMed]
  97. Sherman, B.T.; Lempicki, R.A. Systematic and Integrative Analysis of Large Gene Lists Using DAVID Bioinformatics Resources. Nat. Protoc. 2009, 4, 44. [Google Scholar]
  98. Banliat, C.; Tsikis, G.; Labas, V.; Teixeira-Gomes, A.-P.; Com, E.; Lavigne, R.; Pineau, C.; Guyonnet, B.; Mermillod, P.; Saint-Dizier, M. Identification of 56 Proteins Involved in Embryo–Maternal Interactions in the Bovine Oviduct. Int. J. Mol. Sci. 2020, 21, 466. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Lee, Y.-L.; Lee, K.-F.; Xu, J.-S.; He, Q.-Y.; Chiu, J.-F.; Lee, W.M.; Luk, J.M.; Yeung, W.S.B. The Embryotrophic Activity of Oviductal Cell-Derived Complement C3b and iC3b, a Novel Function of Complement Protein in Reproduction. J. Biol. Chem. 2004, 279, 12763–12768. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Georgiou, A.S.; Snijders, A.P.L.; Sostaric, E.; Aflatoonian, R.; Vazquez, J.L.; Vazquez, J.M.; Roca, J.; Martinez, E.A.; Wright, P.C.; Fazeli, A. Modulation of The Oviductal Environment by Gametes. J. Proteome Res. 2007, 6, 4656–4666. [Google Scholar] [CrossRef] [PubMed]
  101. Klein, C. Novel Equine conceptus–endometrial Interactions on Day 16 of Pregnancy Based on RNA Sequencing. Reprod. Fertil. Dev. 2016, 28, 1712–1720. [Google Scholar] [CrossRef]
  102. Ward, P.P.; Mendoza-Meneses, M.; Mulac-Jericevic, B.; Cunningham, G.A.; Saucedo-Cardenas, O.; Teng, C.T.; Conneely, O.M. Restricted Spatiotemporal Expression of Lactoferrin During Murine Embryonic Development. Endocrinology 1999, 140, 1852–1860. [Google Scholar] [CrossRef]
  103. Østrup, O.; Olbricht, G.; Østrup, E.; Hyttel, P.; Collas, P.; Cabot, R. RNA Profiles of Porcine Embryos During Genome Activation Reveal Complex Metabolic Switch Sensitive to In Vitro Conditions. PLoS ONE 2013, 8, e61547. [Google Scholar] [CrossRef] [Green Version]
  104. Soleilhavoup, C.; Riou, C.; Tsikis, G.; Labas, V.; Harichaux, G.; Kohnke, P.L.; Reynaud, K.; De Graaf, S.P.; Gerard, N.; Druart, X. Proteomes of the Female Genital Tract During the Oestrous Cycle. Mol. Cell. Proteom. 2016, 15, 93–108. [Google Scholar] [CrossRef] [Green Version]
  105. Larson, R.C.; Ignotz, G.G.; Currie, W.B. Effect of Fibronectin on Early Embryo Development in Cows. Reproduction 1992, 96, 289–297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Lafrenie, R.M.; Yamada, K.M. Integrin-Dependent Signal Transduction. J. Cell. Biochem. 1996, 61, 543–553. [Google Scholar] [CrossRef]
  107. Gabler, C.; Chapman, D.A.; Killian, G.J. Expression and Presence of Osteopontin and Integrins in the Bovine Oviduct During the Oestrous Cycle. Reproduction 2003, 126, 721–729. [Google Scholar] [CrossRef] [PubMed]
  108. Newbold, R.R.; Teng, T.; Beckman, W.C., Jr.; Jefferson, W.N.; Hanson, R.B.; Miller, J.V.; Mclachlan, J.A. Fluctuations of Lactoferrin Protein and Messenger Ribonucleic Acid in the Reproductive Tract of the Mouse During the Estrous Cycle. Biol. Reprod. 1992, 47, 903–915. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Zumoffen, C.M.; Gil, R.; Caille, A.M.; Morente, C.; Munuce, M.J.; Ghersevich, S.A. A Protein Isolated from Human Oviductal Tissue in Vitro Secretion, Identified as Human Lactoferrin, Interacts with Spermatozoa and Oocytes and Modulates Gamete Interaction. Hum. Reprod. 2013, 28, 1297–1308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Ward, P.P.; Mendoza-Meneses, M.; Cunningham, G.A.; Conneely, O.M. Iron Status in Mice Carrying a Targeted Disruption of Lactoferrin. Mol. Cell. Biol. 2003, 23, 178–185. [Google Scholar] [CrossRef] [Green Version]
  111. Buhi, W.; Alvarez, I. Identification, Characterization and Localization of Three Proteins Expressed by the Porcine Oviduct. Theriogenology 2003, 60, 225–238. [Google Scholar] [CrossRef]
  112. Georgiou, A.; Gil, M.; Almiñana, C.; Cuello, C.; Vazquez, J.M.; Roca, J.; Martinez, E.A.; Fazeli, A. Effects of Complement Component 3 Derivatives on Pig Oocyte Maturation, Fertilization and Early Embryo Development In Vitro. Reprod. Domest. Anim. 2011, 46, 1017–1021. [Google Scholar] [CrossRef]
  113. Algarra, B.; Han, L.; Soriano-Úbeda, C.; Avilés, M.; Coy, P.; Jovine, L.; Jiménez-Movilla, M. The C-Terminal Region of OVGP1 Remodels the Zona Pellucida and Modifies Fertility Parameters. Sci. Rep. 2016, 6, 32556. [Google Scholar] [CrossRef] [Green Version]
  114. Oliphant, G.; Bowling, A.; Eng, L.A.; Keen, S.; Randall, P.A. The Permeability of Rabbit Oviduct to Proteins Present in the Serum. Biol. Reprod. 1978, 18, 516–520. [Google Scholar] [CrossRef] [Green Version]
  115. Rodríguez-Alonso, B.; Maillo, V.; Acuña, O.S.; López-Úbeda, R.; Torrecillas, A.; Simintiras, C.A.; Sturmey, R.; Avilés, M.; Lonergan, P.; Rizos, D. Spatial and Pregnancy-Related Changes in the Protein, Amino Acid, and Carbohydrate Composition of Bovine Oviduct Fluid. Int. J. Mol. Sci. 2020, 21, 1681. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Oliphant, G.; Reynolds, A.B.; Smith, P.F.; Ross, P.R.; Marta, J.S. Immunocytochemical Localization and Determination of Hormone-Induced Synthesis of the Sulfated Oviductal Glycoproteins. Biol. Reprod. 1984, 31, 165–174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Brown, C.R.; Cheng, W.K.T. Changes in Composition of the Porcine Zona-Pellucida During Development of the Oocyte to the 2-Cell to 4-Cell Embryo. J. Embryol. Exp. Morphol. 1986, 92, 183–191. [Google Scholar]
