Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Reproduction ; 163(2): R11-R23, 2022 02 01.
Article in English | MEDLINE | ID: mdl-35007205

ABSTRACT

Pregnancy-specific glycoproteins (PSGs) are members of the immunoglobulin superfamily and are closely related to the predominantly membrane-bound CEACAM proteins. PSGs are produced by placental trophoblasts and secreted into the maternal bloodstream at high levels where they may regulate maternal immune and vascular functions through receptor binding and modulation of cytokine and chemokine expression and activity. PSGs may have autocrine and paracrine functions in the placental bed, and PSGs can activate soluble and extracellular matrix bound TGF-ß, with potentially diverse effects on multiple cell types. PSGs are also found at high levels in the maternal circulation, at least in human, where they may have endocrine functions. In a non-reproductive context, PSGs are expressed in the gastrointestinal tract and their deregulation may be associated with colorectal cancer and other diseases. Like many placental hormones, PSGs are encoded by multigene families and they have an unusual phylogenetic distribution, being found predominantly in species with hemochorial placentation, with the notable exception of the horse in which PSG-like proteins are expressed in the endometrial cups of the epitheliochorial placenta. The evolution and expansion of PSG gene families appear to be a highly active process, with significant changes in gene numbers and protein domain structures in different mammalian lineages and reports of extensive copy number variation at the human locus. Against this apparent diversification, the available evidence indicates extensive conservation of PSG functions in multiple species. These observations are consistent with maternal-fetal conflict underpinning the evolution of PSGs.


Subject(s)
DNA Copy Number Variations , Placenta , Animals , Female , Glycoproteins/metabolism , Horses , Mammals/metabolism , Phylogeny , Placenta/metabolism , Placentation , Pregnancy , Trophoblasts/metabolism
2.
Glycobiology ; 30(11): 895-909, 2020 10 21.
Article in English | MEDLINE | ID: mdl-32280962

ABSTRACT

Pregnancy-specific beta 1 glycoprotein (PSG1) is secreted from trophoblast cells of the human placenta in increasing concentrations as pregnancy progresses, becoming one of the most abundant proteins in maternal serum in the third trimester. PSG1 has seven potential N-linked glycosylation sites across its four domains. We carried out glycomic and glycoproteomic studies to characterize the glycan composition of PSG1 purified from serum of pregnant women and identified the presence of complex N-glycans containing poly LacNAc epitopes with α2,3 sialyation at four sites. Using different techniques, we explored whether PSG1 can bind to galectin-1 (Gal-1) as these two proteins were previously shown to participate in processes required for a successful pregnancy. We confirmed that PSG1 binds to Gal-1 in a carbohydrate-dependent manner with an affinity of the interaction of 0.13 µM. In addition, we determined that out of the three N-glycosylation-carrying domains, only the N and A2 domains of recombinant PSG1 interact with Gal-1. Lastly, we observed that the interaction between PSG1 and Gal-1 protects this lectin from oxidative inactivation and that PSG1 competes the ability of Gal-1 to bind to some but not all of its glycoprotein ligands.


Subject(s)
Galectin 1/metabolism , Polysaccharides/metabolism , Pregnancy-Specific beta 1-Glycoproteins/metabolism , Female , Galectin 1/chemistry , Humans , Ligands , Polysaccharides/chemistry , Pregnancy , Pregnancy-Specific beta 1-Glycoproteins/chemistry , Pregnancy-Specific beta 1-Glycoproteins/isolation & purification
3.
Reproduction ; 160(5): 685-694, 2020 11.
Article in English | MEDLINE | ID: mdl-33065543

ABSTRACT

In early equine pregnancy, a highly invasive trophoblast cell subpopulation, the chorionic girdle cells, invade the endometrium and form endometrial cups (EC). These cells express classical MHC molecules, thereby stimulating a humoral and cellular immune response, resulting in a massive accumulation of maternal CD4+ and CD8+ T cells around the EC. Nevertheless, no immediate destruction of endometrial cups by maternal lymphoid cells occurs, presumably due to immune tolerance. Although the environment of EC is rich in TGFB and in FOXP3+, CD4+ T cells, the mechanisms leading to tolerance have not been elucidated. Recently, we discovered that equine trophoblast cells secrete pregnancy-specific glycoproteins (PSGs). Since human and murine PSGs activate latent TGFB, we hypothesized that equine PSGs may have a similar activity. We performed plasmon surface resonance experiments to show that equine PSG CEACAM49 can directly bind to the latency-associated peptide (LAP) of both TGFB1 and TGFB2. We then found that the binding of CEACAM49 leads to the activation of TGFB1 as determined by both ELISA and cell-based assays. Furthermore, the activation of TGFB is a unique function of PSGs within the human CEA family, because CEACAM1, 3, 5, 6, 8 do not activate this cytokine. This finding further strengthens the classification of CEACAM49 as an equine PSG. Based on our results, we hypothesize that activation of latent TGFB in the EC environment by equine PSGs secreted by invasive trophoblast cells, could contribute to the generation of regulatory T cells (Tregs) to maintain immune tolerance.


