Skip to main content

Functional glycosylation in the human and mammalian uterus

Background

Glycosylation is the most common and structurally diverse of all the post-translational modifications of proteins. Lipids and extracellular matrices are also often glycosylated. The mammalian uterus is highly enriched in glycoconjugates that are associated with the apical surfaces of epithelial cells and the secretions released by both epithelial and stromal cells. These glycoconjugates interact primarily with sperm, the implanting embryo, the fetus, and any pathogen that happens to gain entry into the uterus. Secretions of the endometrial glands increase substantially during the luteal phase of the menstrual cycle. These secretions are highly enriched in glycoproteins and mucins that promote specific uterine functions.

Findings

Lectins and antibodies have been employed in the majority of the studies focused on uterine glycosylation have employed to define the expression of carbohydrate sequences. However, while these studies provide insight about potential glycosylation, precise information about glycan structure is lacking. Direct sequencing studies that employ biochemical or mass spectrometric methods are far more definitive, but have rarely been employed with uterine glycoproteins. Both lectin/antibody binding and direct carbohydrate sequencing studies that have been focused on the mammalian uterus are reviewed. The primary functional role of the eutherian uterus is to facilitate fertilization and nurture the developing embryo/fetus. Trophoblasts are the primary cells that mediate the binding of the embryo and placenta to the uterine lining. In mammals that utilize hemochorial placentation, they invade the decidua, the specialized endometrial lining that forms during pregnancy. Trophoblasts have also been analyzed for their lectin/antibody binding as a complement to the analysis of the uterine cells and tissues. They will also be reviewed here.

Conclusions

The functional roles of the glycans linked to uterine and trophoblast glycoconjugates remain enigmatic. Another major question in the human is whether defects in placental or uterine glycosylation play a role in the development the Great Obstetrical Syndromes. More recent findings indicate that changes in glycosylation occur in trophoblasts obtained from patients that develop preeclampsia and preterm birth. The functional significance of these changes remain to be defined. Whether such shifts happen during the development of other types of obstetrical syndromes remains to be determined.

Introduction

Glycosylation is a specific type of post-translational modification of proteins, lipids and other cellular components that is universally observed throughout the plant and animal kingdoms [1, 2]. The plasma membranes of cells, extracellular matrices and connective tissues are the primary sites where abundant glycosylation is observed [2]. Carbohydrate sequences expressed on the outer surfaces of cells participate in binding to other cell types and crucial signaling events during both physiological and pathological states [3, 4]. It is therefore not that surprising that glycans are profusely expressed in the mammalian uterus, an organ that must undergo many different transformations to support fertilization and subsequent fetal development [5, 6]. Though there is currently rather limited data available about the precise sequences of glycans linked to uterine glycoconjugates, the advent of ultrasensitive mass spectrometric (MS) methods combined with a greater appreciation of the role of glycosylation in reproduction should provide strong incentives in the future for the glycomic analysis of cells and tissues within this organ [710]. Here both classical and modern studies focused on uterine glycosylation will be reviewed.

Glycans as essential functional groups that facilitate reproduction

Monroy provided the first convincing evidence that carbohydrate recognition is essential for sperm-egg binding. Specifically, he proposed that polysaccharides or glycans presented on the egg jelly coat of marine organisms were recognized by lectin-like egg binding proteins on the surface of sperm, enabling robust gamete binding [11]. This model for specific cell-cell recognition relies on the strict regulation of carbohydrate expression on both sperm and eggs. The carbohydrate ligands for sperm binding must be expressed at elevated levels on the extracellular matrix of the egg, but not on the sperm surface where they could interact with the lectin-like egg binding proteins on the plasma membrane and inhibit binding. This same type of regulation also applies to cell signaling events involving the specific recognition of carbohydrate ligands. Expression of the carbohydrate ligand for a receptor on the same surface as the receptor could inhibit signaling.

This logic can be directly applied to initial murine and human sperm-egg binding, where substantial evidence supports a specific carbohydrate binding specificity. Data obtained from many different studies indicate that the major egg binding protein on mouse sperm interacts with triantennary and tetraantennary N-glycans terminated with β-linked Gal presented on the constituent glycoproteins that form the zona pellucida (ZP) [1214]. Similarly, the major egg binding protein on sperm has been proposed to recognize multivalent sialyl-Lewisx sequences presented on both N- and O-glycans of ZP glycoproteins during initial gamete binding in humans. Clearly, these carbohydrate ligands for murine and human gamete binding should not be expressed on the plasma membranes of mouse and human sperm. Otherwise, they could interact with the lectin-like egg binding proteins to block binding. Glycomic studies have demonstrated this restriction in the human model. As noted previously, sialyl-Lewisx (sLex) is the carbohydrate ligand on the ZP that mediates human sperm-ZP binding [15]. However, glycomic analysis did not indicate the expression of sLex on human sperm glycans, in spite of the presence of both highly sialylated and fucosylated glycans [16]. Evidence supporting this type of restriction in other species can be obtained in the future by performing detailed glycomic analyses of mature sperm and ZP glycoproteins.

It is quite easy to understand how receptor-ligand systems involving protein-protein interactions between different cell types could be regulated by the genome, but much more difficult for carbohydrate-dependent interactions [17]. Unlike proteins, carbohydrate sequences are assembled via a template-independent process [2]. Their synthesis relies upon the availability of specific modification enzymes (glycosyltransferases, glycosidases), sugar nucleotide sugars and protein substrates [2, 18, 19]. Other relevant factors include the competition for substrate glycans by different enzymes, and the organization of enzymes into complexes or organelles like the Golgi apparatus [20, 21].

Precisely how the expression of complex glycans is regulated remains an enigma. Nonetheless, recent evidence indicates that glycans have been employed as functional groups since the initiation of life on this planet. The synthesis of carbohydrate sequences has been documented in Archaean prokaryotes that date back more than 3500 mya [22]. Recent findings suggest that glycosylation was likely essential to enable these ancient organisms to survive the very harsh environmental conditions that existed during the early stages of earth’s history. The functional roles for glycans has greatly expanded over the eons. Evidence for sexual reproduction has been identified in the fossils of bangiacean red algae (Bangiomorpha pubescens) that date back 1200 mya [23]. These results indicate that the pathways for regulating the expression of carbohydrate functional groups and their cognate receptors on different gametes have likely existed for millennia. The fact that they are still employed in humans confirms that the functional roles requiring carbohydrate recognition remain under positive selection.

Abnormal glycosylation contributes to the development of many different pathological states in humans [2]. The Great Obstetrical Syndromes (preterm labor, preeclampsia, intrauterine growth restriction, preterm premature rupture of membranes, late spontaneous abortion, abruptio placentae) remain the issues of foremost concern for clinicians devoted to the delivery of healthy infants [2428]. All of these pathological states are clearly associated with disorders of deep placentation [29]. Whether any of these syndromes are the result of defective uterine glycosylation has yet to be determined. Unlike genetic or epigenetic changes, subtle shifts in glycosylation are completely invisible to the current methods of genomic analysis. However, they can be readily revealed by careful glycomic analysis of glycoconjugates and whole cell types isolated from normal and pathological tissue samples [7, 30, 31]. The great majority of the studies focused on analyzing uterine glycosylation have been performed with lectins and carbohydrate-specific antibodies. However, ultrasensitive MS analyses will be essential to precisely define discrete differences in glycosylation between normal and pathological states in the human uterus that could result in the development of these obstetrical syndromes [710]. Studies focused on the expression of glycosyltransferase genes in the uterus will also be reviewed.

Analysis of human uterine glycoconjugates with carbohydrate binding proteins

The human uterus is highly enriched in glycoconjugates that are associated with the apical surfaces of epithelial cells and the aqueous secretions released by both epithelial and stromal cells. These glycoconjugates interact primarily with sperm, the implanting embryo, the fetus, and any pathogen that gains entry into the uterus. Secretions of the endometrial glands increase substantially during the luteal phase of the menstrual cycle. These secretions are highly enriched with growth factors and nutrients that support the implantation of the embryo and its subsequent development into a viable fetus.

