Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 130
Filter
1.
Scand J Clin Lab Invest ; 83(7): 479-488, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37887078

ABSTRACT

Chronic hypertension is one of the major risk factors for preeclampsia. Pregnancy-specific beta-1-glycoprotein (PSG-1) is a protein that plays a critical role in fetomaternal immune modulation and has been shown to be closely associated with pregnancy adverse events such as preeclampsia. It is also known that PSG-1 and its source placenta are associated with many molecular pathways associated with blood pressure regulation. In addition, the nondipping pattern (NDP) of chronic hypertension has been shown to be an independent risk factor for preeclampsia. Dipper individuals experience a notable nighttime drop in blood pressure, typically around 10% or more compared to daytime levels, while nondipper individuals show a smaller nighttime blood pressure decrease, indicating potential circadian blood pressure regulation disruption. In this context, we aimed to reveal the relationship between PSG-1, NDP and preeclampsia in this study. A total of 304 pregnant women who were newly diagnosed in the first trimester and started on antihypertensive medication were included in this study. All subjects performed 24-h ambulatory blood pressure monitoring twice throughout pregnancy, the first in the 1. trimester to confirm the diagnosis of hypertension and the second between 20+0 and 21+1 gestational weeks to determine the dipper-nondipper status of hypertension. Subjects were grouped as dipper and nondipper according to blood pressure, and groups were compared in terms of PSG-1 levels. In this study, low PSG-1 levels and NDP were independently associated with preeclampsia. Findings from this study suggest that PSG-1 may play an important role in the causal relationship between NDP and preeclampsia.


Subject(s)
Hypertension , Pre-Eclampsia , Female , Humans , Pregnancy , Blood Pressure/physiology , Blood Pressure Monitoring, Ambulatory , Circadian Rhythm/physiology , Glycoproteins , Hypertension/complications , Pre-Eclampsia/diagnosis , Pregnant Women , Pregnancy-Specific beta 1-Glycoproteins/metabolism
2.
PLoS One ; 16(4): e0250031, 2021.
Article in English | MEDLINE | ID: mdl-33857242

ABSTRACT

OBJECTIVE: We sought to identify plasma protein biomarkers that are predictive of the outcome of rescue cerclage in patients with cervical insufficiency. METHODS: This retrospective cohort study included 39 singleton pregnant women undergoing rescue cerclage for cervical insufficiency (17-25 weeks) who gave plasma samples. Three sets of pooled plasma samples from controls (cerclage success, n = 10) and cases (cerclage failure, n = 10, defined as spontaneous preterm delivery at <33 weeks) were labeled with 6-plex tandem mass tag (TMT) reagents and analyzed by liquid chromatography-tandem mass spectrometry. Differentially expressed proteins between the two groups were selected from the TMT-based quantitative analysis. Multiple reaction monitoring-mass spectrometry (MRM-MS) analysis was further used to verify the candidate proteins of interest in patients with cervical insufficiency in the final cohort (n = 39). RESULTS: From MRM-MS analysis of the 40 proteins showing statistically significant changes (P < 0.05) from the TMT-based quantitative analysis, plasma IGFBP-2, PSG4, and PGLYRP2 levels were found to be significantly increased, whereas plasma MET and LXN levels were significantly decreased in women with cerclage failure. Of these, IGFBP-2, PSG4, and LXN levels in plasma were independent of cervical dilatation. A multiple-biomarker panel was developed for the prediction of cerclage failure, using a stepwise regression procedure, which included the plasma IGFBP-2, PSG4, and LXN (area under the curve [AUC] = 0.916). The AUC for this multiple-biomarker panel was significantly greater than the AUC for any single biomarker included in the multi-biomarker model. CONCLUSIONS: Proteomic analysis identified useful and independent plasma biomarkers (IGFBP-2, PSG4, and LXN; verified by MRM) that predict poor pregnancy outcome following rescue cerclage. Their combined analysis in a multi-biomarker panel significantly improved predictability.


