Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 815
Filter
1.
Bull Exp Biol Med ; 177(1): 124-132, 2024 May.
Article in English | MEDLINE | ID: mdl-38960961

ABSTRACT

Pregnancy-specific ß1-glycoprotein (PSG), one of the most important proteins of pregnancy, has a pronounced immunosuppressive effect. Short peptides of PSG, the so-called SLiMs (short linear motifs), are promising molecules for mild immunosuppression. We studied in vitro effect of short PSG peptides (YACS, YQCE, YVCS, and YECE) on differentiation and cytokine profile of human T-regulatory lymphocytes (Treg). T helpers isolated from the peripheral blood and polarized into the Treg phenotype with a T-cell activator (anti-CD2/3/28) and the cytokines IL-2 and transforming grown factor ß (TGFß) were used. PSG peptides were shown to have no direct modulatory effect on Treg differentiation in a culture of CD4+ cells polarized to the Treg phenotype. At the same time, PSG peptides had no effect on the viability and number of CD4+ cells in the in vitro culture. PSG peptides also had no effect on the levels of TNFα, IL-8, IL-2, macrophage inflammatory protein 1ß, IL-17, IL-10, IL-6, granulocyte-macrophage CSF, monocyte chemoattractant protein 1, IL-13, IL-5, IL-7, IL-12(p70), IL-1ß, granulocyte CSF, IL-4, but decreased IFNγ levels. The observed ability of the YQCE peptide to reduce the production of this proinflammatory Th1 cytokine by T helper cells can be interpreted as a positive effect. Our findings can be used for further development of safe peptide drugs based on SLiMs sequences.


Subject(s)
Cell Differentiation , Cytokines , Pregnancy-Specific beta 1-Glycoproteins , T-Lymphocytes, Regulatory , Humans , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , Cell Differentiation/drug effects , Pregnancy-Specific beta 1-Glycoproteins/metabolism , Cytokines/metabolism , Female , Pregnancy , Peptides/pharmacology , Interleukin-2/metabolism , Cells, Cultured
2.
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
4.
In Vivo ; 36(6): 2700-2707, 2022.
Article in English | MEDLINE | ID: mdl-36309385

ABSTRACT

BACKGROUND/AIM: Pregnancy specific beta-1-glycoprotein 1 (PSG1) is a member of the immunoglobulin superfamily and associated with carcinoembryonic antigens. It has been reported to be highly expressed in variety of cancers. However, the role of PSG1 in gastric cancer remains unclear. The aim of our study was to examine the clinical significance and functional role of PSG1 in gastric cancer. MATERIALS AND METHODS: We analyzed the association between PSG1 expression levels and clinicopathological features using Kaplan-Meier survival curves and publicly available microarray data. In gastric cancer cell lines, PSG1 expression levels were detected by polymerase chain reaction and western blot analysis. The functional role of PSG1 on the proliferation, migration and invasive abilities were also investigated using PSG1 siRNA or an over-expression plasmid vector through WST, transwell migration and invasion assays. RESULTS: PSG1 expression levels were higher in gastric cancer patient tissues than in normal gastric tissues. Increased expression of PSG1 was associated with poor patient prognosis. Knockdown of PSG1 inhibited cell proliferation, migration, and invasion in gastric cancer cells. In contrast, over-expression of PSG1 enhanced cell proliferation, migration, and invasion. CONCLUSION: PSG1 is up-regulated in gastric cancer and may serve as an oncogene that promotes cell proliferation, migration, and invasion. PSG1 is an independent prognostic factor for the progression of gastric cancer and may be a potential biomarker and therapeutic target for gastric cancer.