Glycosylation in the human uterus has been studied primarily by employing lectins and carbohydrate-specific antibodies [32, 33]. This approach was initially necessary because of the limited amount of available tissue/cells and the relative insensitivity of the methods of carbohydrate structural analysis. Lectins are proteins that recognize and bind to carbohydrate sequences that express specific structural features [34]. Many lectins with different carbohydrate binding specificities have been isolated and purified to homogeneity since 1970 [3438]. The major lectins employed to profile glycosylation in the many different human cell types are shown in Table 1.

Table 1 Binding specificities of lectins commonly employed to analyze glycosylation

Though lectins are useful tools, they cannot provide precise details about glycan expression, due to their potential for cross reactivity with unknown carbohydrate sequences and the enormous structural diversity of glycans, especially those that possess multiple non-reducing terminals due to branching. However, when employed in conjunction with ultrasensitive MS sequencing tools, lectins can be very useful for precisely defining structure-function relationships. The unambiguous identification of the glycoprotein ligands for DC-SIGN in human seminal plasma could only be accomplished by employing lectin affinity chromatography in conjunction with glycomic and proteomic analyses, as demonstrated in a recent study [39].

Differential agglutination of human tumor cells compared to normal progenitor cells by wheat germ agglutinin (WGA) was initially reported in 1965 [40]. This observation led many investigators working in cancer research to employ lectins in their comparative studies of normal versus tumorigenic tissues and cell types. This interest in differential glycosylation during tumorigenesis also stimulated many investigators to define the carbohydrate binding properties of the lectins shown in Table 1.

Many different lectin binding studies have been performed on tissue samples obtained from the human uterus and cervix (Table 2). In an early study, Rowinski and coworkers reported that fibroblasts obtained from the normal human cervix were not agglutinated by the lectin Concanavalin A (ConA). By contrast, fibroblasts underlying different cervical cancer lesions became agglutinated with lower concentrations of this lectin as the tumor progressed [41]. Kluskens et al. [42] analyzed the binding of 7 FITC-labeled lectins to proliferative, hyperplastic and cancerous endometrial samples. They were able to define differences in the binding of WGA and ConA to these samples. The sialic acid binding lectin from Limulus polyphemus was employed to investigate the changes in the expression of sialylated glycoconjugates in human endometrial adenocarcinoma after treatment with medroxyprogesterone acetate [43]. They observed specific quantitative and qualitative differences in lectin binding after therapy with this hormone.

Table 2 Lectin binding to human uterine tissues

Damjanov and coworkers investigated the binding of a panel of 13 different fluoresceinated lectins to normal human endocervical and uterine epithelium at different stages of the menstrual cycle [44]. They reported that MPA, UEA-I, SBA and VVA were selectively bound to the endocervix but not the endometrium, indicating that lectins could be employed to distinguish between epithelia at different uterine sites. They also demonstrated that these variations were independent of the menstrual cycle and blood group status. Bychkov and Toto employed the avidin-biotin-peroxidase method to analyze the binding of PNA, UEA-1, WGA and ConA to samples of endometrium during different stages of the menstrual cycle and early pregnancy [45]. They reported very strong binding of PNA and UEA-1 to apical cells during early pregnancy, but only weak binding during the proliferative and secretory phases. WGA and RCA-1 displayed marginal binding to glandular epithelium during the proliferative phase that increased substantially during the secretory phase. This same investigative group employed WGA and PNA as probes to analyze normal, dysplastic and neoplastic cervical epithelium [46]. They observed minimal binding of these lectins to normal squamous epithelium which increased substantially as the lesions became more malignant.

Wan and coworkers used a panel of nineteen FITC-labeled lectins to define the glycosylation of the epithelial surfaces in the human female reproductive tract including the uterus and cervix. They concluded that the distribution of galactosyl residues displayed variations among the organs, unlike mannosylated and fucosylated residues that were more evenly expressed [47]. Tang studied the binding of UEA-1, GS-I (isolectin B4), and DBA to normal and malignant cells of the uterine endometrium [48]. DBA binding decreased while UEA-1 binding increased on tumorigenic luminal cells compared to normal progenitor cells. Griffin and Wells employed a panel of eleven biotinylated lectins to compare the glycosylation of normal cervical glands for comparison with cervical glandular intraepithelial neoplasia and invasive adenocarcinoma [49]. Foster and coworkers analyzed the binding of an extensive panel of lectins to define changes in glycan expression that accompanied the transition from normal to cancerous lesions in human cervical epithelium [50]. They suggested that the expression of novel carbohydrate sequences by cancer cells could substantially promote their invasion and dissemination.

Nagai and associates investigated the binding of UEA, DBA, ConA and PHA to normal and neoplastic glandular epithelium from the human endocervix and endometrium [51]. They reported that the intensity and staining pattern of lectin binding were useful for differentiating between endocervical and endometrial epithelium derived from either normal or neoplastic tissue. The effect of hormonal cycling on the glycosylation of the human oviduct has been studied by employing five horseradish peroxidase-labeled lectins (PNA, SBA, DBA, WGA, ConA, LTA, UEA-I) [52]. Substantial losses in the binding of DBA, WGA and ConA lectins was observed in postmenopausal women compared to the menstruating women. Argueso et al. employed PNA and ECA to analyze the expression of T antigen and N-acetyllactosamine in the human mucin MUC5B during the menstrual cycle [53]. This investigative group reported that the expression of these sequences increased steadily up to midcycle and then dramatically declined by the end of the cycle.

PWM and WGA have recently been employed to analyze the expression of glycans in the human uterine cervix and cervical lesions [54]. Enhanced binding of PWM was observed in squamous carcinoma compared to premalignant lesions (premalignant cervical intraepithelial neoplasia grades 1–3) and normal cervical epithelium. By contrast, the binding of WGA uniformly decreased as the cancerous lesions became more aggressive. Analysis of cervical lesions by lectin blot and enzyme-linked lectin assay (ELLA) also indicated decreased sialylation and fucosylation of cancerous cervical lesions compared to normal epithelium [55].

Analysis of mammalian uterine glycosylation with carbohydrate binding proteins

The interaction of lectins with uterine tissues has also been studied in many other non-human species (Table 3). Roberts and coworkers isolated a plasma membrane fraction from the luminal surface of the pig uterus during the estrous cycle and early pregnancy [56]. These investigators separated the membrane glycoproteins in this fraction by 2-D gel electrophoresis and stained them with radioiodinated ConA and RCA-I. However, no major changes in either protein expression or glycosylation was detected during either the estrous cycle or early pregnancy with these specific lectin probes [56].

Table 3 Lectin and antibody binding to mammalian uterine tissues

In 1983, Damjanov and coworkers employed a panel of fluorescein-conjugated lectins to probe the luminal epithelium lining the murine oviduct and uterus [57]. They were able to demonstrate that WGA, BPA, RCA-I, MPA and UEA-1 displayed differential binding to the epithelial surface of the pregnant versus non-pregnant uterus. This differential binding was also regionally specific with WGA, indicating that changes in the binding of RCA-I, MPA and WGA delineated pregnancy-related changes in the distal oviduct and colliculus tubaris. WGA could also distinguish pregnancy related changes in the proximal oviduct. UEA-I alone reacted exclusively with the epithelium of the non-pregnant uterus. RCA-II reacted preferentially with the epithelium of the oviduct and uterus as compared with its weak reactivity with the stroma. Two lectins (PSA, LCA) reacted selectively with stromal cells of the uterus and oviduct [57].

Aplin and coworkers analyzed the expression of eleven different lectins and two monoclonal antibodies directed against carbohydrate sequences (keratan sulfate, sialyl-Tn antigen) to investigate glycan expression in the oviduct and the endometrium during the luteal phase of Cebus apella, a New World monkey [58]. Jones et al. recently employed a panel of twenty-four different lectins to investigate the glycosylation of the placenta and the uterus in a marsupial, the tammar wallaby Macropus eugenii [59]. Feline decidual cells displayed weak binding for GS-I, ConA, DBA, DSA, PNA, RCA-I, SBA and SJA in another study [60]. ConA, LCA, SNA, RCA-I, PNA, SBA and HPA were among the lectins that were shown to bind to normal rat endometrium [61]. Several groups have analyzed the binding of a panel of lectins to canine endometrial mucosa that in some cases displayed variation depending on the stage of presentation (immature, oestrus, young anoestrus, aged anoestrus) and disease status [6265]. Georgiades and coworkers analyzed the binding of GS-I to mouse decidual stromal cells during pregnancy. They reported staining of these cells with GS-I in the venous sinusoid area of the decidual basalis by embryonic day 7.5 of pregnancy and in the entire basalis by embryonic day 10.5 and afterwards [66].

