Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
J Reprod Immunol ; 144: 103280, 2021 04.
Article in English | MEDLINE | ID: mdl-33530024

ABSTRACT

In early human gestation, maternal arterial blood flow into the intervillous space of the developing placenta is obstructed by invaded trophoblasts, which form cellular plugs in uterine spiral arteries. These trophoblast plugs have recently been described to be loosely cohesive with clear capillary-sized channels into the intervillous space by 7 weeks of gestation. Here, we analysed localisation of maternal platelets at the maternal-foetal interface of human first trimester pregnancy, and tested the hypothesis whether HLA-G, which is primarily expressed by extravillous trophoblasts, affects aggregation and adhesion of isolated platelets. Immunohistochemistry of first trimester placental sections localised maternal platelets in vessel-like channels and adjacent intercellular gaps of extravillous trophoblasts in distal parts of columns. Furthermore, this localisation was confirmed by transmission electron microscopy. Neither co-incubation of HLA-G overexpressing JAR cells with isolated platelets, nor incubation with cell-derived soluble HLA-G or recombinant HLA-G affected platelet adhesion and aggregation. Our study suggests that maternal platelets flow through vessel-like channels of distal trophoblast columns and spread into adjacent lateral intercellular gaps, where platelet-derived factors could contribute to trophoblast differentiation into the invasive phenotype.


Subject(s)
Blood Platelets/immunology , Cell Differentiation/immunology , Maternal-Fetal Exchange/immunology , Placental Circulation/immunology , Trophoblasts/physiology , Cell Line , Coculture Techniques , Female , HLA-G Antigens/immunology , HLA-G Antigens/isolation & purification , Humans , Microscopy, Electron, Transmission , Placenta/blood supply , Placenta/cytology , Placenta/immunology , Placenta/ultrastructure , Pregnancy , Pregnancy Trimester, First/immunology , Primary Cell Culture , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Trophoblasts/ultrastructure
2.
Front Immunol ; 11: 1920, 2020.
Article in English | MEDLINE | ID: mdl-33013843

ABSTRACT

Transplacental antibody transfer from mother to fetus provides protection from infection in the first weeks of life, and the four different subclasses of IgG (IgG1, IgG2, IgG3, and IgG4) have diverse roles in protection against infection. In this study, we evaluated concentrations and transplacental transfer ratios of the IgG subclasses in a healthy UK-based cohort of mother-cord pairs, and investigated associations with maternal, obstetric, and fetal factors. In agreement with previous studies, we found a strong association between maternal and cord IgG for all subclasses. We report a transfer efficiency hierarchy of IgG1>IgG3>IgG4=IgG2 in our study population, and our review of the literature demonstrates that there is no consensus in the hierarchy of subclass transfer, despite the commonly made statement that the order is IgG1>IgG4>IgG3>IgG2. We report additional data regarding negative associations between elevated maternal IgG concentrations and maternal/cord transfer ratios, finding an effect on IgG1, IgG2, and IgG3 subclasses. Levels of IgG subclasses were the same between venous and arterial blood samples from the umbilical cord, but there was a significantly higher level of total IgG in arterial blood. We found no correlation between placental FcRn protein levels and IgG transfer in our cohort, suggesting that IgG is the main determinant of observed differences in transplacental transfer ratios at term. Neonatal IgG1 and IgG4 levels were increased with later gestation at delivery, independent of any increase in transplacental transfer, indicating that the benefit of later gestation is through accumulation of these subclasses in the fetus. Neonatal IgG2 levels and transfer ratios were reduced in rhesus-negative pregnancies, suggesting that administered anti-D antibodies may compete for transplacental transfer of this subclass. Maternal influenza vaccination resulted in elevated maternal and neonatal levels of IgG4, whereas maternal Tdap vaccination had no impact on neonatal levels of the subclasses, nor transfer. However, within Tdap vaccinated pregnancies, later gestation at Tdap vaccination was associated with higher transplacental transfer. Our study provides information regarding levels and transfer of IgG subclasses in healthy term pregnancies and demonstrates the importance of recording detailed clinical information in studies of antibody transfer, including parity, ethnicity, and timing of maternal vaccine delivery.


Subject(s)
Fetal Blood/immunology , Immunity, Maternally-Acquired , Immunoglobulin G/blood , Maternal-Fetal Exchange , Placental Circulation/immunology , Adult , Diphtheria-Tetanus-acellular Pertussis Vaccines/administration & dosage , Diphtheria-Tetanus-acellular Pertussis Vaccines/immunology , Female , Humans , Immunogenicity, Vaccine , Immunoglobulin G/classification , Infant, Newborn , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , London , Male , Middle Aged , Pregnancy , Vaccination , Young Adult
3.
J Immunotoxicol ; 17(1): 135-143, 2020 12.
Article in English | MEDLINE | ID: mdl-32538211