Subject(s)
Carcinoembryonic Antigen/metabolism , Endometrium/metabolism , Glycoproteins/metabolism , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta1/metabolism , Trophoblasts/metabolism , Animals , Endometrium/immunology , Endometrium/pathology , Female , Horses , Pregnancy , Transforming Growth Factor beta1/genetics , Trophoblasts/immunology , Trophoblasts/pathology
4.
Reproduction ; 160(5): 737-750, 2020 11.
Article in English | MEDLINE | ID: mdl-33065549

ABSTRACT

We previously reported that binding to heparan sulfate (HS) is required for the ability of the placentally secreted pregnancy-specific glycoprotein 1 (PSG1) to induce endothelial tubulogenesis. PSG1 is composed of four immunoglobulin-like domains but which domains of the protein bind to HS remains unknown. To analyze the interaction of PSG1 with HS, we generated several recombinant proteins, including the individual domains, chimeric proteins between two PSG1 domains, and mutants. Using flow cytometric and surface plasmon resonance studies, we determined that the B2 domain of PSG1 binds to HS and that the positively charged amino acids encompassed between amino acids 43-59 are required for this interaction. Furthermore, we showed that the B2 domain of PSG1 is required for the increase in the formation of tubes by endothelial cells (EC) including a human endometrial EC line and two extravillous trophoblast (EVT) cell lines and for the pro-angiogenic activity of PSG1 observed in an aortic ring assay. PSG1 enhanced the migration of ECs while it increased the expression of matrix metalloproteinase-2 in EVTs, indicating that the pro-angiogenic effect of PSG1 on these two cell types may be mediated by different mechanisms. Despite differences in amino acid sequence, we observed that all human PSGs bound to HS proteoglycans and confirmed that at least two other members of the family, PSG6 and PSG9, induce tube formation. These findings contribute to a better understanding of the pro-angiogenic activity of human PSGs and strongly suggest conservation of this function among all PSG family members.


Subject(s)
Angiogenesis Inducing Agents/metabolism , Endothelial Cells/metabolism , Glycoproteins/metabolism , Neovascularization, Physiologic , Placenta/metabolism , Pregnancy Proteins/metabolism , Trophoblasts/metabolism , Endothelial Cells/cytology , Female , Glycoproteins/genetics , Humans , Placenta/cytology , Pregnancy , Pregnancy Proteins/genetics , Pregnancy-Specific beta 1-Glycoproteins/metabolism , Trophoblasts/cytology
5.
Biol Blood Marrow Transplant ; 25(2): 193-203, 2019 02.
Article in English | MEDLINE | ID: mdl-30253241

ABSTRACT

Acute graft-versus-host disease (aGVHD) is an immune-mediated reaction that can occur after hematopoietic stem cell transplantation in which donor T cells recognize the host antigens as foreign, destroying host tissues. Establishment of a tolerogenic immune environment while preserving the immune response to infectious agents is required for successful bone marrow transplantation. Pregnancy-specific glycoprotein 1 (PSG1), which is secreted by the human placenta into the maternal circulation throughout pregnancy, likely plays a role in maintaining immunotolerance to prevent rejection of the fetus by the maternal immune system. We have previously shown that PSG1 activates the latent form of transforming growth factor ß1 (TGF-ß), a cytokine essential for the differentiation of tolerance-inducing CD4+FoxP3+ regulatory T cells (Tregs). Consistent with this observation, treatment of naïve murine T cells with PSG1 resulted in a significant increase in FoxP3+ cells that was blocked by a TGF-ß receptor I inhibitor. We also show here that PSG1 can increase the availability of active TGF-ß in vivo. As the role of CD4+FoxP3+ cells in the prevention of aGVHD is well established, we tested whether PSG1 has beneficial effects in a murine aGHVD transplantation model. PSG1-treated mice had reduced numbers of tissue-infiltrating inflammatory CD3+ T cells and had increased expression of FoxP3 in T cells compared with vehicle-treated mice. In addition, administration of PSG1 significantly inhibited aGVHD-associated weight loss and mortality. On the other hand, administration of PSG1 was less effective in managing aGVHD in the presence of an alloimmune reaction against a malignancy in a graft-versus-leukemia experimental model. Combined, this data strongly suggests that PSG1 could be a promising treatment option for patients with aGVHD following bone marrow transplantation for a nonmalignant condition, such as an autoimmune disorder or a genetic immunodeficiency.