Subject(s)
Stomach Neoplasms , Female , Humans , Pregnancy , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Glycoproteins/genetics , Glycoproteins/metabolism , Neoplasm Invasiveness/genetics , Prognosis , Stomach Neoplasms/pathology , Transcription Factors/genetics , Pregnancy-Specific beta 1-Glycoproteins
5.
J Am Heart Assoc ; 11(7): e024536, 2022 04 05.
Article in English | MEDLINE | ID: mdl-35322669

ABSTRACT

Background Preeclampsia is pregnancy specific, involving significant maternal endothelial dysfunction. Predictive biomarkers are lacking. We evaluated the biomarker potential, expression, and function of PSG7 (pregnancy-specific ß-1 glycoprotein 7) and PSG9 (pregnancy-specific ß-1 glycoprotein 9) in preeclampsia. Methods and Results At 36 weeks gestation preceding term preeclampsia diagnosis, PSG7 and PSG9 (in Australian cohorts of n=918 and n=979, respectively) were significantly increased before the onset of term preeclampsia (PSG7, P=0.013; PSG9, P=0.0011). In samples collected at 28 to 32 weeks from those with preexisting cardiovascular disease and at high risk of preeclampsia (Manchester Antenatal Vascular Service, UK cohort, n=235), both PSG7 and PSG9 were also significantly increased preceding preeclampsia onset (PSG7, P<0.0001; PSG9, P=0.0003) relative to controls. These changes were validated in the plasma and placentas of patients with established preeclampsia who delivered at <34 weeks gestation (PSG7, P=0.0008; PSG9, P<0.0001). To examine whether PSG7 and PSG9 are associated with increasing disease severity, we measured them in a cohort from South Africa stratified for this outcome, the PROVE (Preeclampsia Obstetric Adverse Events) cohort (n=72). PSG7 (P=0.0027) and PSG9 (P=0.0028) were elevated among patients who were preeclamptic with severe features (PROVE cohort), but not significantly changed in those without severe features or with eclampsia. In syncytialized first trimester cytotrophoblast stem cells, exposure to TNFα (tumor necrosis factor α) or IL-6 (interleukin 6) significantly increased the expression and secretion of PSG7 and PSG9. In contrast, when we treated primary endothelial cells with recombinant PSG7 and PSG9, we only observed modest changes in Flt-1 (FMS-like tyrosine kinase-1) expression and Plgf (placental growth factor) expression, and no other effects on proangiogenic/antiangiogenic or endothelial dysfunction markers were observed. Conclusions Circulating PSG7 and PSG9 are increased before preeclampsia onset and among those with established disease with their production and release potentially driven by placental inflammation.


Subject(s)
Pre-Eclampsia , Pregnancy-Specific beta 1-Glycoproteins , Australia/epidemiology , Biomarkers/blood , Endothelial Cells/metabolism , Female , Glycoproteins , Humans , Placenta/metabolism , Placenta Growth Factor , Pre-Eclampsia/diagnosis , Pregnancy , Pregnancy-Specific beta 1-Glycoproteins/analysis
6.
JBRA Assist Reprod ; 26(2): 267-273, 2022 04 17.
Article in English | MEDLINE | ID: mdl-34755504

ABSTRACT

OBJECTIVE: The most used definition for fetal growth restriction (FGR) is a fetus whose estimated weight is below the 10th percentile for its gestational age. Pregnancy-specific beta-1-glycoprotein (PSG-1) is an immunomodulator found in maternal serum during pregnancy. This study aimed to determine the serum levels of PSG-1 and clarify the potential role of this molecule in the etiopathogenesis of FGR. METHODS: Eighty women carrying fetuses with FGR and 80 healthy pregnant women were included in the study. Demographic data, laboratory values, and Doppler Ultrasonography (USG) results of all cases were recorded. Venous blood samples were taken from all cases before birth. PSG-1 values were studied by the ELISA method. An Independent Samples T-test was used to evaluate the results. The correlations between parameters were evaluated based on Spearman's rank correlation coefficient. P-values <0.05 were considered statistically significant. RESULTS: When the groups were evaluated for serum PSG-1 levels, the median serum PSG-1 level was lower in pregnant women carrying fetuses with FGR than in controls (0.05 < p>0.10). Median serum PSG-1 was lower in patients with absent end diastolic flow (AEDF) in the umbilical artery in Doppler ultrasound scans than in patients without AEDF, but the difference was not statistically significant (p>0.05). In patients with serum PSG-1 values below 12.93 with 50% sensitivity and 76% specificity, the risk of FGR was higher. CONCLUSIONS: Serum PSG-1 levels may be lower in complicated pregnancies due to problems related to placental insufficiency and FGR.