Biochemical and MS analyses of uterine glycoconjugates in the human and mouse

Though lectins are useful for analyzing the glycosylation of uterine surfaces, they are not nearly as powerful as carbohydrate sequencing tools involving biochemical and MS approaches. Yurewicz and Moghissi isolated sixteen different O-glycans from a pool of human midcycle cervical mucin samples [67]. These investigators subsequently labeled these oligosaccharides with tritium at their reducing ends, enabling them to be sensitively detected during the procedures that enabled their sequence to be defined. Analysis of the neutral O-glycans yielded evidence for the existence of core 2 type O-glycans terminated with H type 2, Lewisx/a, and potential Lewisy/b sequences [68]. A core 2 O-glycan terminated with either sialyl Lewisx and/or sialyl Lewisa was the most unusual oligosaccharide revealed during the sequencing of the sialylated fraction [69]. A detailed glycomic analysis of human cervical mucins expressed during the menstrual cycle was more recently performed by employing ultrasensitive MS methods [70]. At least 50 different neutral, sialylated and sulfated O-glycans were detected. The previous findings reported by Yurewicz and Moghissi were confirmed during this study [68, 69]. Hansson and coworkers did not detect any changes in protein or mucin composition in the cervical plug during the menstrual cycle, but they did observe a relative increase in the expression of neutral fucosylated O-glycans during the ovulatory phase [70].

MS methods have also been employed to analyze the glycans associated with specific uterine or decidual glycoproteins. Perhaps the best studied is glycodelin, a glycoprotein of endometrial and decidual origin that was originally isolated by Bohn from the placenta and designated PP14 [71]. The amniotic fluid specific form of glycodelin (GdA) was originally shown to display several different immunomodulatory activities and the ability to block human sperm-ZP binding in the hemizona assay system at low concentrations [7275]. Glycomic analysis revealed the presence of some very unusual carbohydrate antennae on its N-glycans, such as the fucosylated lacdiNAc sequence (GalNAcβ1-4[Fucα1-3]GlcNAc) [75, 76]. Currently, there are several isoforms of glycodelin that have been identified in the follicular fluid (GdF), cumulus matrix (GdC) and seminal plasma (GdS). Each form has its own specific glycosylation state and biological activities, though the protein component remains unchanged [76].

Carson and coworkers demonstrated that β-estradiol stimulated the incorporation of [3H]mannose into mouse uterine glycoproteins by 3–6-fold without stimulating protein synthesis [77]. This increased incorporation was due to enhanced secretion of specific glycoproteins rather than changes in the glycan biosynthetic pathways. This same group later demonstrated that polylactosaminoglycans represent a major fraction of the total glycoconjugates synthesized by epithelial cells but not stromal cells in the mouse uterus [78]. These glycans play a role in cell adhesion processes involving this cell population. The synthesis of these glycans was specifically stimulated by estrogen [79].

Insightful MS analyses have been performed on mouse uterine luminal fluid (ULF) glycoproteins. Glycomic analysis of lipocalin 2 (Lcn2, 24p3) revealed the presence of multiple Lewisx and Lewisy antenna on complex type N-glycans at its only glycosylation site [80]. Lewisx, Lewisy and terminal NeuAcα2-6Gal sequences were predominant in mouse ULF glycome. Several other glycoproteins carriers of these antenna were detected in ULF, including abundant lactotransferrin. The exact physiological significance of this unusual glycosylation pattern remains to be determined.

Analysis of glycosyltransferase gene expression in the human and mammalian uterus

Several groups have studied the expression of different glycosyltransferase genes in the human and mammalian uterus. Levesque et al. confirmed the expression of a UDP-glucuronyltransferase gene in the human uterus by employing an amplification method involving the reverse transcriptase-polymerase chain reaction (RT-PCR) [81]. Kubushiro et al. demonstrated that the level of β1-4 galactosyltransferase enzyme was highly elevated in human endometrial cancer compared to normal endometrium by employing both immunohistochemical approaches and measurement of mRNA levels [82].

Lowe and coworkers employed a similar approach to detect an α1-2 fucosyltransferase (FUT1) in the mouse uterus [83]. Tabak and coworkers confirmed the expression of a UDP-GalNAc:polypeptide N-acetylgalactosaminyltransferase gene that plays a role in O-glycan synthesis in the rat uterus [84]. Robertson and coworkers employed quantitative real-time PCR (qPCR) in murine uterine epithelial cells to demonstrate that α1-2 fucosyltransferase (FUT2) expression in mouse uterine epithelial cells is regulated by leukemia inhibitory factor (LIF) and interleukin-1B (IL-1B) secreted by macrophages [85].

Hormonal regulation of glycosyltransferase gene expression in the uterus has also been demonstrated. Domino and Hurd employed LacZ expression in α1-2 fucosyltransferase (FUT2)-LacZ mice to demonstrate estradiol-regulated endocervical glandular expression during the estrous cycle, hormone replacement and pregnancy [86]. Uchiyama and coworkers analyzed the expression pattern of mRNAs for three hyaluronan synthases (HAS-1, −2, and −3) in the uterine cervix of gravid mice. The expression of HAS-1 and HAS-2 was inhibited by progesterone treatment whereas HAS-3 was substantially increased [87]. Chu and coworkers showed that the progesterone regulated expression of the gene for a specific β1-4 N-acetylgalactosaminyltransferase (B4GALNT2) was required from the time of embryo implantation in mice [88]. Administration of tamoxifen (an estrogen receptor α antagonist) to pregnant mice on d15 resulted in reduce hyaluronan synthase 2 (HAS-2) gene expression in the cervix as quantified by qPCR. This finding also correlated with an overall 50 % decrease in hyaluronan content, indicating that the expression of this glycosaminoglycan is estrogen regulated [89].

Glycosylation and the implantation of human and mammalian embryos

The mammalian embryo must successfully implant and initiate placentation to obtain nutrients and establish gas exchange. Successful implantation requires a receptive uterine lining and the development of the embryo to the blastocyst stage. The trophectodermal cells of the blastocyst must bind to the luminal epithelial cells of the uterus for implantation to proceed.

Initial implantation of the human embryo into the uterine epithelium has also been proposed to depend on carbohydrate recognition. Hey and Aplin initially reported that sialyl-Lewisx and sialyl-Lewisa are expressed on MUC-1 in the human endometrium [90]. Fisher and coworkers demonstrated that human trophoblasts express L-selectin, an adhesion molecule that is also employed during lymphocyte extravasation from the vasculature [91]. They further reported that human uterine epithelial cells increase their expression of selectin ligands (especially 6-sulfo-sialyl Lex) during the temporal window of receptivity. This adhesive interaction is functional, since human trophoblasts specifically bind to beads coated with 6-sulfo-sialyl Lex but not to unrelated carbohydrate ligands [91]. Carson and coworkers subsequently reported that MUC1 also binds to antibodies specific for 6-sulfo-sialyl Lewisx (HECA-452, MECA-79), suggesting a possible role for this mucin in blastocyst implantation [92]. However, while useful, such studies cannot provide precise information about the degree of substitution with these carbohydrate sequences or potential changes in presentation that occur during the receptive period of blastocyst implantation.

Many different cell adhesion molecules have been associated with the implantation of the mouse embryo [33]. There is compelling evidence that lectin-like interactions also play a role in this process. Lundblad and coworkers initially demonstrated that a specific milk oligosaccharide designated lacto-N-fucopentaose I (LNF-1), but not other closely related oligosaccharides inhibited the implantation of mouse embryos by 53 % at a millimolar concentration [93]. This finding was correlated with the expression of LNF-1 on the surface of the murine uterine endometrial epithelium during pregnancy [94]. Another terminal sequence that has been implicated in binding is the H type 1 antigen [93]. Its synthesis relies on the expression of a specific estrogen dependent α1-2 fucosyltransferase [95]. The mRNA for this enzyme is elevated in the preimplantation phase but decreases as implantation proceeds. Intrauterine injection of monoclonal antibodies directed against the Lewisy antigen also inhibits implantation, but only if they are introduced just before this process normally occurs [96]. As noted previously, results obtained in a more recent study have confirmed that Lewisy sequences are also profusely expressed on murine uterine luminal fluid glycoproteins [80].