ABSTRACT

Arsenic (As) readily crosses the placenta and exposure of the fetus may cause adverse consequences later in life, including immunomodulation. In the current study, the question was asked how the immune repertoire might respond in postnatal life when there is no further As exposure. Here, pregnant mice (Balb/c [H-2d]) were exposed to arsenic trioxide (As2O3) through their drinking water from time of conception until parturition. Their offspring, 4-week-old mice who had not been exposed again to As, were used for functional analyses of innate, humoral and cellular immunity. Compared to cells from non-As-exposed dam offspring, isolated peritoneal macro-phages (Mϕ) displayed no differences in T-cell stimulating ability. Levels of circulating IgG2a but not IgG1 were decreased in As-exposed dam offspring as compared to control offspring counterparts. Mixed-leukocyte reactions (MLR) indicated that CD4+ T-cells from the prenatal As-exposed mice were significantly less responsive to allogenic stimulation as evidenced by decreases in interferon (IFN)-γ and IL-2 production and in expression of CD44 and CD69 (but not CD25) activation markers. Interestingly, the Mϕ from the prenatal As-exposed mice were capable of stimulating normal allogenic T-cells, indicating that T-cells from these mice were refractory to allogenic signals. There was also a significant decrease in absolute numbers of splenic CD4+ and CD8+ T-cells due to prenatal As exposure (as compared to control). Lastly, the impaired immune function of the prenatal As-exposed mice was correlated with a very strong susceptibility to Escherichia coli infection. Taken together, the data from this study clearly show that in utero As exposure may continue to perpetuate a dampening effect on the immune repertoire of offspring, even into the early stages of postnatal life.


Subject(s)
Arsenic Trioxide/toxicity , CD8-Positive T-Lymphocytes/immunology , Escherichia coli Infections/immunology , Maternal Exposure/adverse effects , Prenatal Exposure Delayed Effects/immunology , Animals , Animals, Newborn , Antigens, CD/analysis , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/analysis , Antigens, Differentiation, T-Lymphocyte/metabolism , Arsenic Trioxide/administration & dosage , Arsenic Trioxide/pharmacokinetics , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Disease Susceptibility/blood , Disease Susceptibility/chemically induced , Disease Susceptibility/immunology , Escherichia coli/immunology , Escherichia coli/pathogenicity , Escherichia coli Infections/blood , Escherichia coli Infections/chemically induced , Female , Humans , Hyaluronan Receptors/analysis , Hyaluronan Receptors/metabolism , Interferon-gamma/analysis , Interferon-gamma/metabolism , Interleukin-2/analysis , Interleukin-2/metabolism , Lectins, C-Type/analysis , Lectins, C-Type/metabolism , Male , Maternal-Fetal Exchange/immunology , Mice , Placental Circulation/immunology , Pregnancy , Prenatal Exposure Delayed Effects/blood , Prenatal Exposure Delayed Effects/chemically induced
4.
Proc Natl Acad Sci U S A ; 117(23): 12943-12951, 2020 06 09.
Article in English | MEDLINE | ID: mdl-32461366

ABSTRACT

The IgG Fc domain has the capacity to interact with diverse types of receptors, including the neonatal Fc receptor (FcRn) and Fcγ receptors (FcγRs), which confer pleiotropic biological activities. Whereas FcRn regulates IgG epithelial transport and recycling, Fc effector activities, such as antibody-dependent cellular cytotoxicity (ADCC) and phagocytosis, are mediated by FcγRs, which upon cross-linking transduce signals that modulate the function of effector leukocytes. Despite the well-defined and nonoverlapping functional properties of FcRn and FcγRs, recent studies have suggested that FcγRs mediate transplacental IgG transport, as certain Fc glycoforms were reported to be enriched in fetal circulation. To determine the contribution of FcγRs and FcRn to the maternal-fetal transport of IgG, we characterized the IgG Fc glycosylation in paired maternal-fetal samples from patient cohorts from Uganda and Nicaragua. No differences in IgG1 Fc glycan profiles and minimal differences in IgG2 Fc glycans were noted, whereas the presence or absence of galactose on the Fc glycan of IgG1 did not alter FcγRIIIa or FcRn binding, half-life, or their ability to deplete target cells in FcγR/FcRn humanized mice. Modeling maternal-fetal transport in FcγR/FcRn humanized mice confirmed that only FcRn contributed to transplacental transport of IgG; IgG selectively enhanced for FcRn binding resulted in enhanced accumulation of maternal antibody in the fetus. In contrast, enhancing FcγRIIIa binding did not result in enhanced maternal-fetal transport. These results argue against a role for FcγRs in IgG transplacental transport, suggesting Fc engineering of maternally administered antibody to enhance only FcRn binding as a means to improve maternal-fetal transport of IgG.