Subject(s)
Bone Marrow Transplantation , Graft vs Host Disease , Pregnancy-Specific beta 1-Glycoproteins/pharmacology , Acute Disease , Animals , Disease Models, Animal , Graft vs Host Disease/genetics , Graft vs Host Disease/metabolism , Graft vs Host Disease/pathology , Graft vs Host Disease/prevention & control , Humans , Mice , Mice, Transgenic , Recombinant Proteins/pharmacology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Transplantation, Homologous
6.
Mol Hum Reprod ; 24(12): 602-612, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30371828

ABSTRACT

STUDY QUESTION: Do all 10 human pregnancy-specific beta 1-glycoproteins (PSGs) and murine PSG23 activate latent transforming growth factor-ß1 (TGF-ß1)? SUMMARY ANSWER: All human PSGs and murine PSG23 activated latent TGF-ß1. WHAT IS KNOWN ALREADY: Two of the 10 members of the PSG1 family, PSG1 and PSG9, were previously shown to activate the soluble small latent complex of TGF-ß1, a cytokine with potent immune suppressive functions. STUDY DESIGN, SIZE, DURATION: Recombinant PSGs were generated and tested for their ability to activate the small latent complex of TGF-ß1 in a cell-free ELISA-based assay and in a bioassay. In addition, we tested the ability of PSG1 and PSG4 to activate latent TGF-ß bound to the extracellular matrix (ECM) or on the membranes of the Jurkat human T-cell line. PARTICIPANTS/MATERIALS, SETTING, METHODS: Recombinant PSGs were generated by transient transfection and purified with a His-Trap column followed by gel filtration chromatography. The purified PSGs were compared to vehicle (PBS) used as control for their ability to activate the small latent complex of TGF-ß1. The concentration of active TGF-ß was measured in an ELISA using the TGF-ß receptor II as capture and a bioassay using transformed mink epithelial cells that express luciferase in response to active TGF-ß. The specificity of the signal was confirmed using a TGF-ß receptor inhibitor. We also measured the binding kinetics of some human PSGs for the latent-associated peptide (LAP) of TGF-ß using surface plasmon resonance and determined whether PSG1 and PSG4 could activate the large latent complex of TGF-ß1 bound to the ECM and latent TGF-ß1 bound to the cell membrane. All experiments were performed in triplicate wells and repeated three times. MAIN RESULTS AND THE ROLE OF CHANCE: All human PSGs activated the small latent complex of TGF-ß1 (P < 0.05 vs. control) and showed similar affinities (KD) for LAP. Despite the lack of sequence conservation with its human counterparts, the ability to activate latent TGF-ß1 was shared by a member of the murine PSG family. We found that PSG1 and PSG4 activated the latent TGF-ß stored in the ECM (P < 0.01) but did not activate latent TGF-ß1 bound to glycoprotein A repetitions predominant (GARP) on the surface of Jurkat T cells. LIMITATIONS, REASONS FOR CAUTION: The affinity of the interaction of LAP and PSGs was calculated using recombinant proteins, which may differ from the native proteins in their post-translational modifications. We also utilized a truncated form of murine PSG23 rather than the full-length protein. For the studies testing the ability of PSGs to activate membrane-bound TGF-ß1, we utilized the T-cell line Jurkat and Jurkat cells expressing GARP rather than primary T regulatory cells. All the studies were performed in vitro. WIDER IMPLICATIONS OF THE FINDINGS: Here, we show that all human PSGs activate TGF-ß1 and that this function is conserved in at least one member of the rodent PSG family. In vivo PSGs could potentially increase the availability of active TGF-ß1 from the soluble and matrix-bound latent forms of the cytokine contributing to the establishment of a tolerogenic environment during pregnancy. LARGE-SCALE DATA: None. STUDY FUNDING/COMPETING INTEREST(S): The research was supported by a grant from the Collaborative Health Initiative Research Program (CHIRP). No conflicts of interests are declared by the authors.


Subject(s)
Pregnancy-Specific beta 1-Glycoproteins/metabolism , Transforming Growth Factor beta1/metabolism , Enzyme-Linked Immunosorbent Assay , Extracellular Matrix/metabolism , Female , Heparitin Sulfate , Humans , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Pregnancy , Pregnancy-Specific beta 1-Glycoproteins/genetics , Transforming Growth Factor beta1/genetics
7.
J Biol Chem ; 291(36): 18700-17, 2016 09 02.
Article in English | MEDLINE | ID: mdl-27405760