The isolation and characterization of galectins with different carbohydrate binding specificities led to the concept that there is a glycocode that directs appropriate functions under different physiological states in complex organisms [97, 98]. Subsequent experiments support the hypothesis that there is the specific recognition of a glycocode that functions during the implantation of the mammalian embryo. Jones and Aplin noted that each mammalian species has its own unique pattern of glycosylation of the maternal uterine epithelium based on lectin binding, which they designated as a glycotype [5]. These investigators pointed out that the uterine glycotypes are very similar in cases where interspecies mating results in successful implantation and the development of viable hybrids (horse and donkey, llama and guanaco). They suggest that hybrid embryos are able to implant and develop based partially on their recognition of the shared glycans on the uterine epithelium, i.e., the glycocode [5].

Analysis of human trophoblast glycosylation

Though not actually a part of the uterus, the placenta functions to provide vital nutrients and gas exchange to support the developing eutherian during pregnancy. The placental cells that make intimate contact with the uterine lining are known as trophoblasts. In addition to providing this vital support, these cells are also crucial for establishing an immunological barrier that protects the histoincompatible fetus from the maternal immune response. Because of their importance and relationships to pathological pregnancy states in humans, trophoblasts have been subjected to many different types of biochemical analyses, including lectin binding studies.

Perhaps the most extensive lectin binding analysis of human trophoblasts obtained from term placenta was carried out by Jones and her colleagues [99]. The results of this study are summarized in Table 4. Ezaki and coworkers also analyzed the binding of a panel of lectins to human trophoblasts [100]. However, these investigators reported much lower binding of ConA, E-PHA and LCA to trophoblasts compared to the findings reported by Jones and coworkers.

Table 4 Lectin staining of human trophoblast Domainsab

At this time, the assignment of any specific functional roles for human trophoblast glycans is purely speculative because of the lack of hard evidence. However, one potential role that such glycans could play involves immune recognition. Human syncytiotrophoblast (STB) come into direct physical contact with uterine NK cells during the early stage of implantation. NK cells are specifically sequestered to the implantation site by MIP-1α, a cytokine secreted by STB [101]. STB on the surface of the placental villi also come into direct contact with immune effector cells in the maternal blood. STB lack human leukocyte antigens (HLA), thus likely avoiding alloimmune responses [102, 103]. However, they encounter circulating maternal natural killer (NK) cells at the villous interface. The lack of HLA class I proteins could potentially put them at risk for cytolysis, but only if they express the appropriate NK cell activating ligands [104, 105].

Of even greater potential immune consequence is the expression of paternal HLA-C by extravillous cytotrophoblasts that invade the decidua and the myometrium [102, 103, 106]. How a powerful histocompatibility-based response directed against this type of trophoblast is avoided during pregnancy is currently unknown. In addition, no studies have demonstrated that differential glycosylation of HLA class I molecules could lead to the evasion of this type of immune response. Clearly, ultrasensitive MS analysis of trophoblast populations in the human placenta combined with other functional studies will be useful for determining if glycosylation plays any functional role in the immune deviations that occur during these interactions.

Analysis of mammalian trophoblast glycosylation

Trophoblasts and trophectodermal cells from many other species have also been subjected to staining with lectins to analyze glycan expression and detect differences between species. Jones and coworkers performed lectin binding analysis of trophoblasts isolated from humans and four other species that employ hemochorial implantation (lesser hedgehog tenrec (Echinops telfairi), spotted hyena (Crocuta crocuta), nine-banded armadillo (Dasypus novemcinctus), and guinea pig (Cavia porcellus)) [99]. These investigators noted that the glycosylation patterns were similar to each other and to human trophoblasts, with only minor differences. They suggested that these findings were evidence for convergent evolution [99]. Lectin binding studies have also been performed on trophoblasts from many other mammalian species. These studies are presented in tabular form in Table 5. The intraepithelial binucleate cells present in ruminants have often been the focus of many of these other investigations.

Table 5 Analysis of lectin binding to mammalian trophoblasts

Glycosylation and the development of the great obstetrical syndromes

The Great Obstetrical Syndromes are of the utmost concern for the practicing obstetrician [107]. The existing data indicate that these syndromes are due to defects in deep implantation. Perhaps one of the most puzzling of these syndromes is preeclampsia (PE). The thoughtful obstetrician Jeffcoate referred to PE as “the Disease of Theories” because of the numerous research challenges associated with this pathological condition [108]. This disorder can be broadly divided into two classes, sometimes referred to as maternal and placental, though in some cases a mixture of the two types is observed [109, 110]. Placental PE is the result of poor placentation during early pregnancy. PE has been hypothesized to be the result of: (i) a disruption of vascular remodeling leading to hypoxia; and/or (ii) an aberrant immune response directed against the allogeneic fetus [111]. There is strong evidence indicating that both processes play crucial roles in the clinical manifestation of PE.

Sgambati and coworkers previously analyzed the distribution of sugar residues in human placentas from uncomplicated pregnancies and those affected by different hypertensive disorders (pregnancy-induced hypertension (PIH), PE, PE with hemolysis, elevated liver enzymes and low platelets (HELLP) syndrome) [112]. They employed ConA, WGA, PNA, SBA, DBA, UEA, GNA, DSA, MAA and DSA in combination with other chemical and enzymatic treatments to perform this analysis. They reported a 40 % increase in ConA binding to STB and CTB in placentas derived from patients that developed PE or PE with HELLP syndrome compared to TB derived from women that developed pregnancy-induced hypertension or that delivered without complications. No binding sites for DBA or SBA were detected on STB and CTB in the placentas of patients with uncomplicated deliveries. However, substantial binding of these lectins was observed on STB and CTB associated with the placentas of patients that developed PIH, PE or PE with HELLP syndrome. Binding sites for SNA were expressed on STB from the placentas of patients that developed PE with HELLP, but not on TB from any other patient group analyzed in this study [112]. Clearly, these results indicate that a shift in glycosylation is occurring during the development of these obstetrical syndromes, but how these changes impact this condition remains to be defined.

Potential shifts in glycosylation have recently been indicated during the development of preterm birth. Integrin β1 was isolated from villous samples obtained 6–9 weeks of gestation from placentas obtained from early spontaneous miscarriage and normal controls [113]. Binding of L-PHA, a lectin that specifically recognizes N-glycans bearing the Galβ1-4GlcNAcβ1-6 Man sequence was decreased in integrin samples isolated from patients that experienced a miscarriage compared to controls. By contrast, the level of binding of E-PHA to integrin β1 substantially increased in samples derived from miscarriage patients compared to normal pregnancies. These shifts in glycosylation were correlated with the level of the N-acetylglucosaminyltransferase enzymes known as Mgat 5 and GnT-III that add β1-6 linked and the bisecting GlcNAc to the trimmanosyl core of N-glycans, respectively [113]. Whether these shifts in glycosylation play a role in the development of preterm birth remains to be defined.

Conclusions

Many studies have focused considerable effort on defining glycosylation in the mammalian uterus and placenta. Nonetheless, the functional roles of glycans have not been explicitly defined. Studies focused in this area could be extremely valuable in the human, where the cause of many of the Great Obstetrical Syndromes remain enigmatic [107]. Though powerful methods of both genetic and epigenetic analysis are currently available to analyze uterine and trophoblast function, no definitive cause of these major obstetrical syndromes has been determined. Evidence is now available indicating that the glycosylation of STB and CTB are undergoing substantial changes during the development of hypertensive disorders of pregnancy including PIH, PE and PE with HELLP syndrome [112]. Shifts in the glycosylation of β1 integrin have been detected in the placentas of women who developed preterm birth [113]. The time has now come to employ biochemical and ultrasensitive MS tool to analyze the pathways for glycosylation in the uterus in both the pregnant and nonpregnant women during both normal and aberrant physiological states. The possibility that aberrant glycosylation could play a major role in the development of the Great Obstetrical Syndromes should now be seriously considered.