Subject(s)
Fetal Blood/immunology , Histocompatibility Antigens Class I/metabolism , Immunoglobulin G/metabolism , Maternal-Fetal Exchange/immunology , Placental Circulation/immunology , Receptors, Fc/metabolism , Animals , Female , Histocompatibility Antigens Class I/genetics , Humans , Immunoglobulin G/immunology , Mice , Mice, Transgenic , Pregnancy , Randomized Controlled Trials as Topic , Receptors, Fc/genetics , Receptors, IgG/genetics , Receptors, IgG/metabolism
5.
J Reprod Immunol ; 133: 52-62, 2019 06.
Article in English | MEDLINE | ID: mdl-31280130

ABSTRACT

PROBLEM: Exposure to systemic maternal inflammation (i.e., maternal sepsis, influenza, human immunodeficiency virus, or pyelonephritis) and intrauterine (IU) inflammation (i.e., chorioamnionitis or preterm labor) have been associated with adverse perinatal sequelae. Whether systemic and localized inflammation leading to adverse outcomes have similar placental mechanisms remain unclear. METHOD OF STUDY: We conducted a study by murine modeling systemic and localized IU inflammation with injections of either intraperitoneal (IP) or IU interleukin-1ß (IL-1ß) and compared fetoplacental hemodynamic changes, cytokine/chemokine expression, and fetal loss. RESULTS: IU IL-1ß exposure reduced offspring survival by 31.1% and IP IL-1ß exposure by 34.5% when compared with control pups. Despite this similar outcome in offspring survival, Doppler analysis revealed a stark difference: IU group displayed worsened fetoplacental hemodynamic changes while no differences were found between IP and control groups. While both IU and IP groups had increases in pro-inflammatory cytokines and chemokines, specific gene expression trends differed between the two groups, once again highlighting their mechanistic differences. CONCLUSION: While both IP and IU IL-1ß exposure similarly affected pup survival, only IU inflammation resulted in fetoplacental hemodynamic changes. We speculate that exposure to maternal systemic and IU inflammation plays a key role in fetal injury by utilizing different placental inflammatory pathways and mechanisms.


Subject(s)
Chorioamnionitis/immunology , Maternal-Fetal Exchange/immunology , Placenta/immunology , Premature Birth/immunology , Animals , Chorioamnionitis/diagnostic imaging , Chorioamnionitis/mortality , Chorioamnionitis/pathology , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Female , Fetus/diagnostic imaging , Fetus/immunology , Humans , Interleukin-1beta/administration & dosage , Interleukin-1beta/immunology , Mice , Placenta/pathology , Placental Circulation/immunology , Pregnancy , Premature Birth/mortality , Premature Birth/pathology , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , Survival Rate , Ultrasonography, Doppler
6.
Hypertension ; 68(5): 1308-1313, 2016 11.
Article in English | MEDLINE | ID: mdl-27698062

ABSTRACT

Preeclamptic women produce agonistic autoantibodies to the angiotensin II type 1 receptor (AT1-AA) and exhibit increased blood pressure (mean arterial pressure), vascular sensitivity to angiotensin II (ANG II), and display a decrease in renal function. The objective of this study was to examine the renal hemodynamic changes during pregnancy in the presence of AT1-AAs with or without a slow pressor dose of ANG II. In this study, mean arterial pressure was elevated in all pregnant rats treated with ANG II with or without AT1-AA. Glomerular filtration rate was reduced from 1.90±0.16 mL/min in normal pregnant (NP) to 1.20±0.08 in ANG II+AT1-AA rats. Renal blood flow was decreased in ANG II+AT1-AA versus NP rats to 7.4±1.09 versus 15.4±1.75 mL/min. Renal vascular resistance was drastically increased between ANG II+AT1-AA versus NP rats (18.4±2.91 versus 6.4±0.77 mm Hg/mL per minute). Isoprostane excretion was increased by 3.5-fold in ANG II+AT1-AA versus NP (1160±321 versus 323±52 pg/mL). In conclusion, ANG II and AT1-AA together significantly decrease glomerular filtration rate by 37% and renal blood flow by 50% and caused a 3-fold increase in renal vascular resistance and isoprostane levels versus NP rats. These data indicate the importance of AT1-AAs to enhance ANG II-induced renal vasoconstriction and reduce renal function as mechanisms to cause hypertension as observed during preeclampsia.


Subject(s)
Autoantibodies/drug effects , Oxidative Stress/drug effects , Pregnancy, Animal , Receptor, Angiotensin, Type 1/drug effects , Receptor, Angiotensin, Type 1/immunology , Analysis of Variance , Angiotensin II/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Autoantibodies/immunology , Female , Glomerular Filtration Rate/drug effects , Humans , Hypertension, Pregnancy-Induced/drug therapy , Hypertension, Pregnancy-Induced/immunology , Kidney Function Tests , Oxidative Stress/physiology , Placental Circulation/drug effects , Placental Circulation/immunology , Pregnancy , Pregnancy Outcome , Rats
7.
Hypertension ; 68(4): 964-73, 2016 10.
Article in English | MEDLINE | ID: mdl-27550919