Subject(s)
Biomarkers/blood , Cerclage, Cervical/methods , Uterine Cervical Incompetence/surgery , Adult , Carrier Proteins/blood , Female , Humans , Insulin-Like Growth Factor II/metabolism , Nerve Tissue Proteins/blood , Pregnancy-Specific beta 1-Glycoproteins/metabolism , Prognosis , Proteomics , Proto-Oncogene Proteins c-met/blood , Treatment Outcome , Uterine Cervical Incompetence/blood
3.
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
4.
BMC Immunol ; 21(1): 56, 2020 10 30.
Article in English | MEDLINE | ID: mdl-33126863

ABSTRACT

BACKGROUND: Pregnancy-specific ß1-glycoproteins are capable of regulating innate and adaptive immunity, exerting predominantly suppressive effects. In this regard, they are of interest in terms of their pharmacological potential for the treatment of autoimmune diseases and post-transplant complications. The effect of these proteins on the main pro-inflammatory subpopulation of T lymphocytes, IL-17-producing helper T cells (Th17), has not been comprehensively studied. Therefore, the effects of the native pregnancy-specific ß1-glycoprotein on the proliferation, Th17 polarization and cytokine profile of human CD4+ cells were assessed. RESULTS: Native human pregnancy-specific ß1-glycoprotein (PSG) at а concentration of 100 µg/mL was shown to decrease the frequency of Th17 (RORγτ+) in CD4+ cell culture and to suppress the proliferation of these cells (RORγτ+Ki-67+), along with the proliferation of other cells (Ki-67+) (n = 11). A PSG concentration of 10 µg/mL showed similar effect, decreasing the frequency of Ki-67+ and RORγτ+Ki67+ cells. Using Luminex xMAP technology, it was shown that PSG decreased IL-4, IL-5, IL-8, IL-12, IL-13, IL-17, MIP-1ß, IL-10, IFN-γ, TNF-α, G-CSF, and GM-CSF concentrations in Th17-polarized CD4+ cell cultures but did not affect IL-2, IL-7, and MCP-1 output. CONCLUSIONS: In the experimental model used, PSG had а mainly suppressive effect on the Th17 polarization and cytokine profile of Th17-polarized CD4+ cell cultures. As Th17 activity and a pro-inflammatory cytokine background are unfavorable during pregnancy, the observed PSG effects may play a fetoprotective role in vivo.


Subject(s)
Pregnancy-Specific beta 1-Glycoproteins/metabolism , Pregnancy/immunology , Th17 Cells/immunology , Adult , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , Female , Humans , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Young Adult
5.
Arch Med Res ; 51(6): 504-514, 2020 08.
Article in English | MEDLINE | ID: mdl-32546445

ABSTRACT

BACKGROUND: Cervical Cancer (CC) is a worldwide public health concern associated with genetic alterations, among these the gain of the 19q chromosome harboring the Pregnancy Specific Glycoproteins (PSG) gene family. These proteins play a critical role in pregnancy, with participation in immunotolerance, angiogenesis, and invasion processes, which are also observed in carcinogenesis. The aim of this study was to determine the molecular alterations of PSG1 and its relationship with CC. METHODS: PSG1 Copy Number Variation (CNV) was evaluated in 31 CC and eight normal cervical tissues by qPCR. PSG1 expression was correlated with HPV detection and IL-10 and TGF-ß expression in CC samples. Finally, PSG1 protein expression was evaluated by immunofluorescence in CC cell lines, by immunohistochemistry in a tissue microarray, and by immunoblotting in the sera of women with normal cervix, pre-invasive lesions, and CC. RESULTS: PSG1 showed a gain of 25.6% in CNV and gene expression in CC. There was a lack of PSG1 expression in normal cervical epithelium and positive immunostaining in 57% of CC tissues, while all CC cell lines expressed PSG1. Finally, PSG1 was immunodetected in 90% of pre-invasive lesions and in all CC serum samples, but not in healthy women. PSG1 expression correlates with the expression of IL-10 and TGF-ß in CC tissues, but not with the presence of HPV. CONCLUSION: These data show evidence of the differential expression of PSG1 in CC that could explain its participation in tumor-biology and immunotolerance mechanisms. Further, its immunodetection could provide early detection of this cancer.