Abbreviations

GdA:

Amniotic glycoforms of glycodelin

GdF:

Follicular fluid glycoforms of glycodelin

GdS:

Seminal plasma glycoforms of glycodelin

HELLP:

Hemolysis, elevated liver enzymes and low platelets

MS:

Mass spectrometric. A list of the standard abbreviations for lectins referred to in text is provided in Table 1

References

  1. Varki A. Biological roles of oligosaccharides: all of the theories are correct. Glycobiology. 1993;3(2):97–130.

    Article  CAS  PubMed  Google Scholar 

  2. Varki A, Cummings RD, Esko JD, Freeze HH, Stanley P, Bertozzi CR, et al., editors. Essentials of Glycobiology. 2nd ed. Cold Spring Harbor: Cold Spring Harbor Laboratory Press; 2009.

    Google Scholar 

  3. Varki A. Glycan-based interactions involving vertebrate sialic-acid-recognizing proteins. Nature. 2007;446(7139):1023–9.

    Article  CAS  PubMed  Google Scholar 

  4. Ohtsubo K, Marth JD. Glycosylation in cellular mechanisms of health and disease. Cell. 2006;126(5):855–67.

    Article  CAS  PubMed  Google Scholar 

  5. Jones CJ, Aplin JD. Glycosylation at the fetomaternal interface: does the glycocode play a critical role in implantation? Glycoconjugate J. 2009;26(3):359–66.

    Article  CAS  Google Scholar 

  6. Carson DD. The glycobiology of implantation. Front Biosci. 2002;7:d1535–44.

    Article  PubMed  Google Scholar 

  7. North SJ, Hitchen PG, Haslam SM, Dell A. Mass spectrometry in the analysis of N-linked and O-linked glycans. Curr Opin Struct Biol. 2009;19(5):498–506.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Geyer H, Geyer R. Strategies for analysis of glycoprotein glycosylation. Biochim Biophys Acta. 2006;1764(12):1853–69.

    Article  CAS  PubMed  Google Scholar 

  9. Marino K, Bones J, Kattla JJ, Rudd PM. A systematic approach to protein glycosylation analysis: a path through the maze. Nat Chem Biol. 2010;6(10):713–23.

    Article  CAS  PubMed  Google Scholar 

  10. Dell A, Morris HR. Glycoprotein structure determination by mass spectrometry. Science. 2001;291(5512):2351–6.

    Article  CAS  PubMed  Google Scholar 

  11. Monroy A. Chemistry and Physiology of Fertilization. New York: Holt, Rinehart and Winston; 1965.

    Google Scholar 

  12. Clark GF, Dell A. Molecular models for murine sperm-egg binding. J Biol Chem. 2006;281(20):13853–6.

    Article  CAS  PubMed  Google Scholar 

  13. Clark GF. Molecular models for mouse sperm-oocyte binding. Glycobiology. 2011;21(1):3–5.

    Article  CAS  PubMed  Google Scholar 

  14. Clark GF, Oehninger S, Seppala M. Role for glycoconjugates in cellular communication in the human reproductive system. Mol Hum Reprod. 1996;2:513–7.

    Article  CAS  PubMed  Google Scholar 

  15. Pang PC, Chiu PC, Lee CL, Chang LY, Panico M, Morris HR, et al. Human sperm binding is mediated by the sialyl-Lewisx oligosaccharide on the zona pellucida. Science. 2011;333(6050):1761–4.

    Article  CAS  PubMed  Google Scholar 

  16. Pang PC, Tissot B, Drobnis EZ, Sutovsky P, Morris HR, Clark GF, et al. Expression of bisecting type and Lewisx/Lewisy terminated N-glycans on human sperm. J Biol Chem. 2007;282(50):36593–602.

    Article  CAS  PubMed  Google Scholar 

  17. Murrell MP, Yarema KJ, Levchenko A. The systems biology of glycosylation. Chembiochem. 2004;5(10):1334–47.

    Article  CAS  PubMed  Google Scholar 

  18. Moremen KW, Tiemeyer M, Nairn AV. Vertebrate protein glycosylation: diversity, synthesis and function. Nat Rev Mol Cell Biol. 2012;13(7):448–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Nairn AV, York WS, Harris K, Hall EM, Pierce JM, Moremen KW. Regulation of glycan structures in animal tissues: transcript profiling of glycan-related genes. J Biol Chem. 2008;283(25):17298–313.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Farquhar MG, Palade GE. The Golgi apparatus: 100 years of progress and controversy. Trends Cell Biol. 1998;8(1):2–10.

    Article  CAS  PubMed  Google Scholar 

  21. Dennis JW, Nabi IR, Demetriou M. Metabolism, cell surface organization, and disease. Cell. 2009;139(7):1229–41.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Eichler J. Extreme sweetness: protein glycosylation in archaea. Nat Rev Microbiol. 2013;11(3):151–6.

    Article  CAS  PubMed  Google Scholar 

  23. Butterfield NJ. Bangiomorpha pubescens n. gen., n. sp.: implications for the evolution of sex, multicellularity, and the Mesoproterozoic/Neoproterozoic radiation of eukaryotes. Paleobiology. 2000;26(3):386–404.

    Article  Google Scholar 

  24. Goldenberg RL, Culhane JF, Iams JD, Romero R. Epidemiology and causes of preterm birth. Lancet. 2008;371(9606):75–84.

    Article  PubMed  Google Scholar 

  25. Fretts RC. Etiology and prevention of stillbirth. Am J Obstet Gynecol. 2005;193(6):1923–35.

    Article  PubMed  Google Scholar 

  26. Duley L. The global impact of pre-eclampsia and eclampsia. Semin Perinatol. 2009;33(3):130–7.

    Article  PubMed  Google Scholar 

  27. Mercer BM. Preterm premature rupture of the membranes. Obstet Gynecol. 2003;101(1):178–93.

    PubMed  Google Scholar 

  28. Romero R, Dey SK, Fisher SJ. Preterm labor: one syndrome, many causes. Science. 2014;345(6198):760–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Brosens I, Pijnenborg R, Vercruysse L, Romero R. The ‘Great Obstetrical Syndromes’ are associated with disorders of deep placentation. Am J Obstet Gynecol. 2011;204(3):193–201.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Chui D, Sellakumar G, Green R, Sutton-Smith M, McQuistan T, Marek K, et al. Genetic remodeling of protein glycosylation in vivo induces autoimmune disease. Proc Natl Acad Sci U S A. 2001;98(3):1142–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Wang Y, Tan J, Sutton-Smith M, Ditto D, Panico M, Campbell RM, et al. Modeling human congenital disorder of glycosylation type IIa in the mouse: conservation of asparagine-linked glycan-dependent functions in mammalian physiology and insights into disease pathogenesis. Glycobiology. 2001;11(12):1051–70.

    Article  CAS  PubMed  Google Scholar 

  32. Aplin JD. Adhesion molecules in implantation. Rev Reprod. 1997;2(2):84–93.

    Article  CAS  PubMed  Google Scholar 

  33. Kimber SJ. Molecular interactions at the maternal-embryonic interface during the early phase of implantation. Semin Reprod Med. 2000;18(3):237–53.

    Article  CAS  PubMed  Google Scholar 

  34. Goldstein IJ, Hayes CE. The lectins: carbohydrate-binding proteins of plants and animals. Adv Carbohydr Chem Biochem. 1978;35:127–340.

    Article  CAS  PubMed  Google Scholar 

  35. Stillmark H. Uber Rizen, ein giftiques Ferment aus dem Samen von Ricinus communis L. und einigen anderen Euphorbiaceen. Dorpat: University of Tartu; 1888.

  36. Liener I, Sharon N, Goldstein IJ. The Lectins: Properties, Functions, and Applications in Biology and Medicine. Orlando: Academic; 1986. p. 603 p.

    Google Scholar 

  37. Goldstein IJ. Lectin structure-activity: the story is never over. J Agric Food Chem. 2002;50(22):6583–5.

    Article  CAS  PubMed  Google Scholar 

  38. Hirabayashi J, Kuno A, Tateno H. Lectin-based structural glycomics: a practical approach to complex glycans. Electrophoresis. 2011;32(10):1118–28.