ABSTRACT

Uterine natural killer cells are important for uteroplacental development and pregnancy maintenance. Their role in pregnancy disorders, such as preeclampsia, is unknown. We reduced the number of natural killer cells by administering rabbit anti-asialo GM1 antiserum in an established rat preeclamptic model (female human angiotensinogen×male human renin) and evaluated the effects at the end of pregnancy (day 21), compared with preeclamptic control rats receiving normal rabbit serum. In 100% of the antiserum-treated, preeclamptic rats (7/7), we observed highly degenerated vessel cross sections in the mesometrial triangle at the end of pregnancy. This maternal uterine vasculopathy was characterized by a total absence of nucleated/living cells in the vessel wall and perivascularly and prominent presence of fibrosis. Furthermore, there were no endovascular trophoblast cells within the vessel lumen. In the control, normal rabbit serum-treated, preeclamptic rats, only 20% (1/5) of the animals displayed such vasculopathy. We confirmed the results in healthy pregnant wild-type rats: after anti-asialo GM1 treatment, 67% of maternal rats displayed vasculopathy at the end of pregnancy compared with 0% in rabbit serum-treated control rats. This vasculopathy was associated with a significantly lower fetal weight in wild-type rats and deterioration of fetal brain/liver weight ratio in preeclamptic rats. Anti-asialo GM1 application had no influence on maternal hypertension and albuminuria during pregnancy. Our results show a new role of natural killer cells during hypertensive pregnancy in maintaining vascular integrity. In normotensive pregnancy, this integrity seems important for fetal growth.


Subject(s)
Killer Cells, Natural/cytology , Placental Circulation/physiology , Pre-Eclampsia/physiopathology , Pregnancy, Animal , Trophoblasts/cytology , Analysis of Variance , Angiotensinogen/metabolism , Animals , Cell Movement/drug effects , Cell Movement/physiology , Female , Fetal Development/immunology , Fetal Development/physiology , Gestational Age , Interleukin-15/metabolism , Killer Cells, Natural/immunology , Placental Circulation/immunology , Pre-Eclampsia/metabolism , Pregnancy , Rats , Rats, Sprague-Dawley , Statistics, Nonparametric , Trophoblasts/metabolism
8.
Hum Immunol ; 76(4): 254-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25636573

ABSTRACT

Human leukocyte antigen (HLA)-G is a class Ib molecule with restricted tissue distribution expressed on trophoblast cells and has been proposed to have immunomodulatory functions during pregnancy. Soluble HLA-G1 (sHLA-G1) can be generated by the shedding of membrane-bound HLA-G molecules; however, three soluble isoforms also exist (HLA-G5 to -G6). During pregnancy, it is unknown whether there is a correlation between sHLA-G levels in maternal and fetal blood. In 246 pregnancies, we have measured the levels of sHLA-G1/-G5 in maternal blood plasma samples from gestational week 20 (GW20) and at term, as well as in umbilical cord blood samples. Soluble HLA-G levels declined by 38.4% in maternal blood from GW20 to term, and sHLA-G levels were significantly lower in maternal blood at term than in GW20 (P<0.001). At term, the sHLA-G levels were significantly higher in maternal blood than in umbilical blood (P<0.001). HLA-G levels in maternal blood in GW20 and at term, and in maternal blood at term and umbilical cord blood, were correlated (P<0.001 and P<0.01, respectively). This is the first large study simultaneously measuring sHLA-G in both maternal and umbilical cord blood. The finding that sHLA-G levels are significantly lower in fetal compared with maternal blood at term documents for the first time that sHLA-G is not freely transferred over the placental barrier. Soluble HLA-G levels in maternal and fetal blood were found to be correlated, which may be due to shared genetic factors of importance for production of sHLA-G in the mother and child, or it may support the theory that sHLA-G in the pregnant woman and the fetus is partly derived from a "shared organ", the placenta.


Subject(s)
Fetal Blood/metabolism , HLA-G Antigens/immunology , Placental Circulation/immunology , Pregnancy Trimester, Second/immunology , Cohort Studies , Female , Follow-Up Studies , Gestational Age , HLA-G Antigens/blood , Humans , Immunity, Maternally-Acquired , Pregnancy
9.
Am J Reprod Immunol ; 73(1): 79-90, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25263526

ABSTRACT

PROBLEM: To evaluate the inflammatory pattern in maternal circulation from women with preterm premature rupture of membranes (PPROM) considering the occurrence of histologically confirmed chorioamnionitis (HCA). METHOD OF STUDY: A prospective study was conducted in 121 women with PPROM between 24 and 34 + 0 weeks of gestation. Association between white blood cells (WBC) count, plasma CRP, IL-6, MCP-1 and IP-10 levels, and HCA was assessed. RESULTS: The rate of HCA was 44.7% (54/121). During the 5 days preceding delivery, median CRP, WBC, and IL-6 levels were significantly higher in the HCA than in no-HCA group (P < 0.001). Variations in IL-6, IP-10 levels, during the 24-72 hr before delivery, were predictors of the occurrence of HCA, but the diagnostic accuracy was low [Receiver Operating Characterictic (ROC) curve, area under the curve (AUC) = 0.56]. CONCLUSION: An increase in IL-6, CRP, IP-10 maternal plasma levels was confirmed in PPROM women with HCA. Longitudinal follow-up of these markers did not add valuable information regarding HCA.