Subject(s)
Pregnancy-Specific beta 1-Glycoproteins/metabolism , Uterine Cervical Neoplasms/metabolism , Female , Humans , Pregnancy
6.
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
7.
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
9.
Nature ; 564(7735): 263-267, 2018 12.
Article in English | MEDLINE | ID: mdl-30487605

ABSTRACT

The placenta is the extraembryonic organ that supports the fetus during intrauterine life. Although placental dysfunction results in major disorders of pregnancy with immediate and lifelong consequences for the mother and child, our knowledge of the human placenta is limited owing to a lack of functional experimental models1. After implantation, the trophectoderm of the blastocyst rapidly proliferates and generates the trophoblast, the unique cell type of the placenta. In vivo, proliferative villous cytotrophoblast cells differentiate into two main sub-populations: syncytiotrophoblast, the multinucleated epithelium of the villi responsible for nutrient exchange and hormone production, and extravillous trophoblast cells, which anchor the placenta to the maternal decidua and transform the maternal spiral arteries2. Here we describe the generation of long-term, genetically stable organoid cultures of trophoblast that can differentiate into both syncytiotrophoblast and extravillous trophoblast. We used human leukocyte antigen (HLA) typing to confirm that the organoids were derived from the fetus, and verified their identities against four trophoblast-specific criteria3. The cultures organize into villous-like structures, and we detected the secretion of placental-specific peptides and hormones, including human chorionic gonadotropin (hCG), growth differentiation factor 15 (GDF15) and pregnancy-specific glycoprotein (PSG) by mass spectrometry. The organoids also differentiate into HLA-G+ extravillous trophoblast cells, which vigorously invade in three-dimensional cultures. Analysis of the methylome reveals that the organoids closely resemble normal first trimester placentas. This organoid model will be transformative for studying human placental development and for investigating trophoblast interactions with the local and systemic maternal environment.


Subject(s)
Maternal-Fetal Relations , Models, Biological , Organoids/cytology , Organoids/physiology , Placentation , Tissue Culture Techniques , Trophoblasts/cytology , Trophoblasts/physiology , Cell Differentiation , Cell Movement , Chorionic Gonadotropin/metabolism , DNA Methylation , Decidua/cytology , Female , Growth Differentiation Factor 15/metabolism , HLA Antigens/metabolism , Humans , Organoids/metabolism , Pregnancy , Pregnancy-Specific beta 1-Glycoproteins/metabolism , Transcriptome/genetics , Trophoblasts/metabolism
10.
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
11.
Sci Rep ; 8(1): 9445, 2018 06 21.
Article in English | MEDLINE | ID: mdl-29930344

ABSTRACT

Cervical cancer (CC) is associated with alterations in immune system balance, which is primarily due to a shift from Th1 to Th2 and the unbalance of Th17/Treg cells. Using in silico DNA copy number analysis, we have demonstrated that ~20% of CC samples exhibit gain of 8q22.3 and 19q13.31; the regions of the genome that encodes the KLF10 and PSG genes, respectively. Gene expression studies demonstrated that there were no alterations in KLF10 mRNA expression, whilst the PSG2 and -5 genes were up-regulated by 1.76 and 3.97-fold respectively in CC compared to normal tissue controls. siRNA and ChIP experiments in SiHa cells have demonstrated that KLF10 participates in immune response through regulation of IL6, IL25 and PSG2 and PSG5 genes. Using cervical tissues from KLF10-/- mice, we have identified down-regulation of PSG17, -21 and -23 and IL11. These results suggest that KLF10 may regulate immune system response genes in cervical cancer among other functions. KLF10 and PSG copy number variations and alterations in mRNA expression levels could represent novel molecular markers in CC.