ABSTRACT

The mannose receptor (ManR, Mrc1) and asialoglycoprotein receptor (ASGR, Asgr1 and Asgr2) are highly abundant endocytic receptors expressed by sinusoidal endothelial cells and parenchymal cells in the liver, respectively. We genetically manipulated either receptor individually or in combination, revealing phenotypic changes in female and male mice associated with changes in circulating levels of many glycoproteins. Both receptors rise and fall in response to progesterone during pregnancy. Thirty percent of Asgr2(-/-) and 65% of Mrc1(-/-)Asgr2(-/-) mice are unable to initiate parturition at the end of pregnancy, whereas Mrc1(-/-) mice initiate normally. Twenty five percent of Mrc1(-/-)Asgr2(-/-) male mice develop priapism when mating due to thrombosis of the penile vein, but neither Mrc1(-/-) nor Asgr2(-/-) mice do so. The half-life for luteinizing hormone (LH) clearance increases in Mrc1(-/-) and Mrc1(-/-)Asgr2(-/-) mice but not in Asgr2(-/-) mice; however, LH and testosterone are elevated in all three knockouts. The ManR clears LH thus regulating testosterone production, whereas the ASGR appears to mediate clearance of an unidentified glycoprotein that increases LH levels. More than 40 circulating glycoproteins are elevated >3.0-fold in pregnant Mrc1(-/-)Asgr2(-/-) mice. Pregnancy-specific glycoprotein 23, undetectable in WT mice (<50 ng/ml plasma), reaches levels of 1-10 mg/ml in the plasma of Mrc1(-/-)Asgr2(-/-) and Asgr2(-/-) mice, indicating it is cleared by the ASGR. Elevation of multiple coagulation factors in Mrc1(-/-)Asgr2(-/-) mice may account for priapism seen in males. These male and female phenotypic changes underscore the key roles of the ManR and ASGR in controlling circulating levels of numerous glycoproteins critical for regulating reproductive hormones and blood coagulation.


Subject(s)
Asialoglycoprotein Receptor/metabolism , Membrane Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , Animals , Asialoglycoprotein Receptor/genetics , Blood Coagulation/genetics , Female , Glycoproteins/blood , Glycoproteins/genetics , Luteinizing Hormone/blood , Luteinizing Hormone/genetics , Male , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Parturition/blood , Parturition/genetics , Pregnancy , Priapism/blood , Priapism/genetics , Priapism/pathology , Receptors, Cell Surface/genetics , Receptors, Immunologic , Testosterone/blood , Testosterone/genetics , Venous Thrombosis/blood , Venous Thrombosis/genetics , Venous Thrombosis/pathology
8.
J Biol Chem ; 290(7): 4422-31, 2015 Feb 13.
Article in English | MEDLINE | ID: mdl-25548275

ABSTRACT

Pregnancy-specific glycoproteins (PSGs) are a family of Ig-like proteins secreted by specialized placental cells. The PSG1 structure is composed of a single Ig variable region-like N-terminal domain and three Ig constant region-like domains termed A1, A2, and B2. Members of the human and murine PSG family have been shown to induce anti-inflammatory cytokines from monocytes and macrophages and to stimulate angiogenesis. We recently showed that recombinant forms of PSG1 (PSG1-Fc and PSG1-His) and PSG1 purified from the serum of pregnant women are associated with the immunoregulatory cytokine TGF-ß1 and activated latent TGF-ß1. Here, we sought to examine the requirement of specific PSG1 domains in the activation of latent TGF-ß1. Plasmon surface resonance studies showed that PSG1 directly bound to the small latent complex and to the latency-associated peptide of TGF-ß1 and that this binding was mediated through the B2 domain. Furthermore, the B2 domain alone was sufficient for activating the small latent complex. In separate experiments, we found that the PSG1-mediated induction of TGF-ß1 secretion in macrophages was dependent on the N-terminal domain. Mutagenesis analysis revealed that four amino acids (LYHY) of the CC' loop of the N-terminal domain were required for induction of latent TGF-ß1 secretion. Together, our results show that two distinct domains of PSG1 are involved in the regulation of TGF-ß1 and provide a mechanistic framework for how PSGs modulate the immunoregulatory environment at the maternal-fetal interface for successful pregnancy outcome.


Subject(s)
Macrophages/metabolism , Monocytes/metabolism , Placenta/metabolism , Pregnancy-Specific beta 1-Glycoproteins/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Blotting, Western , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Female , Humans , Immunoenzyme Techniques , Macrophages/cytology , Mice , Monocytes/cytology , Placenta/cytology , Pregnancy , Pregnancy-Specific beta 1-Glycoproteins/genetics , Protein Conformation , Protein Structure, Tertiary , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta1/genetics
9.
Front Immunol ; 14: 1196395, 2023.
Article in English | MEDLINE | ID: mdl-37475853