Subject(s)
Amnion/pathology , Chorioamnionitis/diagnosis , Obstetric Labor, Premature/diagnosis , Rupture/diagnosis , Adult , Biomarkers/metabolism , C-Reactive Protein/metabolism , Chorioamnionitis/immunology , Cytokines/blood , Female , Follow-Up Studies , Humans , Inflammation Mediators/metabolism , Leukocyte Count , Obstetric Labor, Premature/immunology , Placental Circulation/immunology , Predictive Value of Tests , Pregnancy , Prognosis , Prospective Studies , Rupture/immunology
11.
J Reprod Immunol ; 101-102: 120-126, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24119981

ABSTRACT

Acute atherosis (Aa) affects uteroplacental spiral arteries in 20-40% of cases of preeclampsia. Its hallmark is lipid-filled, CD68-positive foam cells. It usually develops in the decidua (the pregnancy endometrium) at the distal ends of arteries that are often unremodelled in their proximal segments. Aa resembles the early stages of atherosclerosis, which becomes symptomatic in the middle-aged and elderly, in contrast to the young age of pregnant women with Aa. Although the mechanisms of Aa are largely unknown, they are likely to resemble those of early atherosclerosis, which is an inflammatory lesion of the arterial wall. However, Aa is likely to have added pregnancy-specific features. Because it also occurs in normotensive pregnancies, complicated by foetal growth restriction, diabetes mellitus or autoimmune disease or even without any complications, we suggest that Aa is the final manifestation of several inflammatory processes. We revisit an old proposition that immunological incompatibility between mother and foetus may sometimes induce Aa. We propose that excessive inflammatory activation, of other aetiologies, primarily in the decidua basalis, may explain the different ways in which Aa occurs. We speculate that the subset of women who develop these lesions may be at an increased risk of atherosclerotic arterial disease later in life. We hypothesise that use of anti-atherogenic statins during established preeclampsia may ameliorate Aa, improve uteroplacental perfusion and enhance pregnancy outcome.


Subject(s)
Atherosclerosis/immunology , Decidua/immunology , Histocompatibility, Maternal-Fetal/immunology , Pre-Eclampsia/immunology , Acute Disease , Atherosclerosis/drug therapy , Decidua/drug effects , Female , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Inflammation Mediators/antagonists & inhibitors , Placental Circulation/drug effects , Placental Circulation/immunology , Pre-Eclampsia/drug therapy , Pregnancy , Pregnancy Outcome , Risk
12.
Hum Immunol ; 74(7): 872-7, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23619469

ABSTRACT

Studies of maternal-fetal tolerance focus on defining mechanisms for establishment of immunological privilege within the uterus during pregnancy. Fetal trophoblasts play a key role in maternal tolerance, in part through cytokines production. As a novel inhibitory cytokine, IL-35 is produced by Foxp3(+) regulatory T cells (Tregs) and mediates maximal suppression of Tregs. The purpose of the study is to analyze the expression of IL-35 in first-trimester human placental trophoblasts. IL-35 expression was detected at both protein and mRNA levels by immunohistochemical staining and quantitative real-time PCR method, respectively and secretion of IL-35 was measured by ELISA assay. Our results demonstrated that human trophoblasts constitutively expressed IL-35. Ebi3 and p35 (two subunits of IL-35) mRNA was shown to be co-expressed in trophoblast cells. Moreover, large amounts of secreted IL-35 were detected in the supernatants of trophoblast cells. But we did not detect the constitutive expression of IL-35 in decidual stromal cells. Our findings confirmed for the first time that first-trimester human trophoblast cells expressed and secreted IL-35, which might contribute to their suppressive capacity to maternal immune cells. Therefore, IL-35 may be an important factor of the cytokine network regulating local immune responses during human pregnancy.


Subject(s)
Immune Tolerance , Interleukin-12 Subunit p35/metabolism , Interleukins/metabolism , Stromal Cells/immunology , T-Lymphocytes, Regulatory/immunology , Trophoblasts/immunology , Cells, Cultured , Female , Forkhead Transcription Factors/metabolism , Humans , Interleukin-12 Subunit p35/genetics , Interleukins/genetics , Minor Histocompatibility Antigens , Placental Circulation/immunology , Pregnancy , Pregnancy Trimester, First
13.
J Reprod Immunol ; 97(1): 20-6, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23432868

ABSTRACT

This mini-review summarizes the results of recent transcriptome studies of bovine endometrium during the estrous cycle and during the pre-implantation phase, with a focus on immune response genes. Gene expression changes in the bovine endometrium during the estrous cycle were compared to a similar study in equine endometrium. The results indicate species-specific expression patterns, particularly for genes with immune functions. These are presumably the consequence of adaptations to differences in the physiology of reproduction in each species, including development of the conceptus, hormone profiles during the estrous cycle, and insemination. The results from a number of transcriptome studies during the pre-implantation phase, as well as comparison to the effects of human interferon alpha on bovine endometrial gene expression, suggest that during pregnancy there is no general suppression of the maternal immune system, but rather a fine-tuned regulation of immune cells. This presumably facilitates tolerance to the immunologically 'foreign' conceptus and at the same time activation of the immune system to defend against microbial and viral infections. Furthermore, comparison of differentially expressed genes in bovine endometrium to similar studies in human endometrial samples reveals a number of similar changes, indicating the existence of shared mechanisms in preparation for embryo implantation.