Subject(s)
Early Growth Response Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Kruppel-Like Transcription Factors/metabolism , Pregnancy-Specific beta 1-Glycoproteins/genetics , Uterine Cervical Neoplasms/genetics , Animals , Cell Line, Tumor , DNA Copy Number Variations , Early Growth Response Transcription Factors/genetics , Female , Humans , Interleukins/genetics , Interleukins/metabolism , Kruppel-Like Transcription Factors/genetics , Mice , Pregnancy-Specific beta 1-Glycoproteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Uterine Cervical Neoplasms/immunology
12.
Sci China Life Sci ; 60(5): 528-535, 2017 May.
Article in English | MEDLINE | ID: mdl-28078509

ABSTRACT

Hepatocellular carcinoma (HCC) is a common malignant solid tumor characterized by rich vascularization. Pregnancy-specific glycoprotein 9 (PSG9) is a member of the carcinoembryonic antigen (CEA)/PSG family and is produced at high levels during pregnancy. We previously identified PSG9 as an HCC-related protein. However, the expression of PSG9 and its regulation during HCC carcinogenesis remain poorly explored. In the present study, we first found that the levels of PSG9 protein were significantly increased in the plasma of HCC patients. PSG9 overexpression also increased the proliferation ability of an HCC cell line. High expression of PSG9 was associated with angiogenesis by accelerating VEGFA expression. In addition, Cox's proportional hazards model analysis revealed that the plasma level of PSG9 was an independent prognostic factor for overall survival. We propose that PSG9 is a novel indicator of prognosis in patients with HCC and could serve as a novel therapeutic target for HCC. Furthermore, our results indicate that PSG9 protein may facilitate the development of HCC by fostering angiogenesis via promoting VEGFA production in cancer cells.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Neovascularization, Pathologic/metabolism , Pregnancy-Specific beta 1-Glycoproteins/metabolism , Vascular Endothelial Growth Factor A/metabolism , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Liver Neoplasms/blood supply , Liver Neoplasms/genetics , Male , Middle Aged , Neovascularization, Pathologic/blood , Neovascularization, Pathologic/genetics , Pregnancy-Specific beta 1-Glycoproteins/classification , Pregnancy-Specific beta 1-Glycoproteins/genetics , Prognosis , RNA Interference , Vascular Endothelial Growth Factor A/genetics
13.
Curr Med Chem ; 24(3): 245-267, 2017.
Article in English | MEDLINE | ID: mdl-27881070

ABSTRACT

BACKGROUND: Pregnancy specific ß1-glycoproteins (PSGs) have long been recognized as trophoblast quality and embryo viability markers. However, biological roles of PSGs remain obscure, and structure/function relationships to other feto-placental proteins as well as implications for drug design have not been reviewed. This review summarizes and discusses advances in 45-year studies of PSGs with focus on the latest achievements and the challenges for future investigations. METHODS: Literature search was performed to review the majority of recent PSG studies with emphasis on usage of high-throughput integrated proteomic profiling technologies, systems biology and bioinformatics approaches that enhance novel biomarker and drug target discovery as well as protein structure/activity analysis. RESULTS: Clinical significance and screening performance improved when PSG measurements were combined with those of other placenta-derived proteins: hCG, hPL, PAPP-A, and proMBP. Nevertheless, analysis of protein co-expression and co-localization data and the involvement of PSGs in protein interaction networks are being introduced to discover novel, specific and high-sensitive, gestational/cancer biomarkers. Despite biological roles of PSGs are not fully understood, there are evidences of that they exhibit immunomodulatory, antiinflammatory and proangiogenic effects. Investigation of structure/function relationships showed that PSGs may function in cooperative/coordinated manner with numerous regulatory proteins including alpha-fetoprotein and transforming growth factors-ß; this is provided by the presence of conserved short linear motifs (SLiMs) such as RGD, PXXP and AFP14-20-like (YXCX) ones. CONCLUSION: PSG-derived peptides may be used as a rationale to design novel drugs that mimic SLiMs involved in protein-protein interactions to inhibit domain-motif binding and to block cell signaling, and/or exert immunomodulatory, anti-inflammatory and proangiogenic effects.