    Article  CAS  PubMed  Google Scholar 

  39. Clark GF, Grassi P, Pang PC, Panico M, Lafrenz D, Drobnis EZ, et al. Tumor biomarker glycoproteins in the seminal plasma of healthy human males are endogenous ligands for DC-SIGN. Mol Cell Proteomics. 2012;11(1):M111.008730.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Aub JC, Sanford BH, Cote MN. Studies on reactivity of tumor and normal cells to a wheat germ agglutinin. Proc Natl Acad Sci U S A. 1965;54(2):396–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Chaudhuri S, Koprowska I, Rowinski J. Different agglutinability of fibroblasts underlying various precursor lesions of human uterine cervical carcinoma. Cancer Res. 1975;35(9):2350–54.

    CAS  PubMed  Google Scholar 

  42. Kluskens LF, Kluskens JL, Bibbo M. Lectin binding in endometrial adenocarcinoma. Am J Clin Pathol. 1984;82(3):259–66.

    Article  CAS  PubMed  Google Scholar 

  43. Barni S, Gerzeli G, Novelli G. Cytochemistry of cell surface sialoglycoconjugates in endometrial adenocarcinoma. Effects of medroxyprogesterone therapy. Applied Pathol. 1984;2(3):135–45.

    CAS  Google Scholar 

  44. Yen Y, Lee MC, Salzmann M, Damjanov I. Lectin binding sites on human endocervix: a comparison with secretory and proliferative endometrium. Anat Rec. 1986;215(3):262–6.

    Article  CAS  PubMed  Google Scholar 

  45. Bychkov V, Toto PD. Lectin binding to normal human endometrium. Gynecol Obstet Invest. 1986;22(1):29–33.

    Article  CAS  PubMed  Google Scholar 

  46. Bychkov V, Toto PD. Wheat germ and peanut agglutinin binding to normal, dysplastic and neoplastic cervical epithelium. Gynecol Obstet Invest. 1986;21(3):158–63.

    Article  CAS  PubMed  Google Scholar 

  47. Wu TC, Lee SM, Jih MH, Liu JT, Wan YJ. Differential distribution of glycoconjugates in human reproductive tract. Fertil Steril. 1993;59(1):60–4.

    Article  CAS  PubMed  Google Scholar 

  48. Tang J-W. Expression of blood group antigen associated lectin receptors in normal and malignant tissues of uterine endometrium Chinese. J Clin Oncol. 1994;21(12):922–5.

    Google Scholar 

  49. Griffin NR, Wells M. Characterisation of complex carbohydrates in cervical glandular intraepithelial neoplasia and invasive adenocarcinoma. Int J Gynecol Pathol. 1994;13(4):319–29.

    Article  CAS  PubMed  Google Scholar 

  50. Banerjee S, Robson P, Soutter WP, Foster CS. Modulated expression of glycoprotein oligosaccharides identifies phenotypic differentiation in squamous carcinomas of the human cervix. Hum Pathol. 1995;26(9):1005–13.

    Article  CAS  PubMed  Google Scholar 

  51. Toda T, Sadi AM, Egawa H, Atari E, Qureshi B, Nagai Y. Affinity of four lectins for endocervical and endometrial non-neoplastic and neoplastic glandular epithelium. Histopathology. 1998;32(3):257–63.

    Article  CAS  PubMed  Google Scholar 

  52. Gheri G, Noci I, Sgambati E, Borri P, Taddei G, Bryk SG. Ageing of the human oviduct: lectin histochemistry. Histol Histopathol. 2001;16(1):21–8.

    CAS  PubMed  Google Scholar 

  53. Argueso P, Spurr-Michaud S, Tisdale A, Gipson IK. Variation in the amount of T antigen and N-acetyllactosamine oligosaccharides in human cervical mucus secretions with the menstrual cycle. J Clin Endocrinol Metab. 2002;87(12):5641–8.

    Article  CAS  PubMed  Google Scholar 

  54. Clark AT, Guimaraes Da Costa VM, Bandeira Costa L, Bezerra Cavalcanti CL, De Melo Rego MJ, Beltrao EI. Differential expression patterns of N-acetylglucosaminyl transferases and polylactosamines in uterine lesions. Eur J Histochem. 2014;58(2):2334.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Kim HJ, Kim SC, Ju W, Kim YH, Yin SY, Kim HJ. Aberrant sialylation and fucosylation of intracellular proteins in cervical tissue are critical markers of cervical carcinogenesis. Oncol Rep. 2014;31(3):1417–22.

    CAS  PubMed  Google Scholar 

  56. Mullins DE, Horst MN, Bazer FW, Roberts RM. Isolation and characterization of a plasma membrane fraction derived from the luminal surface of the pig uterus during the estrous cycle and early pregnancy. Biol Reprod. 1980;22(5):1181–92.

    CAS  PubMed  Google Scholar 

  57. Lee MC, Wu TC, Wan YJ, Damjanov I. Pregnancy-related changes in the mouse oviduct and uterus revealed by differential binding of fluoresceinated lectins. Histochemistry. 1983;79(3):365–75.

    Article  CAS  PubMed  Google Scholar 

  58. Jones CJ, Ortiz ME, Croxatto HB, Manzur A, Slevin G, Aplin JD. Muc1 and glycan expression in the oviduct and endometrium of a New World monkey. Cebus apella Biol Reprod. 2001;64(5):1535–44.

    Article  CAS  PubMed  Google Scholar 

  59. Jones CJ, Skepper JN, Renfree MB, Aplin JD. Trophoblast specialisations during pregnancy in the tammar wallaby, Macropus eugenii: a morphological and lectin histochemical study. Placenta. 2014;35(7):467–75.

    Article  CAS  PubMed  Google Scholar 

  60. Fernandez PE, Diessler ME, Pachame A, Ortega HH, Gimeno EJ, Portiansky EL, et al. Intermediate filament proteins expression and carbohydrate moieties in trophoblast and decidual cells of mature cat placenta. Reprod Domest Anim. 2014;49(2):263–9.

    Article  CAS  PubMed  Google Scholar 

  61. Lutsyk A, Sogomonian E. Structural, functional, and lectin histochemical characteristics of rat ovaries and endometrium in experimental hyper- and hypothyroidism. Folia Histochem Cytobiol. 2012;50(3):331–9.

    Article  CAS  PubMed  Google Scholar 

  62. Stadler K, Handler J, Schoenkypl S, Walter I. A three-dimensional culture model of canine uterine glands. In Vitro Cell Dev Biol Anim. 2009;45(1–2):35–43.

    Article  CAS  PubMed  Google Scholar 

  63. Leitner M, Aurich JE, Galabova G, Aurich C, Walter I. Lectin binding patterns in normal canine endometrium and in bitches with pyometra and cystic endometrial hyperplasia. Histol Histopathol. 2003;18(3):787–95.

    CAS  PubMed  Google Scholar 

  64. Sandoval C, Fisher PJ, Schlafer DH. Characterization of trophoblast cell populations by lectin histochemistry in canine placenta during development. J Reprod Fertil Suppl. 2001;57:199–206.

    CAS  PubMed  Google Scholar 

  65. Haines DM. Peanut agglutinin lectin immunohistochemical staining of normal and neoplastic canine tissues. Vet Pathol. 1993;30(4):333–42.

    Article  CAS  PubMed  Google Scholar 

  66. Charalambous C, Drakou K, Nicolaou S, Georgiades P. Novel spatiotemporal glycome changes in the murine placenta during placentation based on BS-I lectin binding patterns. Anat Rec (Hoboken). 2013;296(6):921–32.

    Article  CAS  Google Scholar 

  67. Yurewicz EC, Moghissi KS. Purification of human midcycle cervical mucin and characterization of its oligosaccharides with respect to size, composition, and microheterogeneity. J Biol Chem. 1981;256(22):11895–904.

    CAS  PubMed  Google Scholar 

  68. Yurewicz EC, Matsuura F, Moghissi KS. Structural characterization of neutral oligosaccharides of human midcycle cervical mucin. J Biol Chem. 1982;257(5):2314–22.

    CAS  PubMed  Google Scholar 

  69. Yurewicz EC, Matsuura F, Moghissi KS. Structural studies of sialylated oligosaccharides of human midcycle cervical mucin. J Biol Chem. 1987;262(10):4733–9.

    CAS  PubMed  Google Scholar 

  70. Andersch-Bjorkman Y, Thomsson KA, Holmen Larsson JM, Ekerhovd E, Hansson GC. Large scale identification of proteins, mucins, and their O-glycosylation in the endocervical mucus during the menstrual cycle. Mol Cell Proteomics. 2007;6(4):708–16.