Subject(s)
Endometrium/immunology , Estrous Cycle/immunology , Maternal-Fetal Exchange/immunology , Pregnancy, Animal/immunology , Animals , Cattle , Embryo Implantation/genetics , Embryo Implantation/immunology , Estrous Cycle/genetics , Female , Humans , Maternal-Fetal Exchange/genetics , Placental Circulation/immunology , Pregnancy , Pregnancy, Animal/genetics , Transcriptome/genetics , Transplantation Tolerance
14.
J Reprod Immunol ; 97(1): 36-42, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23432870

ABSTRACT

Viviparity has many evolutionary advantages but brings with it the problem of the semi-allogeneic foetus having to coexist with the mother for the duration of pregnancy. In species with haemochorial placentation this problem is particularly evident as foetal trophoblast cells are extensively intermingled with maternal tissue and are directly exposed to maternal blood. Fascinating adaptations on both the foetal and maternal side have allowed for this interaction to be re-directed away from an immune rejection response not only towards immunotolerance, but in fact towards actively supporting reproductive success. Recent data have shown that some of these remarkable adaptations are conserved between mice and humans. Thus, a subset of trophoblast cells that is directly exposed to the maternal uterine environment shares the feature of expressing an unusual antigen repertoire on their surface. Paternal antigens can be recognized by maternal immune cells, in particular uterine natural killer cells that express cognate receptors, to regulate the extensive remodelling events that take place at the implantation site. Detailed genetic dissection experiments in the mouse have further demonstrated the direct impact of antigenic dissimilarity on foetal growth. With the availability of inbred strains, in vitro culture systems of trophoblast stem cells, and in-depth genetic, genomic and epigenomic data the mouse will be a valuable model system to study the intricate immune crosstalk at the foeto-maternal boundary. These insights will pave the way towards unravelling the mutual and synergistic interactions between trophoblast and its surrounding maternal environment, and in doing so help understand pregnancy pathologies.


Subject(s)
Homeostasis , Immune System , Killer Cells, Natural/immunology , Maternal-Fetal Exchange/immunology , Reproduction/immunology , Trophoblasts/immunology , Animals , Biological Evolution , Cell Communication , Female , Humans , Mice , Placental Circulation/immunology , Pregnancy/immunology , Transplantation Tolerance
15.
J Cell Physiol ; 228(1): 142-8, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22566240

ABSTRACT

Increased vascular resistance in the fetoplacental circulation is a characteristic of preeclampsia. However, the potential molecular mechanisms of this condition remain obscure. The current study aimed to determine the direct effect of the peptide antigen corresponding to the second extracellular loop of the angiotensin II type 1 receptor (AT1R-EC(II) ) activating autoantibody (AT1-AA), a novel risk factor in preeclamptic patients, on fetoplacental villus stem blood vessels. Immunohistochemistry revealed that AT1 receptors were localized in the veins and arteries of human placental villi. Among 58 serum samples from preeclamptic patients, 28 (48.28%) were proved AT1-AA-positive by enzyme-linked immunosorbent assay [P<0.01 vs. 2/51 (3.92%) in the normal pregnancy group]. Total IgGs purified from AT1-AA-positive patients' sera (AT1-AA-IgGs) were added to isolated normal human placental blood vessels. The IgG significantly constricted both the villus veins and arteries in a dose-dependent manner in vitro, which could be blocked by the peptide corresponding to the human AT1R-EC(II) , anti-human IgG or the AT1 receptor antagonist losartan. Additionally, the venous constriction induced by AT1-AA-IgGs remained unchanged even at the end of the experiment (about half an hour), but the vasoconstriction caused by the AT1 receptor agonist angiotensin II underwent desensitization within three minutes. Collectively, our results demonstrated that AT1-AA in preeclamptic sera can directly constrict fetoplacental villus blood vessels without desensitization via the AT1 receptor in vitro, which might contribute to poor fetoplacental perfusion in preeclampsia.


Subject(s)
Autoantibodies/adverse effects , Placental Circulation/immunology , Pre-Eclampsia/metabolism , Receptor, Angiotensin, Type 1/immunology , Vasoconstriction/immunology , Adult , Autoantibodies/blood , Autoantibodies/immunology , Female , Humans , Immunoglobulin G/blood , Immunoglobulin G/isolation & purification , Placenta/blood supply , Pre-Eclampsia/immunology , Pregnancy , Receptor, Angiotensin, Type 2/immunology , Tissue Culture Techniques , Young Adult
16.
J Reprod Immunol ; 94(2): 131-41, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22542910