  118. Oikawa, T.; Sendai, Y.; Kurata, S.-I.; Yanagimachi, R. A Glycoprotein of Oviductal Origin Alters Biochemical Properties of the Zona Pellucida of Hamster Egg. Gamete Res. 1988, 19, 113–122. [Google Scholar] [CrossRef] [PubMed]
  119. Buhi, W.C. Characterization and Biological Roles of Oviduct-Specific, Oestrogen-Dependent Glycoprotein. Reproduction 2002, 123, 355–362. [Google Scholar] [CrossRef] [PubMed]
  120. Araki, Y.; Nohara, M.; Yoshida-Komiya, H.; Kuramochi, T.; Ito, M.; Hoshi, H.; Shinkai, Y.; Sendai, Y. Effect of a Null Mutation of the Oviduct-Specific Glycoprotein Gene on Mouse Fertilization. Biochem. J. 2003, 374, 551–557. [Google Scholar] [CrossRef] [PubMed]
  121. Murray, M.K.; DeSouza, M.M. Messenger RNA Encoding an Estrogen-Dependent Oviduct Secretory Protein in the Sheep Is Localized in the Apical Tips and Basal Compartments of Fimbria and Ampulla Epithelial Cells Implying Translation at Unique Cytoplasmic Foci. Mol. Reprod. Dev. 1995, 42, 268–283. [Google Scholar] [CrossRef]
  122. Kan, F.W.; St-Jacques, S.; Bleau, G. Immunoelectron Microscopic Localization of an Oviductal Antigen in Hamster Zona Pellucida by Use of a Monoclonal Antibody. J. Histochem. Cytochem. 1988, 36, 1441–1447. [Google Scholar] [CrossRef] [Green Version]
  123. Abé, H.; Oikawa, T. Immunocytochemical Localization of an Oviductal Zona Pellucida Glycoprotein in the Oviductal Epithelium of the Golden Hamster. Anat. Rec. Adv. Integr. Anat. Evol. Biol. 1991, 229, 305–314. [Google Scholar]
  124. McBride, D.S.; Boisvert, C.; Bleau, G.; Kan, F.W. Detection of Nascent and/Or Mature Forms of Oviductin in the Female Reproductive Tract and Post-Ovulatory Oocytes by Use of a Polyclonal Antibody Against Recombinant Hamster Oviductin. J. Histochem. Cytochem. 2004, 52, 1001–1009. [Google Scholar] [CrossRef] [Green Version]
  125. Boice, M.L.; Geisert, R.D.; Blair, R.M.; Verhage, H.G. Identification and Characterization of Bovine Oviductal Glycoproteins Synthesized at Estrus. Biol. Reprod. 1990, 43, 457–465. [Google Scholar] [CrossRef] [PubMed]
  126. Abé, H.; Numazawa, C.; Abe, M.; Onodera, M.; Katsumi, A. Immunocytochemical Localization of Oviduct-Specific Glycoproteins in the Oviductal Epithelium from Cows at Follicular and Luteal Phases. Cell Tissue Res. 1993, 274, 41–47. [Google Scholar] [CrossRef] [PubMed]
  127. Buhi, W.C.; O’Brien, B.; Alvarez, I.M.; Erdos, G.; Dubois, D. Immunogold Localization of Porcine Oviductal Secretory Proteins Within the Zona Pellucida, Perivitelline Space, and Plasma Membrane of Oviductal and Uterine Oocytes and Early Embryos1. Biol. Reprod. 1993, 48, 1274–1283. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Verhage, H.G.; Mavrogianis, P.A.; Boice, M.L.; Li, W.; Fazleabas, A.T. Oviductal Epithelium of the Baboon: Hormonal Control and the Immuno-Gold Localization of Oviduct-Specific Glycoproteins. Am. J. Anat. 1990, 187, 81–90. [Google Scholar] [CrossRef] [PubMed]
  129. Rapisarda, J.J.; Mavrogianis, P.A.; O’Day-Bowman, M.B.; Fazleabas, A.T.; Verhage, H.G. Immunological Characterization and Immunocytochemical Localization of an Oviduct-Specific Glycoprotein in the Human. J. Clin. Endocrinol. Metab. 1993, 76, 1483–1488. [Google Scholar] [PubMed]
  130. Kapur, R.P.; Johnson, L.V. Ultrastructural Evidence That Specialized Regions of the Murine Oviduct Contribute a Glycoprotein to the Extracellular Matrix of Mouse Oocytes. Anat. Rec. Adv. Integr. Anat. Evol. Biol. 1988, 221, 720–729. [Google Scholar] [CrossRef]
  131. Arias, E.B.; Verhage, H.G.; Jaffe, R.C. Complementary Deoxyribonucleic Acid Cloning and Molecular Characterization of an Estrogen-Dependent Human Oviductal Glycoprotein1. Biol. Reprod. 1994, 51, 685–694. [Google Scholar] [CrossRef] [Green Version]
  132. Tian, X.; Pascal, G.; Fouchécourt, S.; Pontarotti, P.; Monget, P. Gene Birth, Death, and Divergence: The Different Scenarios of Reproduction-Related Gene Evolution1. Biol. Reprod. 2009, 80, 616–621. [Google Scholar] [CrossRef] [Green Version]
  133. Moros-Nicolás, C.; Fouchécourt, S.; Goudet, G.; Monget, P. Genes Encoding Mammalian oviductal proteins involved in fertilization are subjected to gene death and positive selection. J. Mol. Evol. 2018, 86, 655–667. [Google Scholar] [CrossRef]
  134. Fazleabas, A.T.; Verhage, H.G. The Detection of Oviduct-Specific Proteins in the Baboon (Papio anubis). Biol. Reprod. 1986, 35, 455–462. [Google Scholar] [CrossRef]
  135. Verhage, H.G.; Fazleabas, A.T.; Donnelly, K. The in vitro synthesis and release of proteins by the human oviduct. Endocrinology 1988, 122, 1639–1645. [Google Scholar] [CrossRef] [PubMed]
  136. Donnelly, K.M.; Fazleabas, A.T.; Verhage, H.G.; Mavrogianis, P.A.; Jaffe, R.C. Cloning of a Recombinant Complementary DNA to a Baboon (Papio Anubis) Estradiol-Dependent Oviduct-Specific Glycoprotein. Mol. Endocrinol. 1991, 5, 356–364. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. O’Day-Bowman, M.B.; Mavrogianis, P.A.; Reuter, L.M.; Johnson, D.E.; Fazleabas, A.T.; Verhage, H.G. Association of oviduct-specific glycoproteins with human and baboon (Papio anubis) ovarian oocytes and enhancement of human sperm binding to human hemizonae following in vitro incubation. Biol. Reprod. 1996, 54, 60–69. [Google Scholar] [CrossRef] [PubMed]