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic imposed a risk of infection and disease in pregnant women and neonates. Successful pregnancy requires a fine-tuned regulation of the maternal immune system to accommodate the growing fetus and to protect the mother from infection. Galectins, a family of ß-galactoside-binding proteins, modulate immune and inflammatory processes and have been recognized as critical factors in reproductive orchestration, including maternal immune adaptation in pregnancy. Pregnancy-specific glycoprotein 1 (PSG1) is a recently identified gal-1 ligand at the maternal-fetal interface, which may facilitate a successful pregnancy. Several studies suggest that galectins are involved in the immune response in SARS-CoV-2-infected patients. However, the galectins and PSG1 signature upon SARS-CoV-2 infection and vaccination during pregnancy remain unclear. In the present study, we examined the maternal circulating levels of galectins (gal-1, gal-3, gal-7, and gal-9) and PSG1 in pregnant women infected with SARS-CoV-2 before vaccination or uninfected women who were vaccinated against SARS-CoV-2 and correlated their expression with different pregnancy parameters. SARS-CoV-2 infection or vaccination during pregnancy provoked an increase in maternal gal-1 circulating levels. On the other hand, levels of PSG1 were only augmented upon SARS-CoV-2 infection. A healthy pregnancy is associated with a positive correlation between gal-1 concentrations and gal-3 or gal-9; however, no correlation was observed between these lectins during SARS-CoV-2 infection. Transcriptome analysis of the placenta showed that gal-1, gal-3, and several PSG and glycoenzymes responsible for the synthesis of gal-1-binding glycotopes (such as linkage-specific N-acetyl-glucosaminyltransferases (MGATs)) are upregulated in pregnant women infected with SARS-CoV-2. Collectively, our findings identify a dynamically regulated "galectin-specific signature" that accompanies the SARS-CoV-2 infection and vaccination in pregnancy, and they highlight a potentially significant role for gal-1 as a key pregnancy protective alarmin during virus infection.


Subject(s)
COVID-19 , Placenta , Female , Humans , Infant, Newborn , Pregnancy , Alarmins/metabolism , COVID-19/metabolism , Galectin 1/metabolism , Galectins/metabolism , SARS-CoV-2/metabolism
10.
PNAS Nexus ; 2(8): pgad247, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37575671

ABSTRACT

Placental abnormalities cause impaired fetal growth and poor pregnancy outcome (e.g. preeclampsia [PE]) with long-lasting consequences for the mother and offspring. The molecular dialogue between the maternal niche and the developing placenta is critical for the function of this organ. Galectin-1 (gal-1), a highly expressed glycan-binding protein at the maternal-fetal interface, orchestrates the maternal adaptation to pregnancy and placenta development. Down-regulation or deficiency of gal-1 during pregnancy is associated with the development of PE; however, the maternal- and placental-derived gal-1 contributions to the disease onset are largely unknown. We demonstrate that lack of gal-1 imposes a risk for PE development in a niche-specific manner, and this is accompanied by a placental dysfunction highly influenced by the absence of maternal-derived gal-1. Notably, differential placental glycosylation through the Sda-capped N-glycans dominates the invasive trophoblast capacity triggered by maternal-derived gal-1. Our findings show that gal-1 derived from the maternal niche is essential for healthy placenta development and indicate that impairment of the gal-1 signaling pathway within the maternal niche could be a molecular cause for maternal cardiovascular maladaptation during pregnancy.

11.
J Biol Chem ; 286(9): 7577-86, 2011 Mar 04.
Article in English | MEDLINE | ID: mdl-21193412

ABSTRACT

Pregnancy-specific ß1 glycoproteins (PSGs) are the most abundant fetal proteins in the maternal bloodstream in late pregnancy. They are secreted by the syncytiotrophoblast and are detected around day 14 postfertilization. There are 11 human PSG genes, which encode a family of proteins exhibiting significant conservation at the amino acid level. We and others have proposed that PSGs have an immune modulatory function. In addition, we recently postulated that they are proangiogenic due to their ability to induce the secretion of VEGF-A and the formation of tubes by endothelial cells. The cellular receptor(s) for human PSGs remain unknown. Therefore, we conducted these studies to identify the receptor for PSG1, the highest expressed member of the family. We show that removal of cell surface glycosaminoglycans (GAGs) by enzymatic or chemical treatment of cells or competition with heparin completely inhibited binding of PSG1. In addition, PSG1 did not bind to cells lacking heparan or chondroitin sulfate on their surface, and binding was restored upon transfection with all four syndecans and glypican-1. Importantly, the presence of GAGs on the surface of endothelial cells was required for the ability of PSG1 to induce tube formation. This finding indicates that the PSG1-GAG interaction mediates at least some of the PSG1 proposed functions.


Subject(s)
Chondroitin Sulfates/metabolism , Heparitin Sulfate/metabolism , Pregnancy-Specific beta 1-Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , Trophoblasts/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Endothelial Cells/metabolism , Female , HeLa Cells , Heparin/metabolism , Humans , Jurkat Cells , Mice , NIH 3T3 Cells , Neovascularization, Physiologic/physiology , Pregnancy , Pregnancy-Specific beta 1-Glycoproteins/genetics , Syndecans/metabolism , Transfection , Trophoblasts/cytology
12.
Cell Mol Bioeng ; 15(2): 175-191, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35401843

ABSTRACT

Introduction: Trophoblast invasion is a complex biological process necessary for establishment of pregnancy; however, much remains unknown regarding what signaling factors coordinate the extent of invasion. Pregnancy-specific glycoproteins (PSGs) are some of the most abundant circulating trophoblastic proteins in maternal blood during human pregnancy, with maternal serum concentrations rising to as high as 200-400 µg/mL at term. Methods: Here, we employ three-dimensional (3D) trophoblast motility assays consisting of trophoblast spheroids encapsulated in 3D gelatin hydrogels to quantify trophoblast outgrowth area, viability, and cytotoxicity in the presence of PSG1 and PSG9 as well as epidermal growth factor and Nodal. Results: We show PSG9 reduces trophoblast motility whereas PSG1 increases motility. Further, we assess bulk nascent protein production by encapsulated spheroids to highlight the potential of this approach to assess trophoblast response (motility, remodeling) to soluble factors and extracellular matrix cues. Conclusions: Such models provide an important platform to develop a deeper understanding of early pregnancy.