ABSTRACT

Interactions between maternal immune cells and the placenta are of substantial interest since diseases of pregnancy, such as recurrent miscarriage, villitis of unknown etiology and preeclampsia may arise due to inadequate adaptation of the maternal immune system. During normal pregnancy trophoblast debris is shed from the placenta into the maternal blood in large quantities. This trophoblast debris is then rapidly cleared from the maternal circulation. In this study, we exposed trophoblast debris generated from an in vitro placental explant model to peripheral blood-derived macrophages and quantified a variety of molecules that are important in immune responses by ELISA or flow cytometry. Phagocytosis of trophoblast debris resulted in reduced cell-surface expression of MHC-II molecules, the costimulatory molecules (CD80, CD86, CD40 and B7H3), monocyte chemoattractant protein-1 (MCP-1), inter-cellular adhesion molecule 1 (ICAM-1) and IL-8 receptors in macrophages while the expression of programmed death-1 ligand 1 (PD-L1) was upregulated. In addition, phagocytosis of trophoblast debris induced the secretion of the anti-inflammatory cytokines IL-10, IL6 and IL1Ra and decreased the secretion of pro-inflammatory cytokines IL-1ß, IL12p70 and IL-8 by macrophages. Phagocytosis of trophoblast debris also increased macrophage expression of the immunosuppressive enzyme indoleamine 2,3-dioxygenase (IDO). We have shown that phagocytosis of trophoblast debris from normal placentae alters the phenotype of macrophages such that they are likely to deviate maternal immune responses towards tolerance and away from inflammation. This may be one of the mechanisms that allow the human fetal allograft to survive in direct contact with the maternal immune system.


Subject(s)
Cellular Structures/immunology , Fetus/immunology , Macrophages/immunology , Transplantation Tolerance , Transplantation, Homologous/immunology , Trophoblasts/immunology , Antigens, CD/genetics , Antigens, CD/metabolism , Cells, Cultured , Cellular Structures/cytology , Cellular Structures/metabolism , Cytokines/genetics , Cytokines/metabolism , Female , Gene Expression Regulation, Developmental/immunology , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Humans , Immune Tolerance , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Inflammation Mediators/metabolism , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Placental Circulation/immunology , Pregnancy , Trophoblasts/cytology , Trophoblasts/metabolism
17.
Iran J Immunol ; 8(4): 201-8, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22201617

ABSTRACT

BACKGROUND: Repeated Implantation Failure (RIF) is one of the most intricate obstacles in assisted reproduction. The cytokine and chemokine composition of uterine cavity seem to play important roles in the implantation process. OBJECTIVE: To compare the cytokine profile in the endometrium of normal fertile women and those with repeated implantation failure. METHODS: After enzymatic digestion of endometrial tissues, whole endometrial cells and endometrial stromal cells from RIF and normal fertile women were cultivated and stimulated for cytokine secretion. The levels of IL-10, TGF-ß, IFN-γ, IL-6, IL-8 and IL-17 in culture supernatants of the two groups were assayed by ELISA and compared together. RESULTS: Endometrial stromal cells and whole endometrial cells of normal fertile women produced higher levels of IL-6, IL-8 and TGF-ß compared to RIF group, although this difference was statistically significant only in endometrial stromal cells (p=0.005, 0.002 and 0.001, respectively). In addition, endometrial stromal cells of normal fertile women produced lower levels of IL-10 in comparison with RIF group (p=0.005). CONCLUSION: Disturbances in cytokine production at the feto-maternal interface could be a cause of implantation failure. A pro-inflammatory cytokine milieu seems to be pivotal for successful implantation.


Subject(s)
Embryo Implantation/immunology , Endometrium/metabolism , Fertilization in Vitro , Infertility, Female/immunology , Stromal Cells/immunology , Adult , Cells, Cultured , Cytokines/metabolism , Endometrium/immunology , Endometrium/pathology , Enzyme-Linked Immunosorbent Assay , Female , Fertility/immunology , Humans , Infertility, Female/therapy , Placental Circulation/immunology , Pregnancy , Th1-Th2 Balance , Treatment Failure
18.
J Reprod Immunol ; 89(1): 18-25, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21470690

ABSTRACT

The placenta forms the interface between the mother and the fetus. During placental development cytotrophoblasts differentiate to form the syncytium or to invade the decidual wall to breach maternal vessels and establish the blood flow in the intervillous space. This process is still not well understood but it is proposed that chemokines and their receptors are involved in guiding cytotrophoblasts to the decidua and maternal vessels as well as attracting immunocompetent cells to the implantation site. CXCL12 is a chemokine expressed by cytotrophoblasts and is involved in cytotrophoblast invasion, differentiation and survival. One of its receptors, CXCR4, has been detected on cytotrophoblasts. Recent data show that CXCR7 and syndecan-4 might partially mediate CXCL12 function in other cell types. In this study, we examined CXCR7 and syndecan-4 expression at the maternal-fetal interface via immmunolocalization in placental tissue sections and in isolated cytotrophoblasts. We further used immunoblot analyses to confirm the data. We were able to show that cytotrophoblasts express both receptors and that upregulation occurs during the differentiation process of cytotrophoblasts towards the invasive phenotype. On a functional level CXCR7 seems not to be involved in JAR cell chemotaxis, suggesting a different function of this receptor. In conclusion, we propose that CXCL12 binds to CXCR4, but also to CXCR7 and syndecan-4. These three receptors could mediate different functions of CXCL12, such as cell migration, directed invasion, proliferation and survival. The latter molecules might also be involved in the development of placental pathologies, such as preeclampsia or choriocarcinoma growth.