  138. Verhage, H.G.; Fazleabas, A.T.; Mavrogianis, P.A.; O’Day-Bowman, M.B.; Donnelly, K.M.; Jaffe, R.C. The baboon oviduct: Characteristics of an oestradiol-dependent oviduct-specific glycoprotein. Hum. Reprod. Update 1997, 3, 541–552. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Hachen, A.; Jewgenow, K.; Braun, B.C. Sequence analysis of feline oviductin and its expression during the estrous cycle in the domestic cat (Felis catus). Theriogenology 2012, 77, 539–549. [Google Scholar] [CrossRef] [PubMed]
  140. Hribal, R.; Hachen, A.; Zahmel, J.; Gonzalez, L.F.; Jewgenow, K.; Braun, B.C. The effect of recombinant feline oviductin used in the cat IVF system. Reprod. Domest. Anim. 2014, 49, 24–25. [Google Scholar]
  141. Hribal, R.; Hachen, A.; Jewgenow, K.; Zahmel, J.; Fernandez-Gonzalez, L.; Braun, B.C. The influence of recombinant feline oviductin on different aspects of domestic cat (Felis catus) IVF and embryo quality. Theriogenology 2014, 82, 742–749. [Google Scholar] [CrossRef]
  142. Kim, R.N.; Kim, D.W.; Choi, S.H.; Chae, S.H.; Nam, S.H.; Kim, A.; Kang, A.; Park, K.H.; Lee, Y.S.; Hirai, M.; et al. Major chimpanzee-specific structural changes in sperm development-associated genes. Funct. Integr. Genom. 2011, 11, 507–517. [Google Scholar] [CrossRef]
  143. Malayer, J.; Hansen, P.; Buhi, W. Secretion of proteins by cultured bovine oviducts collected from estrus through early diestrus. J. Exp. Zool. 1988, 248, 345–353. [Google Scholar] [CrossRef]
  144. King, R.S.; Anderson, S.H.; Killian, G.J. Effect of bovine oviductal estrus-associated protein on the ability of sperm to capacitate and fertilize oocytes. J. Androl. 1994, 15, 468–478. [Google Scholar]
  145. Abe, H.; Sendai, Y.; Satoh, T.; Hoshi, H. Bovine oviduct-specific glycoprotein: A potent factor for maintenance of viability and motility of bovine spermatozoa in vitro. Mol. Reprod. Dev. 1995, 42, 226–232. [Google Scholar] [CrossRef] [PubMed]
  146. Martus, N.S.; Verhage, H.G.; Mavrogianis, P.A.; Thibodeaux, J.K. Enhancement of bovine oocyte fertilization in vitro with a bovine oviductal specific glycoprotein. J. Reprod. Fertil. 1998, 113, 323–329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Saint-Dizier, M.; Marnier, C.; Tahir, M.Z.; Grimard, B.; Thoumire, S.; Chastant-Maillard, S.; Reynaud, K. OVGP1 Is Expressed in the Canine Oviduct at the Time and Place of Oocyte Maturation and Fertilization. Mol. Reprod. Dev. 2014, 81, 972–982. [Google Scholar] [CrossRef] [PubMed]
  148. Abe, H.; Onodera, M.; Sugawara, S. Immunological detection and characterization of an estrus-associated antigen in the goat oviduct. J. Exp. Zool. 1995, 272, 134–141. [Google Scholar] [CrossRef]
  149. Pradeep, M.A.; Jagadeesh, J.; De, A.K.; Kaushik, J.K.; Malakar, D.; Kumar, S.; Dang, A.K.; Das, S.K.; Mohanty, A.K. Purification, sequence characterization and effect of goat oviduct-specific glycoprotein on in vitro embryo development. Theriogenology 2011, 75, 1005–1015. [Google Scholar] [CrossRef]
  150. Léveillé, M.-C.; Roberts, K.D.; Chevalier, S.; Chapdelaine, A.; Bleau, G. Uptake of an oviductal antigen by the hamster zona pellucida. Biol. Reprod. 1987, 36, 227–238. [Google Scholar] [CrossRef] [Green Version]
  151. Robitaille, G.; St-Jacques, S.; Potier, M.; Bleau, G. Characterization of an oviductal glycoprotein associated with the ovulated hamster oocyte. Biol. Reprod. 1988, 38, 687–694. [Google Scholar] [CrossRef] [Green Version]
  152. Kan, F.W.; Roux, E.; St.-Jacquesz, S.; Bleau, G. Demonstration by lectin-gold cytochemistry of transfer of glycoconjugates of oviductal origin to the zona pellucida of oocytes after ovulation in hamsters. Anat. Rec. 1990, 226, 37–47. [Google Scholar] [CrossRef]
  153. Suzuki, K.; Sendai, Y.; Onuma, T.; Hoshi, H.; Hiroi, M.; Araki, Y. Molecular characterization of a hamster oviduct-specific glycoprotein. Biol. Reprod. 1995, 53, 345–354. [Google Scholar] [CrossRef] [Green Version]
  154. Schmidt, A.; Mavrogianis, P.A.; O’dayBowman, M.B.; Jaffe, R.C.; Verhage, H.G. Characterization of antibodies generated against a conserved portion of oviductal glycoprotein (OGP) and endogenous hamster OGP and their ability to decrease sperm binding to the zona pellucida in vitro. Am. J. Reprod. Immunol. 1997, 38, 377–383. [Google Scholar] [CrossRef]
  155. Abe, H.; Satoh, T.; Hoshi, H. Primary modulation by oestradiol of the production of an oviduct-specific glycoprotein by the epithelial cells in the oviduct of newborn golden hamsters. J. Reprod. Fertil. 1998, 112, 157–163. [Google Scholar] [CrossRef] [PubMed]
  156. O’Day-Bowman, M.B.; Mavrogianis, P.A.; Minshall, R.D.; Verhage, H.G. In vivo versus in vitro oviductal glycoprotein (OGP) association with the zona pellucida (ZP) in the hamster and baboon. Mol. Reprod. Dev. 2002, 62, 248–256. [Google Scholar] [CrossRef] [PubMed]