Subject(s)
Fetal Growth Retardation , Pregnancy-Specific beta 1-Glycoproteins , Female , Fetal Growth Retardation/etiology , Glycoproteins , Humans , Placenta/blood supply , Pregnancy , Ultrasonography, Prenatal
7.
Bull Exp Biol Med ; 172(2): 169-174, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34855088

ABSTRACT

We studied the effects of pregnancy-specific ß1-glycoprotein (PSG) on the replicative potential of naïve T cells (CD45RA+) and immune memory T cells (CD45R0+) in vitro by evaluating the expression of the hTERT gene in combination with the proliferative activity of cells. Human PSG was obtained by the author's patented method of immunopurification using a biospecific sorbent with subsequent removal of immunoglobulin contamination on a HiTrap Protein G HP column. We used monocultures of CD45RA+ and CD45R0+ lymphocytes isolated from peripheral blood mononuclear cells of reproductive-age women. It was found that PSG in physiological concentrations inhibited the expression of the hTERT gene mRNA in naïve T cells and immune memory T cells and simultaneously reduced the number of proliferating T cells estimated by the differential gating method. At the same time, PSG reduced CD71 expression only on naïve T cells without affecting this molecule on immune memory T cells. Thus, PSG decreased the replication potential and suppressed the proliferation of T cells and immune memory T cells, which in the context of pregnancy can contribute to the formation of immune tolerance to the semi-allogeneic embryo.


Subject(s)
Memory T Cells/drug effects , Pregnancy-Specific beta 1-Glycoproteins/pharmacology , T-Lymphocytes/drug effects , Apoptosis/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Female , Humans , Immune Tolerance/immunology , Immunologic Memory/drug effects , Immunologic Memory/physiology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/physiology , Lymphocyte Activation/drug effects , Memory T Cells/physiology , Pregnancy , Pregnancy-Specific beta 1-Glycoproteins/physiology , T-Lymphocytes/physiology
8.
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
9.
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
10.
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
11.
Proc Natl Acad Sci U S A ; 117(28): 16339-16345, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32601217

ABSTRACT

We present a technique to construct a simplification of a feature network which can be used for interactive data exploration, biological hypothesis generation, and the detection of communities or modules of cofunctional features. These are modules of features that are not necessarily correlated, but nevertheless exhibit common function in their network context as measured by similarity of relationships with neighboring features. In the case of genetic networks, traditional pathway analyses tend to assume that, ideally, all genes in a module exhibit very similar function, independent of relationships with other genes. The proposed technique explicitly relaxes this assumption by employing the comparison of relational profiles. For example, two genes which always activate a third gene are grouped together even if they never do so concurrently. They have common, but not identical, function. The comparison is driven by an average of a certain computationally efficient comparison metric between Gaussian mixture models. The method has its basis in the local connection structure of the network and the collection of joint distributions of the data associated with nodal neighborhoods. It is benchmarked on networks with known community structures. As the main application, we analyzed the gene regulatory network in lung adenocarcinoma, finding a cofunctional module of genes including the pregnancy-specific glycoproteins (PSGs). About 20% of patients with lung, breast, uterus, and colon cancer in The Cancer Genome Atlas (TCGA) have an elevated PSG+ signature, with associated poor group prognosis. In conjunction with previous results relating PSGs to tolerance in the immune system, these findings implicate the PSGs in a potential immune tolerance mechanism of cancers.