13.
Viruses ; 14(7)2022 07 20.
Article in English | MEDLINE | ID: mdl-35891553

ABSTRACT

Over the past two years, scientific research has moved at an unprecedented rate in response to the COVID-19 pandemic. The rapid development of effective vaccines and therapeutics would not have been possible without extensive background knowledge on coronaviruses developed over decades by researchers, including Kathryn (Kay) Holmes. Kay's research team discovered the first coronavirus receptors for mouse hepatitis virus and human coronavirus 229E and contributed a wealth of information on coronaviral spike glycoproteins and receptor interactions that are critical determinants of host and tissue specificity. She collaborated with several research laboratories to contribute knowledge in additional areas, including coronaviral pathogenesis, epidemiology, and evolution. Throughout her career, Kay was an extremely dedicated and thoughtful mentor to numerous graduate students and post-doctoral fellows. This article provides a review of her contributions to the coronavirus field and her exemplary mentoring.


Subject(s)
Coronavirus 229E, Human , Receptors, Coronavirus , Animals , COVID-19 , History, 21st Century , Humans , Mice , Pandemics , Spike Glycoprotein, Coronavirus/genetics
14.
Cell Death Dis ; 12(4): 400, 2021 04 14.
Article in English | MEDLINE | ID: mdl-33854046

ABSTRACT

Immune homeostasis depends upon effective clearance of pathogens while simultaneously preventing autoimmunity and immunopathology in the host. Restimulation-induced cell death (RICD) is one such mechanism where by activated T cells receive subsequent antigenic stimulation, reach a critical signal threshold through the T cell receptor (TCR), and commit to apoptosis. Many details of this process remain unclear, including the role of co-stimulatory and co-inhibitory proteins that influence the TCR signaling cascade. Here we characterize the role of T cell immunoglobulin and mucin domain containing 3 (TIM-3) in RICD regulation. TIM-3 protected newly activated CD8+ effector T cells from premature RICD during clonal expansion. Surprisingly, however, we found that TIM-3 potentiated RICD in late-stage effector T cells. The presence of TIM-3 increased proximal TCR signaling and proapoptotic protein expression in late-stage effector T cells, with no consistent signaling effects noted in newly activated cells with or without TIM-3. To better explain these differences in TIM-3 function as T cells aged, we characterized the temporal pattern of TIM-3 expression in effector T cells. We found that TIM-3 was expressed on the surface of newly activated effector T cells, but remained largely intracellular in late-stage effector cells. Consistent with this, TIM-3 required a ligand to prevent early RICD, whereas ligand manipulation had no effects at later stages. Of the known TIM-3 ligands, carcinoembryonic antigen-related cell adhesion molecule (CEACAM1) showed the greatest difference in surface expression over time and also protected newly activated cells from premature RICD, with no measurable effects in late-stage effectors. Indeed, CEACAM1 enabled TIM-3 surface expression on T cells, implying a co-dependency for these proteins in protecting expanding T cells from premature RICD. Our findings suggest that co-signaling proteins like TIM-3 and CEACAM1 can alter RICD sensitivity at different stages of the effector T cell response, with important implications for checkpoint blockade therapy.


Subject(s)
Antigens, CD/metabolism , Apoptosis/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Adhesion Molecules/metabolism , Hepatitis A Virus Cellular Receptor 2/metabolism , Antigens, CD/immunology , Cell Adhesion Molecules/immunology , Humans , Lymphocyte Activation/immunology , Membrane Proteins/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism
15.
Front Immunol ; 12: 784473, 2021.
Article in English | MEDLINE | ID: mdl-34975875

ABSTRACT

Lectin-glycan interactions, in particular those mediated by the galectin family, regulate many processes required for a successful pregnancy. Over the past decades, increasing evidence gathered from in vitro and in vivo experiments indicate that members of the galectin family specifically bind to both intracellular and membrane bound carbohydrate ligands regulating angiogenesis, immune-cell adaptations required to tolerate the fetal semi-allograft and mammalian embryogenesis. Therefore, galectins play important roles in fetal development and placentation contributing to maternal and fetal health. This review discusses the expression and role of galectins during the course of pregnancy, with an emphasis on maternal immune adaptions and galectin-glycan interactions uncovered in the recent years. In addition, we summarize the galectin fingerprints associated with pathological gestation with particular focus on preeclampsia.