  157. O’day-Bowman, M.B.; Mavrogianis, P.A.; Fazleabas, A.T.; Verhage, H.G. A human oviduct-specific glycoprotein: Synthesis, secretion, and localization during the menstrual cycle. Microsc. Res. Tech. 1995, 32, 57–69. [Google Scholar] [CrossRef] [PubMed]
  158. Briton-Jones, C.; Lok, I.H.; Yuen, P.M.; Chiu, T.T.Y.; Cheung, L.P.; Haines, C. Regulation of human oviductin mRNA expression in vivo. Fertil. Steril. 2001, 75, 942–946. [Google Scholar] [CrossRef]
  159. Briton-Jones, C.; Lok, I.H.; Cheung, C.K.; Chiu, T.T.Y.; Cheung, L.P.; Haines, C. Estradiol regulation of oviductin/oviduct-specific glycoprotein messenger ribonucleic acid expression in human oviduct mucosal cells in vitro. Fertil. Steril. 2004, 81, 749–756. [Google Scholar] [CrossRef]
  160. Verhage, H.G.; Mavrogianis, P.A.; Boomsma, R.A.; Schmidt, A.; Brenner, R.M.; Slayden, O.V.; Jaffe, R.C. Immunologic and molecular characterization of an estrogen-dependent glycoprotein in the rhesus (Macaca mulatta) oviduct. Biol. Reprod. 1997, 57, 525–531. [Google Scholar] [CrossRef] [Green Version]
  161. Yan, G.M.; Zhang, G.J.; Fang, X.D.; Zhang, Y.F.; Li, C.; Ling, F.; Cooper, D.N.; Li, Q.Y.; Li, Y.; van Gool, A.J.; et al. Genome sequencing and comparison of two nonhuman primate animal models, the cynomolgus and Chinese rhesus macaques. Nat. Biotechnol. 2011, 29, 1019–U1089. [Google Scholar] [CrossRef] [Green Version]
  162. Kapur, R.P.; Johnson, L.V. An oviductal fluid glycoprotein associated with ovulated mouse ova and early embryos. Dev. Biol. 1985, 112, 89–93. [Google Scholar] [CrossRef]
  163. Sendai, Y.; Komiya, H.; Suzuki, K.; Onuma, T.; Kikuchi, M.; Hoshi, H.; Araki, Y. Molecular cloning and characterization of a mouse oviduct-specific glycoprotein. Biol. Reprod. 1995, 53, 285–294. [Google Scholar] [CrossRef] [Green Version]
  164. Buhi, W.; Vallet, J.; Bazer, F. De novo synthesis and release of polypeptides from cyclic and early pregnant porcine oviductal tissue in explant culture. J. Exp. Zool. 1989, 252, 79–88. [Google Scholar] [CrossRef]
  165. Buhi, W.; Alvarez, I.; Sudhipong, V.; Dones-Smith, M. Identification and characterization of de novo-synthesized porcine oviductal secretory proteins. Biol. Reprod. 1990, 43, 929–938. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Buhi, W.C.; Alvarez, I.M.; Choi, I.; Cleaver, B.D.; Simmen, F.A. Molecular cloning and characterization of an estrogen-dependent porcine oviductal secretory glycoprotein. Biol. Reprod. 1996, 55, 1305–1314. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Kouba, A.J.; Abeydeera, L.R.; Alvarez, I.M.; Day, B.N.; Buhi, W.C. Effects of the porcine oviduct-specific glycoprotein on fertilization, polyspermy, and embryonic development in vitro. Biol. Reprod. 2000, 63, 242–250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Merchan, M.; Rendon, M.; Folch, J.M. Assignment of the oviductal glycoprotein 1 gene (OVGP1) to porcine chromosome 4q22 -> q23 by radiation hybrid panel mapping. Cytogenet. Genome Res. 2006, 114, 93C. [Google Scholar] [CrossRef] [PubMed]
  169. Oliphant, G.; Ross, P.R. Demonstration of production and isolation of three sulfated glycoproteins from the rabbit oviduct. Biol. Reprod. 1982, 26, 537–544. [Google Scholar] [CrossRef] [Green Version]
  170. Merchan, M.; Peiro, R.; Argente, M.J.; Santacreu, M.A.; Garcia, M.L.; Blasco, A.; Folch, J.M. Analysis of the oviductal glycoprotein 1 polymorphisms and their effects on components of litter size in rabbits. Anim. Genet. 2009, 40, 756–758. [Google Scholar] [CrossRef]
  171. Garcia, M.L.; Peiro, R.; Argente, M.J.; Merchan, M.; Folch, J.M.; Blasco, A.; Santacreu, M.A. Investigation of the oviductal glycoprotein 1 (OVGP1) gene associated with embryo survival and development in the rabbit. J. Anim. Sci. 2010, 88, 1597–1602. [Google Scholar] [CrossRef] [Green Version]
  172. Sutton, R.; Nancarrow, C.; Wallace, A.; Rigby, N. Identification of an oestrus-associated glycoprotein in oviducal fluid of the sheep. J. Reprod. Fertil. 1984, 72, 415–422. [Google Scholar] [CrossRef] [Green Version]
  173. Gandolfi, F.; Brevini, T.; Richardson, L.; Brown, C.; Moor, R. Characterization of proteins secreted by sheep oviduct epithelial cells and their function in embryonic development. Development 1989, 106, 303–312. [Google Scholar]
  174. Gandolfi, F.; Modina, S.; Brevini, T.; Galli, C.; Moor, R.; Lauria, A. Oviduct ampullary epithelium contributes a glycoprotein to the zona pellucida, perivitelline space and blastomeres membrane of sheep embryos. Eur. J. Basic Appl. Histochem. 1991, 35, 383–392. [Google Scholar]
  175. DeSouza, M.M.; Murray, M.K. An estrogen-dependent secretory protein, which shares identity with chitinases, is expressed in a temporally and regionally specific manner in the sheep oviduct at the time of fertilization and embryo development. Endocrinology 1995, 136, 2485–2496. [Google Scholar] [CrossRef] [PubMed]
  176. Hill, J.L.; Wade, M.G.; Nancarrow, C.D.; Kelleher, D.L.; Boland, M.P. Influence of ovine oviducal amino acid concentrations and an ovine oestrus-associated glycoprotein on development and viability of bovine embryos. Mol. Reprod. Dev. 1997, 47, 164–169. [Google Scholar] [CrossRef]