Subject(s)
Computational Biology/methods , Immune Tolerance/genetics , Neoplasms/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Models, Statistical , Neoplasms/immunology , Pregnancy-Specific beta 1-Glycoproteins/genetics , Prognosis
12.
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
13.
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
14.
J Obstet Gynaecol ; 40(8): 1074-1078, 2020 Nov.
Article in English | MEDLINE | ID: mdl-31790616

ABSTRACT

The aim of this study was to investigate the relationship between the maternal serum levels of pregnancy-specific beta-1-glycoprotein 1 (PSG1) and preeclampsia, and to compare levels of PSG1 in pregnancies with preeclampsia and uneventful pregnancies. A case-control study was conducted in a research and training hospital. A total of 40 women with preeclampsia and 42 healthy pregnant women who were gestational age-matched were included. Serum PSG1 levels were measured using enzyme-linked immunosorbent assay. The maternal serum PSG1 levels were significantly lower in patients with preeclampsia compared with controls (11.60 ± 8.08 vs. 17.58 ± 9.72 ng/mL, p = .003). Circulating PSG1 levels were negatively correlated with age in the preeclampsia and control groups (r = -0.322, p = .043), (r = -0.430, p = .005). PSG1 levels, age, blood urea nitrogen levels and birth weight were significantly associated with high odds of having preeclampsia. Receiver operating characteristic (ROC) curve analysis confirmed that the area under ROC curve was 0.707 (95% CI: [0.595-0.819], p < .001) for PSG1. The optimal cut-off value of PSG1 for detecting preeclampsia was ≤ 11.80 ng/mL. There may be a decrease in PSG1 production in preeclampsia-complicated pregnancies where there are pathologies related to placenta formation. A decline in PSG1 concentrations may reflect placental dysfunction.Impact StatementWhat is already known on this subject? Previous studies have reported abnormal pregnancy-specific glycoprotein (PSG) levels in complicated pregnancies and demonstrated their importance in maintaining a healthy pregnancy. Human PSG homologues have been identified in species with haemochorial placentation such as non-human primates, rats and mice, where foetal cells are in direct contact with the maternal circulation. There are studies in which there is no clear relationship between PSGs and preeclampsia.What the results of this study add? We have demonstrated that circulating PSG1 levels were significantly lower in women with preeclampsia than in healthy pregnant women. There may be a decrease in PSG1 production in preeclampsia-complicated pregnancies where there are pathologies related to placenta formation and function. The results obtained from this current study could be used to clarify the relationship between PSG1 levels and preeclampsia.What the implications are for clinical practice and/or further research? Evaluation of the role of circulating PSG1 levels in preeclampsia would be helpful in order to design further studies to determine the feasibility of using PSG1 as a serum marker to predict the risk of developing preeclampsia. The screening performance of PSG1 for preeclampsia is not yet clinically relevant, but may become so when evaluated together with other placental proteins. This will give a lead to further researches which could focus on the early detection of preeclampsia with the combination of several serum markers.


Subject(s)
Pre-Eclampsia/blood , Pregnancy Complications/blood , Pregnancy-Specific beta 1-Glycoproteins/analysis , Adult , Biomarkers/blood , Birth Weight , Blood Urea Nitrogen , Case-Control Studies , Enzyme-Linked Immunosorbent Assay , Female , Gestational Age , Humans , Infant, Newborn , Odds Ratio , Pregnancy , ROC Curve
15.
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
16.
PLoS Genet ; 15(6): e1008107, 2019 06.
Article in English | MEDLINE | ID: mdl-31194736