Subject(s)
Adaptation, Physiological , Fetal Development/physiology , Galectins/physiology , Placentation/physiology , Female , Galectins/chemistry , Glycoproteins/physiology , Humans , Pregnancy
16.
J Exp Med ; 195(2): 277-82, 2002 Jan 21.
Article in English | MEDLINE | ID: mdl-11805154

ABSTRACT

Pregnancy-specific glycoproteins (PSGs) are a family of highly similar secreted proteins produced by the placenta. PSG homologs have been identified in primates and rodents. Members of the human and murine PSG family induce secretion of antiinflammatory cytokines in mononuclear phagocytes. For the purpose of cloning the receptor, we screened a RAW 264.7 cell cDNA expression library. The PSG17 receptor was identified as the tetraspanin, CD9. We confirmed binding of PSG17 to CD9 by ELISA, flow cytometry, alkaline phosphatase binding assays, and in situ rosetting. Anti-CD9 monoclonal antibody inhibited binding of PSG17 to CD9-transfected cells and RAW 264.7 cells. Moreover, PSG17 binding to macrophages from CD9-deficient mice was significantly reduced. We then tested whether PSG17 binds to other members of the murine tetraspanin family. PSG17 did not bind to cells transfected with CD53, CD63, CD81, CD82, or CD151, suggesting that PSG17-CD9 binding is a specific interaction. We have identified the first receptor for a murine PSG as well as the first natural ligand for a member of the tetraspanin superfamily.


Subject(s)
Antigens, CD/metabolism , Pregnancy Proteins/metabolism , Animals , Antigens, CD/genetics , Cell Line , Cloning, Molecular , DNA, Complementary/analysis , DNA, Complementary/genetics , Female , Humans , Ligands , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Placenta/metabolism , Pregnancy , Protein Binding , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Tetraspanin 29
17.
Biol Reprod ; 83(1): 27-35, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20335639

ABSTRACT

Previous studies suggest that human pregnancy specific beta-1-glycoproteins (PSGs) play immunomodulatory roles during pregnancy; however, other possible functions of PSGs have yet to be explored. We have observed that PSGs induce transforming growth factor beta 1 (TGFB1), which among its other diverse functions inhibits T-cell function and has proangiogenic properties. The present study investigates a potential role for PSG1, the most abundant PSG in maternal serum, as a possible inducer of proangiogenic growth factors known to play an important role in establishment of the vasculature at the maternal-fetal interface. To this end, we measured TGFB1, vascular endothelial growth factors (VEGFs) A and C, and placental growth factor (PGF) protein levels in several cell types after PSG1 treatment. In addition, tube formation and wound healing assays were performed to investigate a possible direct interaction between PSG1 and endothelial cells. PSG1 induced up-regulation of both TGFB1 and VEGFA in human monocytes, macrophages, and two human extravillous trophoblast cell lines. We did not observe induction of VEGFC or PGF by PSG1 in any of the cells tested. PSG1 treatment resulted in endothelial tube formation in the presence and absence of VEGFA. Site-directed mutagenesis was performed to map the essential regions within the N-domain of PSG1 required for functional activity. We found that the aspartic acid at position 95, previously believed to be required for binding of PSGs to cells, is not required for PSG1 activity but that the amino acids implicated in the formation of a salt bridge within the N-domain are essential for PSG1 function.


Subject(s)
Neovascularization, Physiologic , Placenta/metabolism , Pregnancy-Specific beta 1-Glycoproteins/metabolism , Transforming Growth Factor beta1/metabolism , Vascular Endothelial Growth Factor A/metabolism , Endothelial Cells/metabolism , Female , Humans , Macrophages/metabolism , Mutagenesis, Site-Directed , Placenta/blood supply , Placenta Growth Factor , Placentation , Pregnancy , Pregnancy Proteins/metabolism , Pregnancy-Specific beta 1-Glycoproteins/genetics , Recombinant Proteins/metabolism , Trophoblasts/metabolism
18.
Front Immunol ; 10: 1166, 2019.
Article in English | MEDLINE | ID: mdl-31231368

ABSTRACT

Galectins are a phylogenetically conserved family of soluble ß-galactoside binding proteins, consisting of 15 different types, each with a specific function. Galectins contribute to placentation by regulating trophoblast development, migration, and invasion during early pregnancy. In addition, galectins are critical players regulating maternal immune tolerance to the embedded embryo. Recently, the role of galectins in angiogenesis during decidualization and in placenta formation has gained attention. Altered expression of galectins is associated with abnormal pregnancies and infertility. This review focuses on the role of galectins in pregnancy-associated processes and discusses the relevance of galectin-glycan interactions as potential therapeutic targets in pregnancy disorders.