  177. Malette, B.; Paquette, Y.; Merlen, Y.; Bleau, G. Oviductins possess chitinase-and mucin-like domains: A lead in the search for the biological function of these oviduct-specific ZP-associating glycoproteins. Mol. Reprod. Dev. 1995, 41, 384–397. [Google Scholar] [CrossRef]
  178. Laheri, S.; Ashary, N.; Bhatt, P.; Modi, D. Oviductal glycoprotein 1 (OVGP1) is expressed by endometrial epithelium that regulates receptivity and trophoblast adhesion. J. Assist. Reprod. Genet. 2018, 35, 1419–1429. [Google Scholar] [CrossRef] [PubMed]
  179. Buhi, W.; Bazer, F.; Alvarez, I.; Mirandot, M. In vitro synthesis of oviductal proteins associated with estrus and 17β-estradiol-treated ovariectomized ewes. Endocrinology 1991, 128, 3086–3095. [Google Scholar] [CrossRef]
  180. Sendai, Y.; Abe, H.; Kikuchi, M.L.; Satoh, T.; Hoshi, H. Purification and molecular cloning of bovine oviduct-specific glycoprotein. Biol. Reprod. 1994, 50, 927–934. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  181. Nancarrow, C.; Hill, J. Oviduct proteins in fertilization and early embryo development. J. Reprod. Fertil. Suppl. Only 1995, 49, 3–14. [Google Scholar] [CrossRef]
  182. Jaffe, R.C.; Arias, E.B.; OdayBowman, M.B.; Donnelly, K.M.; Mavrogianis, P.A.; Verhage, H.G. Regional distribution and hormonal control of estrogen-dependent oviduct-specific glycoprotein messenger ribonucleic acid in the baboon (Papio anubis). Biol. Reprod. 1996, 55, 421–426. [Google Scholar] [CrossRef] [Green Version]
  183. Buhi, W.C.; Alvarez, I.M.; Kouba, A.J. Secreted proteins of the oviduct. Cells Tissues Organs 2000, 166, 165–179. [Google Scholar] [CrossRef]
  184. Lok, I.H.; Briton-Jones, C.M.; Yuen, P.M.; Haines, C.J. Variable expression of oviductin mRNA at different stages of human reproductive cycle. J. Assist. Reprod. Genet. 2002, 19, 569–576. [Google Scholar] [CrossRef]
  185. Laheri, S.; Modi, D.; Bhatt, P. Extra-oviductal expression of oviductal glycoprotein 1 in mouse: Detection in testis, epididymis and ovary. J. Biosci. 2017, 42, 69–80. [Google Scholar] [CrossRef] [PubMed]
  186. Paquette, Y.; Merlen, Y.; Malette, B.; Bleau, G. Allelic polymorphism in the hamster oviductin gene is due to a variable number of mucin-like tandem repeats. Mol. Reprod. Dev. 1995, 42, 388–396. [Google Scholar] [CrossRef] [PubMed]
  187. Merchan, M.; Peiro, R.; Santacreu, M.A.; Francino, O.; Folch, J.M. Rabbit oviductal glycoprotein 1 gene: Genomic organization polymorphism analysis and mRNA expression. Mol. Reprod. Dev. 2007, 74, 687–693. [Google Scholar] [CrossRef] [PubMed]
  188. Komiya, H.; Onuma, T.; Hiroi, M.; Araki, Y. In situ localization of messenger ribonucleic acid for an oviduct-specific glycoprotein during various hormonal conditions in the golden hamster. Biol. Reprod. 1996, 55, 1107–1118. [Google Scholar] [CrossRef]
  189. Roux, E.; Kan, F.W. Stage-specific immunolabeling for oviductin in the secretory granules of the oviductal epithelium of the golden hamster during the estrous cycle. Anat. Rec. 1995, 241, 369–376. [Google Scholar] [CrossRef]
  190. Buhi, W.; Alvarez, I.; Kouba, A. Oviductal regulation of fertilization and early embryonic development. J. Reprod. Fertil. Suppl. 1997, 52, 285. [Google Scholar]
  191. Murray, M.K. An estrogen-dependent glycoprotein is synthesized and released from the oviduct in a temporal-and region-specific manner during early pregnancy in the ewe. Biol. Reprod. 1993, 48, 446–453. [Google Scholar] [CrossRef] [Green Version]
  192. Hyde, B.; Black, D. Synthesis and secretion of sulphated glycoproteins by rabbit oviduct explants in vitro. Reproduction 1986, 78, 83–91. [Google Scholar] [CrossRef] [Green Version]
  193. Erickson-Lawrence, M.F.; Turner, T.T.; Thomas, T.S.; Oliphant, G. Effect of steroid hormones on sulfated oviductal glycoprotein secretion by oviductal explants in vitro. Biol. Reprod. 1989, 40, 1311–1319. [Google Scholar] [CrossRef]
  194. Gonella-Diaza, A.M.; da Silva Andrade, S.C.; Sponchiado, M.; Pugliesi, G.; Mesquita, F.S.; Van Hoeck, V.; de Francisco Strefezzi, R.; Gasparin, G.R.; Coutinho, L.L.; Binelli, M. Size of the ovulatory follicle dictates spatial differences in the oviductal transcriptome in cattle. PLoS ONE 2015, 10, e0145321. [Google Scholar] [CrossRef] [Green Version]
  195. Kadam, K.M.; D’Souza, S.J.; Bandivdekar, A.H.; Natraj, U. Identification and characterization of oviductal glycoprotein-binding protein partner on gametes: Epitopic similarity to non-muscle myosin IIA, MYH 9. Mol. Hum. Reprod. 2006, 12, 275–282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. DeSouza, M.M.; Murray, M.K. An estrogen-dependent sheep oviductal glycoprotein has glycan linkages typical of sialomucins and does not contain chitinase activity. Biol. Reprod. 1995, 53, 1517–1526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Choudhary, S.; Janjanam, J.; Kumar, S.; Kaushik, J.K.; Mohanty, A.K. Structural and functional characterization of buffalo oviduct-specific glycoprotein (OVGP1) expressed during estrous cycle. Biosci. Rep. 2019, 39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  198. Kadam, K.M.; D’Souza, S.J.; Natraj, U. Identification of cellular isoform of oviduct-specific glycoprotein: Role in oviduct tissue remodeling? Cell Tissue Res. 2007, 330, 545–556. [Google Scholar] [CrossRef]