    Article  PubMed  CAS  Google Scholar 

  71. Bohn H, Kraus W, Winckler W. New soluble placental tissue proteins: their isolation, characterization, localization and quantification. Placenta Suppl. 1982;4:67–81.

    CAS  PubMed  Google Scholar 

  72. Oehninger S, Coddington CC, Hodgen GD, Seppala M. Factors affecting fertilization: endometrial placental protein 14 reduces the capacity of human spermatozoa to bind to the human zona pellucida. Fertil Steril. 1995;63(2):377–83.

    Article  CAS  PubMed  Google Scholar 

  73. Seppala M, Koistinen H, Koistinen R, Chiu PC, Yeung WS. Glycosylation related actions of glycodelin: gamete, cumulus cell, immune cell and clinical associations. Hum Reprod Update. 2007;13(3):275–87.

    Article  CAS  PubMed  Google Scholar 

  74. Clark GF, Dell A, Morris HR, Patankar M, Oehninger S, Seppala M. Viewing AIDS from a glycobiological perspective: potential linkages to the human fetoembryonic defence system hypothesis. Mol Hum Reprod. 1997;3(1):5–13.

    Article  CAS  PubMed  Google Scholar 

  75. Dell A, Morris HR, Easton RL, Panico M, Patankar M, Oehniger S, et al. Structural analysis of the oligosaccharides derived from glycodelin, a human glycoprotein with potent immunosuppressive and contraceptive activities. J Biol Chem. 1995;270(41):24116–26.

    Article  CAS  PubMed  Google Scholar 

  76. Lee CL, Pang PC, Yeung WS, Tissot B, Panico M, Lao TT, et al. Effects of differential glycosylation of glycodelins on lymphocyte survival. J Biol Chem. 2009;284(22):15084–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Dutt A, Tang JP, Welply JK, Carson DD. Regulation of N-linked glycoprotein assembly in uteri by steroid hormones. Endocrinology. 1986;118(2):661–73.

    Article  CAS  PubMed  Google Scholar 

  78. Dutt A, Tang JP, Carson DD. Lactosaminoglycans are involved in uterine epithelial cell adhesion in vitro. Dev Biol. 1987;119(1):27–37.

    Article  CAS  PubMed  Google Scholar 

  79. Dutt A, Tang JP, Carson DD. Estrogen preferentially stimulates lactosaminoglycan-containing oligosaccharide synthesis in mouse uteri. J Biol Chem. 1988;263(5):2270–9.

    CAS  PubMed  Google Scholar 

  80. Kuo CW, Chen CM, Lee YC, Chu ST, Khoo KH. Glycomics and proteomics analyses of mouse uterine luminal fluid revealed a predominance of Lewis Y and X epitopes on specific protein carriers. Mol Cell Proteomics. 2009;8(2):325–42.

    Article  CAS  PubMed  Google Scholar 

  81. Levesque E, Beaulieu M, Green MD, Tephly TR, Belanger A, Hum DW. Isolation and characterization of UGT2B15(Y85): a UDP-glucuronosyltransferase encoded by a polymorphic gene. Pharmacogenetics. 1997;7(4):317–25.

    Article  CAS  PubMed  Google Scholar 

  82. Kubushiro K, Ma J, Fukuchi T, Banno K, Muramatsu Y, Tsukazaki K, et al. Changes of β-1,4-galactosyltransferase with the development of endometrial cancer. Gynecol Obstet Invest. 1999;48(3):211–4.

    Article  CAS  PubMed  Google Scholar 

  83. Domino SE, Hiraiwa N, Lowe JB. Molecular cloning, chromosomal assignment and tissue-specific expression of a murine alpha(1,2)fucosyltransferase expressed in thymic and epididymal epithelial cells. Biochem J. 1997;327(Pt 1):105–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Ten Hagen KG, Bedi GS, Tetaert D, Kingsley PD, Hagen FK, Balys MM, et al. Cloning and characterization of a ninth member of the UDP-GalNAc:polypeptide N-acetylgalactosaminyltransferase family, ppGaNTase-T9. J Biol Chem. 2001;276(20):17395–404.

    Article  PubMed  Google Scholar 

  85. Nakamura H, Jasper MJ, Hull ML, Aplin JD, Robertson SA. Macrophages regulate expression of α1,2-fucosyltransferase genes in human endometrial epithelial cells. Mol Hum Reprod. 2012;18(4):204–15.

    Article  CAS  PubMed  Google Scholar 

  86. Domino SE, Hurd EA. LacZ expression in Fut2-LacZ reporter mice reveals estrogen-regulated endocervical glandular expression during estrous cycle, hormone replacement, and pregnancy. Glycobiology. 2004;14(2):169–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Uchiyama T, Sakuta T, Kanayama T. Regulation of hyaluronan synthases in mouse uterine cervix. Biochem Biophys Res Commun. 2005;327(3):927–32.

    Article  CAS  PubMed  Google Scholar 

  88. Li PT, Liao CJ, Wu WG, Yu LC, Chu ST. Progesterone-regulated B4galnt2 expression is a requirement for embryo implantation in mice. Fertil Steril. 2011;95(7):2404–9. 9 e1-3.

    Article  CAS  PubMed  Google Scholar 

  89. Akgul Y, Holt R, Mummert M, Word A, Mahendroo M. Dynamic changes in cervical glycosaminoglycan composition during normal pregnancy and preterm birth. Endocrinology. 2012;153(7):3493–503.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Hey NA, Aplin JD. Sialyl-Lewis x and Sialyl-Lewis a are associated with MUC1 in human endometrium. Glycoconj J. 1996;13(5):769–79.

    Article  CAS  PubMed  Google Scholar 

  91. Genbacev OD, Prakobphol A, Foulk RA, Krtolica AR, Ilic D, Singer MS, et al. Trophoblast L-selectin-mediated adhesion at the maternal-fetal interface. Science. 2003;299(5605):405–8.

    Article  CAS  PubMed  Google Scholar 

  92. Carson DD, Julian J, Lessey BA, Prakobphol A, Fisher SJ. MUC1 is a scaffold for selectin ligands in the human uterus. Front Biosci. 2006;11:2903–8.

    Article  CAS  PubMed  Google Scholar 

  93. Lindenberg S, Sundberg K, Kimber SJ, Lundblad A. The milk oligosaccharide, lacto-N-fucopentaose I, inhibits attachment of mouse blastocysts on endometrial monolayers. J Reprod Fertil. 1988;83(1):149–58.

    Article  CAS  PubMed  Google Scholar 

  94. Kimber SJ, Lindenberg S, Lundblad A. Distribution of some Gal beta 1-3(4)GlcNAc related carbohydrate antigens on the mouse uterine epithelium in relation to the peri- implantational period. J Reprod Immunol. 1988;12(4):297–313.

    Article  CAS  PubMed  Google Scholar 

  95. White S, Kimber SJ. Changes in alpha (1–2)-fucosyltransferase activity in the murine endometrial epithelium during the estrous cycle, early pregnancy, and after ovariectomy and hormone replacement. Biol Reprod. 1994;50(1):73–81.

    Article  CAS  PubMed  Google Scholar 

  96. Zhu ZM, Kojima N, Stroud MR, Hakomori S, Fenderson BA. Monoclonal antibody directed to Le(y) oligosaccharide inhibits implantation in the mouse. Biol Reprod. 1995;52(4):903–12.

    Article  CAS  PubMed  Google Scholar 

  97. Kasai K, Hirabayashi J. Galectins: a family of animal lectins that decipher glycocodes. J Biochem. 1996;119(1):1–8.

    Article  CAS  PubMed  Google Scholar 

  98. Hirabayashi J, Hashidate T, Arata Y, Nishi N, Nakamura T, Hirashima M, et al. Oligosaccharide specificity of galectins: a search by frontal affinity chromatography. Biochim Biophys Acta. 2002;1572(2–3):232–54.

    Article  CAS  PubMed  Google Scholar 

  99. Jones CJ, Carter AM, Aplin JD, Enders AC. Glycosylation at the fetomaternal interface in hemomonochorial placentae from five widely separated species of mammal: is there evidence for convergent evolution? Cells Tissues Organs. 2007;185(4):269–84.

    Article  CAS  PubMed  Google Scholar 

  100. Tatsuzuki A, Ezaki T, Makino Y, Matsuda Y, Ohta H. Characterization of the sugar chain expression of normal term human placental villi using lectin histochemistry combined with immunohistochemistry. Arch Histol Cytol. 2009;72(1):35–49.

    Article  PubMed  Google Scholar 

  101. Drake PM, Gunn MD, Charo IF, Tsou CL, Zhou Y, Huang L, et al. Human placental cytotrophoblasts attract monocytes and CD56(bright) natural killer cells via the actions of monocyte inflammatory protein 1alpha. J Exp Med. 2001;193(10):1199–212.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Juch H, Blaschitz A, Dohr G, Hutter H. HLA class I expression in the human placenta. Wien Med Wochenschr. 2012;162(9–10):196–200.

    Article  PubMed  Google Scholar 

  103. Hutter H, Hammer A, Blaschitz A, Hartmann M, Ebbesen P, Dohr G, et al. Expression of HLA class I molecules in human first trimester and term placenta trophoblast. Cell Tissue Res. 1996;286(3):439–47.

    Article  CAS  PubMed  Google Scholar 

  104. Ljunggren HG, Karre K. In search of the ‘missing self’: MHC molecules and NK cell recognition. Immunol Today. 1990;11(7):237–44.

    Article  CAS  PubMed  Google Scholar 

  105. Long EO, Kim HS, Liu D, Peterson ME, Rajagopalan S. Controlling natural killer cell responses: integration of signals for activation and inhibition. Annu Rev Immunol. 2013;31:227–58.

    Article  CAS  PubMed  Google Scholar 

  106. Handwerger S. New insights into the regulation of human cytotrophoblast cell differentiation. Mol Cell Endocrinol. 2010;323(1):94–104.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Romero R, Espinoza J, Kusanovic JP, Gotsch F, Hassan S, Erez O, et al. The preterm parturition syndrome. BJOG. 2005;113(Suppl 3):17-42.

  108. Jeffcoate TN. Pre-eclampsia and eclampsia: the disease of theories. Proc R Soc Med. 1966;59(5):397–404.

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Walker JJ. Pre-eclampsia. Lancet. 2000;356(9237):1260–5.

    Article  CAS  PubMed  Google Scholar 

  110. Ilekis JV, Reddy UM, Roberts JM. Preeclampsia--a pressing problem: an executive summary of a National Institute of Child Health and Human Development workshop. Reprod Sci. 2007;14(6):508–23.

    Article  CAS  PubMed  Google Scholar 

  111. Pennington KA, Schlitt JM, Jackson DL, Schulz LC, Schust DJ. Preeclampsia: multiple approaches for a multifactorial disease. Dis Model Mech. 2012;5(1):9–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Marini M, Bonaccini L, Thyrion GD, Vichi D, Parretti E, Sgambati E. Distribution of sugar residues in human placentas from pregnancies complicated by hypertensive disorders. Acta Histochem. 2011;113(8):815–25.

    Article  CAS  PubMed  Google Scholar 

  113. Zhang M, Wang M, Gao R, Liu X, Chen X, Geng Y, et al. Altered β1,6-GlcNAc and bisecting GlcNAc-branched N-glycan on integrin β1 are associated with early spontaneous miscarriage in humans. Hum Reprod. 2015;30(9):2064–75.

    Article  PubMed  Google Scholar 

  114. Jones CJ, Wooding FB, Mathias SS, Allen WR. Fetomaternal glycosylation of early placentation events in the African elephant Loxodonta africana. Placenta. 2004;25(4):308–20.

    Article  CAS  PubMed  Google Scholar 

  115. Jones CJ, Abd-Elnaeim M, Bevilacqua E, Oliveira LV, Leiser R. Comparison of uteroplacental glycosylation in the camel (Camelus dromedarius) and alpaca (Lama pacos). Reproduction. 2002;123(1):115–26.

    Article  CAS  PubMed  Google Scholar 

  116. Jones CJ, Koob B, Stoddart RW, Hoffmann B, Leiser R. Lectin-histochemical analysis of glycans in ovine and bovine near-term placental binucleate cells. Cell Tissue Res. 1994;278(3):601–10.

    Article  CAS  PubMed  Google Scholar 

  117. Pijnenborg R, Vercruysse L, Carter AM. Deep trophoblast invasion and spiral artery remodelling in the placental bed of the chimpanzee. Placenta. 2011;32(5):400–8.

    Article  CAS  PubMed  Google Scholar 

  118. Klisch K, Wooding FB, Jones CJ. The glycosylation pattern of secretory granules in binucleate trophoblast cells is highly conserved in ruminants. Placenta. 2010;31(1):11–7.

    Article  CAS  PubMed  Google Scholar 

  119. Jones CJ, Santos TC, Abd-Elnaeim M, Dantzer V, Miglino MA. Placental glycosylation in peccary species and its relation to that of swine and dromedary. Placenta. 2004;25(7):649–57.

    Article  CAS  PubMed  Google Scholar 

  120. Enders AC, Jones CJ, Taylor PJ, Carter AM. Placentation in the Egyptian slit-faced bat Nycteris thebaica (Chiroptera: Nycteridae). Placenta. 2009;30(9):792–9.

    Article  CAS  PubMed  Google Scholar 

  121. Jones CJ, Wilsher SA, Wooding FB, Benirschke K, Allen WR. The binucleate cell of Okapi and Giraffe placenta shows distinctive glycosylation compared with other ruminants: A lectin histochemical study. Mol Phylogenet Evol. 2015;83:184–90.

    Article  CAS  PubMed  Google Scholar 

  122. Jones CJ, Carter AM, Bennett NC, Blankenship TN, Enders AC. Placentation in the Hottentot golden mole, Amblysomus hottentotus (Afrosoricida: Chrysochloridae). Placenta. 2009;30(7):571–8.

    Article  CAS  PubMed  Google Scholar 

  123. Jones CJ, Enders AC, Wooding FP, Dantzer V, Leiser R, Stoddart RW. Equine placental cup cells show glycan expression distinct from that of both chorionic girdle progenitor cells and early allantochorionic trophoblast of the placenta. Placenta. 1999;20(4):347–60.

    Article  CAS  PubMed  Google Scholar 

  124. Jones CJ, Wooding FB, Dantzer V, Leiser R, Stoddart RW. A lectin binding analysis of glycosylation patterns during development of the equine placenta. Placenta. 1999;20(1):45–57.

    Article  CAS  PubMed  Google Scholar 

  125. Pijnenborg R, Vercruysse L, Carter AM. Deep trophoblast invasion and spiral artery remodelling in the placental bed of the lowland gorilla. Placenta. 2011;32(8):586–91.

    Article  CAS  PubMed  Google Scholar 

  126. Jones CJ, Backlin BM, Stoddart RW, Dantzer V. Environmental pollutants as aetiological agents in female reproductive pathology: placental glycan expression in normal and polychlorinated biphenyl (PCB)-exposed mink (Mustela vison). Placenta. 1997;18(8):689–99.

    Article  CAS  PubMed  Google Scholar 

  127. Charalambous F, Elia A, Georgiades P. Decidual spiral artery remodeling during early post-implantation period in mice: investigation of associations with decidual uNK cells and invasive trophoblast. Biochem Biophys Res Commun. 2012;417(2):847–52.

    Article  CAS  PubMed  Google Scholar 

  128. Carvalho AF, Klisch K, Miglino MA, Pereira FT, Bevilacqua E. Binucleate trophoblast giant cells in the water buffalo (Bubalus bubalis) placenta. J Morphol. 2006;267(1):50–6.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The author thanks Dr. Danny Schust for his help in reviewing this manuscript. The author also thanks Julie Schultz and Samantha Belvin for their editorial assistance. Support for the author’s research program has been obtained from the Life Sciences Mission Enhancement Reproductive Biology Program funded by the State of Missouri.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Gary F. Clark.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors’ contribution

GFC conceived of the review and drafted the manuscript.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Clark, G.F. Functional glycosylation in the human and mammalian uterus. Fertil Res and Pract 1, 17 (2015). https://doi.org/10.1186/s40738-015-0007-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40738-015-0007-0

Keywords