ABSTRACT

Spontaneous preterm birth (SPTB) is the leading cause of neonatal death and morbidity worldwide. Both maternal and fetal genetic factors likely contribute to SPTB. We performed a genome-wide association study (GWAS) on a population of Finnish origin that included 247 infants with SPTB (gestational age [GA] < 36 weeks) and 419 term controls (GA 38-41 weeks). The strongest signal came within the gene encoding slit guidance ligand 2 (SLIT2; rs116461311, minor allele frequency 0.05, p = 1.6×10-6). Pathway analysis revealed the top-ranking pathway was axon guidance, which includes SLIT2. In 172 very preterm-born infants (GA <32 weeks), rs116461311 was clearly overrepresented (odds ratio 4.06, p = 1.55×10-7). SLIT2 variants were associated with SPTB in another European population that comprised 260 very preterm infants and 9,630 controls. To gain functional insight, we used immunohistochemistry to visualize SLIT2 and its receptor ROBO1 in placentas from spontaneous preterm and term births. Both SLIT2 and ROBO1 were located in villous and decidual trophoblasts of embryonic origin. Based on qRT-PCR, the mRNA levels of SLIT2 and ROBO1 were higher in the basal plate of SPTB placentas compared to those from term or elective preterm deliveries. In addition, in spontaneous term and preterm births, placental SLIT2 expression was correlated with variations in fetal growth. Knockdown of ROBO1 in trophoblast-derived HTR8/SVneo cells by siRNA indicated that it regulate expression of several pregnancy-specific beta-1-glycoprotein (PSG) genes and genes involved in inflammation. Our results show that the fetal SLIT2 variant and both SLIT2 and ROBO1 expression in placenta and trophoblast cells may be correlated with susceptibility to SPTB. SLIT2-ROBO1 signaling was linked with regulation of genes involved in inflammation, PSG genes, decidualization and fetal growth. We propose that this receptor-ligand couple is a component of the signaling network that promotes SPTB.


Subject(s)
Fetal Development/genetics , Genetic Predisposition to Disease , Intercellular Signaling Peptides and Proteins/genetics , Nerve Tissue Proteins/genetics , Premature Birth/genetics , Receptors, Immunologic/genetics , Female , Fetus , Finland , Gene Expression Regulation/genetics , Gene Frequency , Genome-Wide Association Study , Humans , Placenta/pathology , Polymorphism, Single Nucleotide , Pregnancy , Pregnancy-Specific beta 1-Glycoproteins/genetics , Premature Birth/pathology , Signal Transduction , Trophoblasts/pathology , Roundabout Proteins
17.
Domest Anim Endocrinol ; 67: 54-62, 2019 04.
Article in English | MEDLINE | ID: mdl-30690258

ABSTRACT

The efficacy of several protocols for ovulation synchronization and timed artificial insemination (TAI) in goats was examined. In addition, the relationship between levels of pregnancy specific protein B (PSPB) during gestation assessed with a commercially available ELISA and the number of offspring at birth was determined. In Experiment 1, 70 does were randomized into four treatments: (1) breed by estrus [BBE], (2) 6-d treatment with a new [C6N], (3) once-used [C61], or (4) twice-used Controled Internal Drug Release (CIDR) device [C62)]. BBE does received two 15 mg doses of prostaglandin-F2α (PGF) at a 10-d interval and were bred 12 h after estrus onset. CIDR groups received a CIDR for 6 d with 15 mg PGF given at CIDR removal. TAI was performed 48 h after CIDR removal and does were given 50 µg GnRH. All does were inseminated with a single dose of frozen semen using a non-surgical, transcervical technique. Pregnancy rates for the BBE, C6N, C61 and C62 treatment groups were 39% ± 12%, 64% ± 12%, 77% ± 12% and 57% ± 12%, respectively, and did not differ. Reuse of CIDRs, even with reuse extending for a total of 21 d, was as effective as new CIDRs for synchronization of ovulation. In Experiment 2, 68 does were randomized into four treatments: (1) BBE, (2) C6N, (3) NC.Synch [NCS], (4) modified NCS [NCSM]. The BBE and C6N groups were as described for Experiment 1. The NCS and NCSM groups received 15 mg PGF on Day 1, 50 µg GnRH on Day 8 and 15 mg PGF on Day 15 (NCS) or Day 15.5 (NCSM). Does were bred by TAI at 72 h (NCS) or 60 h (NCSM) after the second PGF injection. All does in the NCS and NCSM groups received 50 µg GnRH at TAI. Pregnancy rates were 53% ± 12%, 30% ± 11%, 50% ± 11% and 41% ± 12% for does in the BBE, C6N, NCS and NCSM group, respectively, and did not differ. In Experiment 3, 62 does pregnant to TAI were bled at Days 48 and 85 post-insemination for PSPB. Data on kid numbers and birth weights were subsequently recorded. At Day 48 of gestation, PSPB levels for does birthing singletons were lower than for does birthing twins or triplets (25.0 ± 0.1a, 28.8 ± 0.1b and 30.7 ± 0b ng/mL, respectively, abP<0.05). At Day 85 of gestation, PSPB levels were progressively greater for does birthing singletons versus twins versus triplets (27.0 ± 0.1a, 28.5 ± 0.1b and 31.6 ± 0c ng/mL, abcP<0.05). In conclusion, PSPB concentrations detected using a commercially available ELISA at Day 48 or 85 of gestation could distinguish does carrying single versus multiple fetuses.