  199. Wegner, C.C.; Killian, G.J. In vitro and in vivo association of an oviduct estrus-associated protein with bovine zona pellucida. Mol. Reprod. Dev. 1991, 29, 77–84. [Google Scholar] [CrossRef]
  200. Coy, P.; Canovas, S.; Mondejar, I.; Saavedra, M.D.; Romar, R.; Grullon, L.; Matas, C.; Aviles, M. Oviduct-specific glycoprotein and heparin modulate sperm-zona pellucida interaction during fertilization and contribute to the control of polyspermy. Proc. Natl. Acad. Sci. USA 2008, 105, 15809–15814. [Google Scholar] [CrossRef] [Green Version]
  201. Algarra, B.; Maillo, V.; Avilés, M.; Gutiérrez-Adán, A.; Rizos, D.; Jiménez-Movilla, M. Effects of recombinant OVGP1 protein on in vitro bovine embryo development. J. Reprod Dev. 2018, 64, 433–443. [Google Scholar] [CrossRef] [Green Version]
  202. Kapur, R.P.; Johnson, L.V. Selective sequestration of an oviductal fluid glycoprotein in the perivitelline space of mouse oocytes and embryos. J. Exp. Zool. 1986, 238, 249–260. [Google Scholar] [CrossRef]
  203. Lyng, R.; Shur, B.D. Mouse oviduct-specific glycoprotein is an egg-associated ZP3-independent sperm-adhesion ligand. J. Cell Sci. 2009, 122, 3894–3906. [Google Scholar] [CrossRef] [Green Version]
  204. Boice, M.L.; McCarthy, T.J.; Mavrogianis, P.A.; Fazlebas, A.T.; Verhage, H.G. Localization of Oviductal Glycoproteins within the Zona Pellucida and Perivitelline Space of Ovulated Ova and Early Embryos in Baboons (Paplo Anubis). Biol. Reprod. 1990, 43, 340–346. [Google Scholar] [CrossRef] [Green Version]
  205. Reuter, L.M.; O’Day-Bowman, M.B.; Mavrogianis, P.A.; Fazleabas, A.T.; Verhage, H.G. In vitro incubation of golden (Syrian) hamster ovarian oocytes and human sperm with a human oviduct specific glycoprotein. Mol. Reprod. Dev. 1994, 38, 160–169. [Google Scholar] [CrossRef] [PubMed]
  206. Malette, B.; Bleau, G. Biochemical characterization of hamster oviductin as a sulphated zona pellucida-binding glycoprotein. Biochem. J. 1993, 295, 437–445. [Google Scholar] [CrossRef] [Green Version]
  207. Yang, X.J.; Zhao, Y.W.; Yang, X.L.; Kan, F.W.K. Recombinant Hamster Oviductin Is Biologically Active and Exerts Positive Effects on Sperm Functions and Sperm-Oocyte Binding. PLoS ONE 2015, 10, e0123003. [Google Scholar] [CrossRef] [Green Version]
  208. Lloyd, R.E.; Romar, R.; Matas, C.; Gutierrez-Adan, A.; Holt, W.V.; Coy, P. Effects of oviductal fluid on the development, quality, and gene expression of porcine blastocysts produced in vitro. Reproduction 2009, 137, 679–687. [Google Scholar] [CrossRef] [Green Version]
  209. Goncalves, R.F.; Staros, A.L.; Killian, G.J. Oviductal Fluid Proteins Associated with the Bovine Zona Pellucida and the Effect on In Vitro Sperm-Egg Binding, Fertilization and Embryo Development. Reprod. Domest. Anim. 2008, 43, 720–729. [Google Scholar] [CrossRef] [PubMed]
  210. Broermann, D.; Xie, S.; Nephew, K.; Pope, W. Effects of the oviduct and wheat germ agglutinin on enzymatic digestion of porcine zona pellucidae. J. Anim. Sci. 1989, 67, 1324–1329. [Google Scholar] [CrossRef] [PubMed]
  211. Davachi, N.D.; Shahneh, A.Z.; Kohram, H.; Zhandi, M.; Shamsi, H.; Hajiyavand, A.M.; Saadat, M. Differential influence of ampullary and isthmic derived epithelial cells on zona pellucida hardening and in vitro fertilization in ovine. Reprod. Biol. 2016, 16, 61–69. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Kim, N.H.; Funahashi, H.; Abeydeera, L.R.; Moon, S.J.; Prather, R.S.; Day, B.N. Effects of oviductal fluid on sperm penetration and cortical granule exocytosis during fertilization of pig oocytes in vitro. J. Reprod. Fertil. 1996, 107, 79–86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Kolbe, T.; Holtz, W. Differences in proteinase digestibility of the zona pellucida of in vivo and in vitro derived porcine oocytes and embryos. Theriogenology 2005, 63, 1695–1705. [Google Scholar] [CrossRef]
  214. Mondejar, I.; Aviles, M.; Coy, P. The human is an exception to the evolutionarily-conserved phenomenon of pre-fertilization zona pellucida resistance to proteolysis induced by oviductal fluid. Hum. Reprod. 2013, 28, 718–728. [Google Scholar] [CrossRef] [Green Version]
  215. Mondejar, I.; Martinez-Martinez, I.; Aviles, M.; Coy, P. Identification of Potential Oviductal Factors Responsible for Zona Pellucida Hardening and Monospermy During Fertilization in Mammals. Biol. Reprod. 2013, 89, 67. [Google Scholar] [CrossRef] [PubMed]
  216. Wang, W.H.; Abeydeera, L.R.; Prather, R.S.; Day, B.N. Morphologic comparison of ovulated and in vitro-matured porcine oocytes, with particular reference to polyspermy after in vitro fertilization. Mol. Reprod. Dev. 1998, 49, 308–316. [Google Scholar] [CrossRef]