Subject(s)
Drug Design , Pregnancy-Specific beta 1-Glycoproteins/chemistry , Pregnancy-Specific beta 1-Glycoproteins/metabolism , Amino Acid Sequence , Animals , Biomarkers/chemistry , Biomarkers/metabolism , Humans , Proteomics , Structure-Activity Relationship
14.
Dokl Biochem Biophys ; 470(1): 361-363, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27817017

ABSTRACT

The role of human pregnancy-specific ß1-glycoprotein (PSG) in the regulation of expression of the transcription factor FOXP3 was studied. It is found that, under conditions of directed induction of phenotypic changes in CD4+ lymphocytes to regulatory T cells (Treg), PSG at high concentrations (10 and 100 µg/mL) increased FOXP3 expression. The evaluation of the spontaneous expression level of FOXP3 mRNA showed that PSG (1 and 100 µg/mL) stimulated the expression of this factor in mononuclear cells and isolated CD4+ lymphocytes. Thus, PSG stimulates FOXP3 expression in immunocompetent cells.


Subject(s)
Forkhead Transcription Factors/metabolism , Pregnancy-Specific beta 1-Glycoproteins/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Female , Gene Expression , Humans , Pregnancy , RNA, Messenger/metabolism , T-Lymphocytes, Regulatory/metabolism
15.
Clin Sci (Lond) ; 130(24): 2267-2276, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27653744

ABSTRACT

Chemotherapeutic response is critical for the successful treatment and good prognosis in cancer patients. In this study, we analysed the gene expression profiles of preoperative samples from oestrogen receptor (ER)-negative breast cancer patients with different responses to taxane-anthracycline-based (TA-based) chemotherapy, and identified a group of genes that was predictive. Pregnancy specific beta-1-glycoprotein 1 (PSG1) played a central role within signalling pathways of these genes. Inhibiting PSG1 can effectively reduce chemoresistance via a transforming growth factor-ß (TGF-ß)-related pathway in ER-negative breast cancer cells. Drug screening then identified dicumarol (DCM) to target the PSG1 and inhibit chemoresistance to TA-based chemotherapy in vitro, in vivo, and in clinical samples. Taken together, this study highlights PSG1 as an important mediator of chemoresistance, whose effect could be diminished by DCM.


Subject(s)
Anticoagulants/therapeutic use , Breast Neoplasms/genetics , Dicumarol/therapeutic use , Pregnancy-Specific beta 1-Glycoproteins/antagonists & inhibitors , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Bridged-Ring Compounds/therapeutic use , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Humans , Pregnancy-Specific beta 1-Glycoproteins/genetics , Pregnancy-Specific beta 1-Glycoproteins/metabolism , Taxoids/therapeutic use , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
16.
Dokl Biol Sci ; 469(1): 206-8, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27595833

ABSTRACT

The role of heterogenic human pregnancy-specific glycoprotein (PSG), obtained by the authors' technology, in the regulation of the indoleamine-2,3-dioxygenase (IDO) activity in female blood monocytes has been studied in vitro. PSG stimulated IDO activity under the conditions of induction of the monocytes by interferon-γ. Upon the induction of cell proliferation by lipopolysaccharides, the stimulating effect was obtained only with 10 µg/mL of PSG. Enhanced IDO activity is probably a factor of peripheral immunological tolerance and antimicrobial protection against intracellular infections in the gestation period.


Subject(s)
Immune Tolerance , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Monocytes/immunology , Pregnancy-Specific beta 1-Glycoproteins/immunology , Pregnancy/immunology , Adult , Female , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Monocytes/metabolism , Pregnancy/blood , Pregnancy-Specific beta 1-Glycoproteins/metabolism
17.
Oncotarget ; 7(38): 61562-61574, 2016 Sep 20.
Article in English | MEDLINE | ID: mdl-27528036

ABSTRACT

PSG9 is a member of the pregnancy-specific glycoprotein (PSG) family and has been shown to contribute to the progression of colorectal cancer (CRC) and cancer-related angiogenesis. Here, we aim to investigate abnormal PSG9 levels in patients with CRC and to emphasize the role of PSG9 in driving tumorigenesis. Serum from 140 patients with CRC and 125 healthy controls as well as 74 paired tumors and adjacent normal tissue were used to determine PSG9 levels. We discovered that PSG9 was significantly increased in serum (P<0.001) and in tumor tissues (P<0.001) from patients with CRC. Interestingly, the increased PSG9 levels correlated with poor survival (P=0.009) and microvessel density (MVD) (P=0.034). The overexpression of PSG9 strongly promoted the proliferation and migration of HCT-116 and HT-29 cells. However, PSG9 depletion inhibited the proliferation of SW-480 cells. Using a human umbilical vein endothelial cell tube-forming assay, we found that PSG9 promoted angiogenesis. The overexpression of PSG9 also increased the growth of tumor xenografts in nude mice. Co-immunoprecipitation experiments revealed that PSG9 was bound to SMAD4. The PSG9/SMAD4 complex recruited cytoplasmic SMAD2/3 to form a complex, which enhanced SMAD4 nuclear retention. The PSG9 and SMAD4 complex activated the expression of multiple angiogenesis-related genes (included IGFBP-3, PDGF-AA, GM-CSF, and VEGFA). Together, our findings illustrate the innovative mechanism by which PSG9 drives the progression of CRC and tumor angiogenesis. This occurs via nuclear translocation of PSG9/SMAD4, which activates angiogenic cytokines. Therefore, our study may provide evidence for novel treatment strategies by targeting PSG9 in antiangiogenic cancer therapy.


Subject(s)
Carcinogenesis/pathology , Colorectal Neoplasms/pathology , Neovascularization, Pathologic/metabolism , Pregnancy-Specific beta 1-Glycoproteins/metabolism , Smad4 Protein/metabolism , Animals , Cell Movement , Cell Nucleus/metabolism , Cell Proliferation , Colorectal Neoplasms/blood , Colorectal Neoplasms/mortality , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , HCT116 Cells , HT29 Cells , Humans , Immunoprecipitation , Insulin-Like Growth Factor Binding Protein 3/metabolism , Kaplan-Meier Estimate , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Platelet-Derived Growth Factor/metabolism , Pregnancy-Specific beta 1-Glycoproteins/analysis , Signal Transduction , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
18.
PLoS One ; 11(7): e0158050, 2016.
Article in English | MEDLINE | ID: mdl-27389696

ABSTRACT

The pregnancy-specific glycoproteins (PSGs) are a family of proteins secreted by the syncytiotrophoblast of the placenta and are the most abundant trophoblastic proteins in maternal blood during the third trimester. The human PSG family consists of 10 protein-coding genes, some of which have a possible role in maintaining maternal immune tolerance to the fetus. PSG9 was reported as a potential predictive biomarker of pre-eclampsia, a serious complication of pregnancy that has been related to immunological dysfunction at the fetal-maternal interface. Therefore, we hypothesized that PSG9 may have an immunoregulatory role during pregnancy. We found that PSG9 binds to LAP and activates the latent form of TGF-ß1. In addition, PSG9 induces the secretion of TGF-ß1 from macrophages but not from CD4+ T-cells. TGF-ß1 is required for the ex vivo differentiation of regulatory T-cells and, consistent with the ability of PSG9 to activate this cytokine, we observed that PSG9 induces the differentiation of FoxP3+ regulatory T-cells from naïve murine and human T-cells. Cytokines that are associated with inflammatory responses were also reduced in the supernatants of T-cells treated with PSG9, suggesting that PSG9, through its activation of TGFß-1, could be a potent inducer of immune tolerance.


Subject(s)
Forkhead Transcription Factors/metabolism , Pregnancy-Specific beta 1-Glycoproteins/metabolism , T-Lymphocytes, Regulatory/cytology , Transforming Growth Factor beta1/metabolism , Animals , Cell Differentiation , Chemokines/metabolism , Female , Humans , Immune System , Inflammation , Kinetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy , Surface Plasmon Resonance
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...