Subject(s)
Estrus Synchronization/methods , Goats/physiology , Litter Size , Ovulation/physiology , Pregnancy-Specific beta 1-Glycoproteins/analysis , Animals , Delayed-Action Preparations , Dinoprost/administration & dosage , Drug Delivery Systems/instrumentation , Enzyme-Linked Immunosorbent Assay/veterinary , Equipment Reuse/veterinary , Female , Gestational Age , Goats/blood , Gonadotropin-Releasing Hormone/administration & dosage , Insemination, Artificial/methods , Insemination, Artificial/veterinary , Pregnancy , Pregnancy Outcome , Progesterone/administration & dosage
18.
Front Med ; 13(2): 250-258, 2019 Apr.
Article in English | MEDLINE | ID: mdl-29770948

ABSTRACT

Biomarkers for hepatocellular carcinoma (HCC) following curative resection are not currently sufficient for prognostic indication of overall survival (OS) and disease-free survival (DFS). The aim of this study was to investigate the prognostic performance of osteopontin (OPN), matrix metalloproteinase 7 (MMP7), and pregnancy specific glycoprotein 9 (PSG9) in patients with HCC. A total of 179 prospective patients with HCC provided plasma before hepatectomy. Plasma OPN, MMP7, and PSG9 levels were determined by enzyme-linked immunosorbent assay. Correlations between plasma levels, clinical parameters, and outcomes (OS and DFS) were overall analyzed. High OPN ( ⩾ 149.97 ng/mL), MMP7 ( ⩾ 2.28 ng/mL), and PSG9 ( ⩾ 45.59 ng/mL) were prognostic indicators of reduced OS (P < 0.001, P < 0.001, and P = 0.007, respectively). Plasma PSG9 protein level was an independent factor in predicting OS (P = 0.008) and DFS (P = 0.038). Plasma OPN + MMP7 + PSG9 elevation in combination was a prognostic factor for OS (P < 0.001). OPN was demonstrated to be a risk factorassociated OS in stage I patients with HCC and patients with low α-fetoprotein levels ( < 20 ng/mL). These findings suggested that OPN, MMP7, PSG9 and their combined panels may be useful for aiding in tumor recurrence and mortality risk prediction of patients with HCC, particularly in the early stage of HCC carcinogenesis.


Subject(s)
Carcinoma, Hepatocellular/mortality , Liver Neoplasms/mortality , Matrix Metalloproteinase 7/blood , Osteopontin/blood , Pregnancy-Specific beta 1-Glycoproteins/analysis , Adult , Aged , Biomarkers, Tumor/blood , Carcinoma, Hepatocellular/blood , Enzyme-Linked Immunosorbent Assay , Female , Hepatectomy , Humans , Liver Neoplasms/blood , Male , Middle Aged , Prognosis , Prospective Studies , Risk Assessment , Survival Analysis
19.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...