  217. Wright Jr, R.W.; Cupps, P.T.; Gaskins, C.T.; Hillers, J.K. Comparative solubility properties of the zona pellucidae of unfertilized murine, ovine and bovine ova. J. Anim. Sci. 1977, 44, 850–853. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  218. McCauley, T.C.; Buhi, W.C.; Wu, G.M.; Mao, J.; Caamano, J.N.; Didion, B.A.; Day, B.N. Oviduct-specific glycoprotein modulates sperm-zona binding and improves efficiency of porcine fertilization in vitro. Biol. Reprod. 2003, 69, 828–834. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  219. Schmidt, A.; Mavrogianis, P.A.; OdayBowman, M.B.; Verhage, H.G. Species-specific effect of oviductal glycoproteins on hamster sperm binding to hamster oocytes. Mol. Reprod. Dev. 1997, 46, 201–207. [Google Scholar] [CrossRef]
  220. Boatman, D.E.; Magnoni, G.E. Identification of a sperm penetration factor in the oviduct of the golden hamster. Biol. Reprod. 1995, 52, 199–207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  221. Yong, P.; Gu, Z.; Luo, J.P.; Wang, J.R.; Tso, J.K. Antibodies against the C-terminal peptide of rabbit oviductin inhibit mouse early embryo development to pass 2-cell stage. Cell Res. 2002, 12, 69–78. [Google Scholar] [CrossRef] [Green Version]
  222. Hill, J.; Walker, S.; Brown, G.; Nancarrow, C. The effects of an estrus-associated oviductal glycoprotein on the in vitro fertilization and development of ovine oocytes matured in vitro. Theriogenology 1996, 46, 1379–1388. [Google Scholar] [CrossRef]
  223. Hill, J.; Walker, S.; Brown, G.; Nancarrow, C. The effects of an ovine oviductal estrus-associated glycoprotein on early embryo development. Theriogenology 1996, 46, 1367–1377. [Google Scholar] [CrossRef]
Figure 1. DEGs between oviducts containing embryos and oviducts on the equivalent day post-ovulation across in bovine, equine and porcine species. (a) Total DEGs. (b) Up-regulated genes in the presence of embryos. (c) Down-regulated genes in the presence of embryos.
Figure 1. DEGs between oviducts containing embryos and oviducts on the equivalent day post-ovulation across in bovine, equine and porcine species. (a) Total DEGs. (b) Up-regulated genes in the presence of embryos. (c) Down-regulated genes in the presence of embryos.
Biomolecules 10 01690 g001
Figure 2. Analysis of the DEG in the oviduct of non-pregnant and pregnant animals related with secreted and/or exosomal proteins. (a) Differentially expressed genes coding for exosomal and/or secreted proteins in the ipsilateral oviduct of pregnant vs non-pregnant cows. Data taken from Maillo et al., 2015 [74]. When more than 20 genes were present in an area of the Venn diagram, gene names were selected according the highest fold change. (b) Differentially expressed genes coding for exosomal and/or secreted proteins in the ipsilateral oviduct of pregnant vs. non-pregnant mares. Data taken from Smits et al., 2016 [75]. (c) Differentially expressed genes coding for exosomal and/or secreted proteins in the pregnant vs. nonpregnant oviduct of sows. Data taken from Martyniak et al., 2018 [76]. When more than 20 genes were present in an area of the Venn diagram, gene names were selected according the highest fold change.
Figure 2. Analysis of the DEG in the oviduct of non-pregnant and pregnant animals related with secreted and/or exosomal proteins. (a) Differentially expressed genes coding for exosomal and/or secreted proteins in the ipsilateral oviduct of pregnant vs non-pregnant cows. Data taken from Maillo et al., 2015 [74]. When more than 20 genes were present in an area of the Venn diagram, gene names were selected according the highest fold change. (b) Differentially expressed genes coding for exosomal and/or secreted proteins in the ipsilateral oviduct of pregnant vs. non-pregnant mares. Data taken from Smits et al., 2016 [75]. (c) Differentially expressed genes coding for exosomal and/or secreted proteins in the pregnant vs. nonpregnant oviduct of sows. Data taken from Martyniak et al., 2018 [76]. When more than 20 genes were present in an area of the Venn diagram, gene names were selected according the highest fold change.
Biomolecules 10 01690 g002
Table 1. Chronology of embryo development in different species.
Table 1. Chronology of embryo development in different species.
HUMANMOUSEPIGCOWSHEEPMARECATBITCHGUINEA PIGRABBIT
2 cells1
(in vitro) [28]
1
[29]
14–16 h
[30]
24–30 h
[31]
1.5

[32]
20 h
[33]
2.5
[34]
4.5–5.5
[35]
2
[36]
16–17 h
[37]
8 cells3
(in vitro) [38]
2.5
[29]
2
[30]
3.5
[39]
2
[32]
3
[40]
3
[34]
4.5–5.5
[35]
4
[36]
29–30 h
[37]
Morula4
(in vitro) [28]
3
[29]
3.5
[30]
5–6
[31]
4
[32]
4
[40]
5
[34]
10
[41]
5
[36]
2.5
[42,43]
Blastocyst5
(in vitro) [28]
3.5
[29]
5
[30]
7
[31]
6
[44]
6
[40]
6–9
[34]
12–13
[41]
6
[36]
3
[43]
Hatching6–7
(in vitro) [45]
4.5
[46]
6
[30]
8
[31]
8
[44]
7–8
[40]
12
[47]
19–20
[48]
6
[49]
4
(in vitro) [37]
no hatching until implantation in vivo [50]
Conceptus reaches the uterus3.5
[51]
3
[29]
2
[30]
4
[31]
4
[44]
5.5–6.5
[40,52]
5–6
[34]
7–10
[41,53,54]
4.5
[36]
3.5
[42]
Conceptus reaches the uterus (stage)Morula
[51]
Morula
[29,55]
4–8 cells
[30]
16 cells
[31]
Morula
(16–32 cells)
[44]
Blastocyst
[40]
Compact morula or blastocyst
[34]
Compact morula or blastocyst
[56]
8–16 cells
[36,57]
Blastocyst
[42]
Implantation6–10
[58]
4.5
[55]
14–18
[59]
19
[31]
16
[44]
~40
[59]
13–14
[60]
From 17–18
[61]
6–7
[49]
7.5
[42]
Time is indicated in days unless otherwise indicated.