Subject(s)
Galectins/physiology , Polysaccharides/metabolism , Pregnancy Proteins/physiology , Animals , Carbohydrate Sequence , Chromosome Mapping , Dimerization , Embryo, Nonmammalian/metabolism , Embryonic Development/physiology , Endothelial Cells/metabolism , Extracellular Vesicles/metabolism , Female , Galectins/chemistry , Galectins/genetics , Glycosylation , Humans , Maternal-Fetal Exchange/physiology , Neovascularization, Physiologic/physiology , Placentation/physiology , Polysaccharides/chemistry , Pre-Eclampsia/metabolism , Pregnancy , Protein Processing, Post-Translational , Structure-Activity Relationship , Substrate Specificity , Trophoblasts/metabolism
19.
Cells ; 8(11)2019 10 31.
Article in English | MEDLINE | ID: mdl-31683744

ABSTRACT

Human pregnancy-specific glycoproteins (PSGs) serve immunomodulatory and pro-angiogenic functions during pregnancy and are mainly expressed by syncytiotrophoblast cells. While PSG mRNA expression in extravillous trophoblasts (EVTs) was reported, the proteins were not previously detected. By immunohistochemistry and immunoblotting, we show that PSGs are expressed by invasive EVTs and co-localize with integrin 5. In addition, we determined that native and recombinant PSG1, the most highly expressed member of the family, binds to 51 and induces the formation of focal adhesion structures resulting in adhesion of primary EVTs and EVT-like cell lines under 21% oxygen and 1% oxygen conditions. Furthermore, we found that PSG1 can simultaneously bind to heparan sulfate in the extracellular matrix and to 51 on the cell membrane. Wound healing assays and single-cell movement tracking showed that immobilized PSG1 enhances EVT migration. Although PSG1 did not affect EVT invasion in the in vitro assays employed, we found that the serum PSG1 concentration is lower in African-American women diagnosed with early-onset and late-onset preeclampsia, a pregnancy pathology characterized by shallow trophoblast invasion, than in their respective healthy controls only when the fetus was a male; therefore, the reduced expression of this molecule should be considered in the context of preeclampsia as a potential therapy.


Subject(s)
Integrin alpha5beta1/metabolism , Pregnancy-Specific beta 1-Glycoproteins/metabolism , Trophoblasts/metabolism , Cell Adhesion , Cell Line , Cell Membrane/metabolism , Cell Movement , Extracellular Matrix/metabolism , Female , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Heparitin Sulfate/metabolism , Humans , Immobilized Proteins/chemistry , Immobilized Proteins/metabolism , Placenta/metabolism , Pre-Eclampsia/diagnosis , Pregnancy , Pregnancy Trimester, First , Pregnancy-Specific beta 1-Glycoproteins/analysis , Pregnancy-Specific beta 1-Glycoproteins/genetics , Protein Binding , Trophoblasts/cytology
20.
Mol Biol Cell ; 14(12): 5098-103, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14528020

ABSTRACT

The function currently attributed to tetraspanins is to organize molecular complexes in the plasma membrane by using multiple cis-interactions. Additionally, the tetraspanin CD9 may be a receptor that binds the soluble ligand PSG17, a member of the immunoglobulin superfamily (IgSF)/CEA subfamily. However, previous data are also consistent with the PSG17 receptor being a CD9 cis-associated protein. In the current study, CD9 extracellular loop (EC2) specifically bound to PSG17-coated beads, indicating a direct interaction between the two proteins. However, CD9-EC2 did not bind to PSG17-coated beads if the CD9-EC2 had the mutation SFQ (173-175) to AAA, a previously studied mutation in egg CD9 that abolishes sperm-egg fusion. Also, PSG17 bound to 293 T cells transfected with wild-type CD9 but not the mutant CD9. By immunofluorescence, PSG17 bound to wild-type eggs but not to CD9 null eggs. The presence of approximately 2 microM recombinant PSG17 produced a significant and reversible inhibition (60-80%) of sperm-egg fusion. Thus, we conclude that CD9 is a receptor for PSG17 and when the PSG17 binding site is mutated or occupied, sperm-egg fusion is impaired. These findings suggest that egg CD9 may function in gamete fusion by binding to a sperm IgSF/CEA subfamily member and such proteins have previously been identified on sperm.


Subject(s)
Antigens, CD/metabolism , Immunoglobulins/metabolism , Membrane Glycoproteins/metabolism , Oocytes/metabolism , Sperm-Ovum Interactions/physiology , Spermatozoa/metabolism , Animals , Antigens, CD/physiology , Cells, Cultured , Female , Fluorescent Antibody Technique , Humans , Male , Membrane Glycoproteins/physiology , Mice , Mice, Inbred ICR , Mutation , Oocytes/physiology , Protein Binding/physiology , Protein Structure, Tertiary/physiology , Sperm Motility/physiology , Spermatozoa/physiology , Tetraspanin 29
SELECTION OF CITATIONS
SEARCH DETAIL