Subject(s)
Chemokine CXCL12/metabolism , Placenta/metabolism , Receptors, CXCR/metabolism , Syndecan-4/metabolism , Trophoblasts/metabolism , Cell Differentiation , Cell Line, Tumor , Chemokine CXCL12/blood , Female , Humans , Immunohistochemistry , Placenta/cytology , Placenta/immunology , Placental Circulation/immunology , Placentation/immunology , Pregnancy , Pregnancy Trimesters , Receptors, CXCR/genetics , Syndecan-4/genetics , Trophoblasts/cytology , Trophoblasts/immunology , Up-Regulation
19.
J Reprod Immunol ; 88(2): 112-7, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21277024

ABSTRACT

There is a strong clinical association between intrauterine infections and pregnancy complications, such as preterm labor. The placenta functions as an active barrier whereby the trophoblast recognizes microbes through pattern recognition receptors, such as the well characterized Toll-like receptors, in order to respond to pathogens at the maternal-fetal interface. Consequently, either an inefficient or overactive placental response to an infectious trigger, may have a significant impact on pregnancy outcome. Recently the placenta has been shown to express a newly identified family of pattern recognition receptors, the cytoplasmic-based Nod-like receptors (NLRs). As a result of their restricted localization, NLRs function as intracellular receptors that respond to infectious components, which have gained access to the cytoplasmic compartment. Thus, NLRs may provide the trophoblast with a recognition system that may be critical in placental responses to microorganisms or their cell wall components that have gained access to the cell's intracellular space, or that have evaded recognition by the TLRs. This review will discuss what is currently known about the role of NOD proteins, NALP proteins, and the inflammasome at the maternal-fetal interface, and their potential role in infection-associated pregnancy complications, like preterm labor. As we learn more about their function at the maternal-fetal interface, we will have a better understanding of their function in normal pregnancy and their potential to contribute to the pathogenesis of infection- and inflammation-associated pregnancy complications.


Subject(s)
Nod Signaling Adaptor Proteins/immunology , Pregnancy Complications, Infectious/immunology , Trophoblasts/metabolism , Animals , Female , Humans , Immunity, Innate , Inflammasomes/metabolism , Placental Circulation/immunology , Pregnancy , Trophoblasts/immunology
20.
Cell Mol Immunol ; 8(1): 1-11, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20711229

ABSTRACT

Natural killer (NK) cells are found in lymphoid and non-lymphoid organs. In addition to important roles in immune surveillance, some NK cells contribute to angiogenesis and circulatory regulation. The uterus of early pregnancy is a non-lymphoid organ enriched in NK cells that are specifically recruited to placental attachment sites. In species with invasive hemochorial placentation, these uterine natural killer (uNK) cells, via secretion of cytokines, chemokines, mucins, enzymes and angiogenic growth factors, contribute to the physiological change of mesometrial endometrium into the unique stromal environment called decidua basalis. In humans, uNK cells have the phenotype CD56(bright)CD16(dim) and they appear in great abundance in the late secretory phase of the menstrual cycle and early pregnancy. Gene expression studies indicate that CD56(bright)CD16(dim) uterine and circulating cells are functionally distinct. In humans but not mice or other species with post-implantation decidualization, uNK cells may contribute to blastocyst implantation and are of interest as therapeutic targets in female infertility. Histological and genetic studies in mice first identified triggering of the process of gestation spiral arterial modification as a major uNK cell function, achieved via interferon (IFN)-γ secretion. During spiral arterial modification, branches from the uterine artery that traverse the endometrium/decidua transiently lose their muscular coat and ability to vasoconstrict. The expression of vascular markers changes from arterial to venous as these vessels dilate and become low-resistance, high-volume channels. Full understanding of the vascular interactions of human uNK cells is difficult to obtain because endometrial time-course studies are not possible in pregnant women. Here we briefly review key information concerning uNK cell functions from studies in rodents, summarize highlights concerning human uNK cells and describe our preliminary studies on development of a humanized, pregnant mouse model for in vivo investigations of human uNK cell functions.


Subject(s)
Killer Cells, Natural , Morphogenesis , Neovascularization, Physiologic , Uterine Artery/immunology , Animals , CD56 Antigen/immunology , Cytokines/immunology , Embryo Implantation/immunology , Female , Fetus/blood supply , Fetus/immunology , GPI-Linked Proteins/immunology , Humans , Killer Cells, Natural/immunology , Mice , Models, Animal , Morphogenesis/immunology , Neovascularization, Physiologic/immunology , Placental Circulation/immunology , Pregnancy , Rats , Receptors, IgG/immunology , Uterine Artery/cytology , Uterine Artery/embryology , Uterus/blood supply , Uterus/embryology , Uterus/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...