Table 2. Common protein-ligand interactions in different species.
Table 2. Common protein-ligand interactions in different species.
OF LigandsReceptor in BlastomeresCitations
Annexin A1ANXA1Epidermal growth factor receptorEGFR[98]
Apolipoprotein C-IIIAPOC3Syndecan 2SDC2
Complement component 3C3CD19 moleculeCD19[99,100]
CD81 moleculeCD81
CalreticulinCALRIntegrin, alpha VITGAV
Fibrinogen gamma chainFGCIntegrin, alpha VITGAV[101]
Integrin, beta 1 (fibronectin receptor, beta polypeptide)ITGB1
Heat shock protein 90 kDa alpha (cytosolic), class A member 1GPICystic fibrosis transmembrane conductance regulatorCFTR
Epidermal growth factor receptorEGFR
LactotransferrinLTFTransferrin receptorTFRC[102]
Glucose-6-phosphate isomeraseGPIAutocrine motility factor receptorAMFR
Table 3. Mammalian species where OVGP1 has been detected.
Table 3. Mammalian species where OVGP1 has been detected.
SpeciesReference
Baboon (Papio anubis)[134,135,136,137,138]
Cat (Felis catus)[139,140,141]
Chimpanzee (Pan troglodytes)[142]
Cow (Bos taurus)[7,125,143,144,145,146]
Dog (Canis lupus familiaris)[147]
Goat (Capra ibex)[148,149]
Hamster (Mesocricetus auratus)[118,124,150,151,152,153,154,155,156]
Human (Homo sapiens)[129,131,157,158,159]
Macaque (Macaca mulatta)[160,161]
Mouse (Mus musculus)[120,162,163]
Pig (Sus scrofa)[164,165,166,167,168]
Rabbit (Oryctolagus cuniculus)[169,170,171]
Sheep (Ovis aries)[172,173,174,175,176]
Table 4. OVGP1 protein detected bonded to the zona pellucida (ZP), oocytes and embryos of different mammalian species. OVGP1 comes from oviductal oocytes (in vivo) or ovarian oocytes exposed to OF, purified protein from oviductal tissue explant or recombinant protein (in vitro).
Table 4. OVGP1 protein detected bonded to the zona pellucida (ZP), oocytes and embryos of different mammalian species. OVGP1 comes from oviductal oocytes (in vivo) or ovarian oocytes exposed to OF, purified protein from oviductal tissue explant or recombinant protein (in vitro).
OVGP1
Source
In Vivo/In VitroTechniqueZPOocyteEmbryoReference
Baboon
(Papio anubis)
Baboonin vivo/in vitroOM++ (PVS)2–4 cells.
+ (ZP, PVS)
[137,156,204]
EM++ (PVS, PM, O)+ (ZP, PVS, BO)
Humanin vitroOM+//[137]
Human
(Homo sapiens)
Humanin vitroOM+//[137]
Baboonin vitro
(Hemizonae)
OM+//[137]
Hamster
(Mesocricetus auratus)
Humanin vitroOM++ (PVS)/[205]
Hamsterin vivo/in vitroOM+-/[96,124,150,151,154,156,205,206,207]
EM++ (PVS, PM, OV)2–8 cells.
+ (ZP, E, L, MVB)
WB+//
Pig
(Sus scrofa)
Pigin vivo/in vitroOM++ (ZP, PM, O)/[113,127,200]
EM++ (PVS, PM, MVB)Day 2–7
+ (ZP, PVS, PM, BM)
MS+//
Cowin vitroOM+//[208]
Cow
(Bos taurus)
Cowin vivo/in vitroOM+-Day 7
+ (ZP)
Day 4–6 and morula + (PVS, BC)
[98,199,209]
MS//+
Pigin vitroOM++ (ZP)+ (ZP, PVS, BM, BC)[201]
Sheep
(Ovis aries)
Sheepin vivo/in vitroOM/EM/WB++ (PVS)+ (ZP, PVS, BC)[173,174]
Mouse
(Mus musculus)
Mousein vivo/in vitroOM+/−+ (PVS)2 cells
+ (ZP, PVS)
[130,162,202,203]
EM+ (PVS)/
WB+/−+/−/
OM: optical microscopy, EM: electron microscopy, WB: Western-blot, MS: Mass spectrometry, ZP: zona pellucida, PVS: perivitelline space, PM: plasma membrane, O: ooplasm, OV ooplasm vesicles, BM: blastomere membrane, B: blastomere cytoplasm, E: endosome, L: secondary lysosome, MVB: multivesicular bodies, (+): positive labelled, (−): negative labelled.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

González-Brusi, L.; Algarra, B.; Moros-Nicolás, C.; Izquierdo-Rico, M.J.; Avilés, M.; Jiménez-Movilla, M. A Comparative View on the Oviductal Environment during the Periconception Period. Biomolecules 2020, 10, 1690. https://doi.org/10.3390/biom10121690

AMA Style

González-Brusi L, Algarra B, Moros-Nicolás C, Izquierdo-Rico MJ, Avilés M, Jiménez-Movilla M. A Comparative View on the Oviductal Environment during the Periconception Period. Biomolecules. 2020; 10(12):1690. https://doi.org/10.3390/biom10121690

Chicago/Turabian Style

González-Brusi, Leopoldo, Blanca Algarra, Carla Moros-Nicolás, Mª José Izquierdo-Rico, Manuel Avilés, and Maria Jiménez-Movilla. 2020. "A Comparative View on the Oviductal Environment during the Periconception Period" Biomolecules 10, no. 12: 1690. https://doi.org/10.3390/biom10121690

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop