Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Am J Obstet Gynecol ; 227(4): 641.e1-641.e13, 2022 10.
Article in English | MEDLINE | ID: mdl-35863458

ABSTRACT

BACKGROUND: The extravillous trophoblast expresses each of the nonclassical major histocompatibility complex class I antigens-human leukocyte antigens E, F, and G-and a single classical class I antigen, human leukocyte antigen C. We recently demonstrated dynamic expression patterns of human leukocyte antigens C, G, and F during early extravillous trophoblast invasion and placentation. OBJECTIVE: This study aimed to investigate the hypothesis that the immune inflammatory mediated complications of pregnancy such as early preeclampsia and preterm labor may show altered expression profiles of nonclassical human leukocyte antigens. STUDY DESIGN: Real-time quantitative polymerase chain reaction, western blot, and immunohistochemistry were performed on placental villous tissues and basal plate sections from term nonlaboring deliveries, preterm deliveries, and severe early-onset preeclampsia, both with and without small-for-gestational-age neonates. RESULTS: Human leukocyte antigen G is strongly and exclusively expressed by the extravillous trophoblast within the placental basal plate, and its levels increase in pregnancies complicated by severe early-onset preeclampsia with small-for-gestational-age neonates relative to those of healthy term controls. Human leukocyte antigen C shows a similar profile in the extravillous trophoblast of preeclamptic pregnancies, but significantly decreases in the villous placenta. Human leukocyte antigen F protein levels are decreased in both extravillous trophoblast and villous placenta of severe early-onset preeclamptic pregnancies, both with and without small-for-gestational-age neonates, compared with those found in term and preterm birth deliveries. Human leukocyte antigen E decreases in blood vessels in placentas from preeclamptic pregnancies relative to its levels in term and preterm birth deliveries. Placental levels of human leukocyte antigens F and C are increased in cases of preterm birth with chorioamnionitis relative to those of cases of idiopathic preterm birth. CONCLUSION: Dysregulation of placental human leukocyte antigen expression at the maternal-fetal interface may contribute to compromised maternal tolerance in preterm birth with chorioamnionitis and excessive maternal systemic inflammation associated with severe early-onset preeclampsia.


Subject(s)
Chorioamnionitis , Pre-Eclampsia , Premature Birth , Chorioamnionitis/metabolism , Female , Fetal Growth Retardation/metabolism , HLA-C Antigens/metabolism , HLA-G Antigens/metabolism , Histocompatibility Antigens Class I , Humans , Infant, Newborn , Placenta/metabolism , Placentation , Pre-Eclampsia/metabolism , Pregnancy , Premature Birth/metabolism , Trophoblasts/metabolism , HLA-E Antigens
2.
Lancet HIV ; 9(2): e120-e129, 2022 02.
Article in English | MEDLINE | ID: mdl-34863352

ABSTRACT

The use of antiretroviral therapy (ART) in pregnancy is important for maternal health, and has been successful in reducing vertical transmission rates to almost zero in those taking effective ART regimens with good adherence. However, there are reports of higher rates of low birthweight and preterm births in women with HIV, which can be further exacerbated by ART usage in pregnancy. Protease inhibitors, and ritonavir-boosted lopinavir in particular, might directly contribute to placental and uteroplacental pathology in part by altering plasma concentrations of the essential steroid hormones of pregnancy, progesterone and oestradiol. In this Review, we collate the increasing evidence of dysregulated maternal endocrinology, reproductive physiology, and placental compromise associated with protease inhibitors. Based on findings of placental and decidual effects, we recommend that ritonavir-boosted lopinavir should be avoided in pregnancy, in line with US and European guidelines. Long-term follow-up of children exposed to protease inhibitors in utero is also recommended.


Subject(s)
Anti-HIV Agents , HIV Infections , HIV Protease Inhibitors , Anti-HIV Agents/therapeutic use , Child , Decidua , Female , HIV Infections/drug therapy , HIV Protease Inhibitors/adverse effects , Humans , Infant, Newborn , Lopinavir/adverse effects , Placenta , Pregnancy , Progesterone , Protease Inhibitors/therapeutic use , Ritonavir/adverse effects
3.
Cells ; 10(7)2021 07 08.
Article in English | MEDLINE | ID: mdl-34359894

ABSTRACT

COVID-19 is associated with increased incidence of preterm birth (PTB). We assessed pathways by which SARS-CoV-2 could access the placenta. Placentae, from PTB with or without chorioamnionitis (ChA), or from term pregnancies (n = 12/13/group) were collected. Peripheral blood was collected from healthy pregnant women (n = 6). Second trimester placental explants (16-20 weeks, n = 5/group) were treated with lipopolysaccharide (LPS, to mimic bacterial infection) and ACE2, CCL2, IL-6/8 and TNFα mRNA was assessed. ChA-placentae exhibited increased ACE2 and CCL2 mRNA expression (p < 0.05). LPS increased cytokine and ACE2 mRNA in placental explants. Placental ACE2 protein localized to syncytiotrophoblast, fetal endothelium, extravillous trophoblast and in immune cells-subsets (M1/M2 macrophage and neutrophils) within the villous stroma. Significantly increased numbers of M1 macrophage and neutrophils were present in the ChA-placenta (p < 0.001). Subsets of peripheral immune cells from pregnant women express the ACE2 mRNA and protein. A greater fraction of granulocytes was positive for ACE2 protein expression compared to lymphocytes or monocytes. These data suggest that in pregnancies complicated by ChA, ACE2 positive immune cells in the maternal circulation have the potential to traffic SARS-CoV-2 virus to the placenta and increase the risk of vertical transmission to the placenta/fetus.


Subject(s)
Angiotensin-Converting Enzyme 2/genetics , Gene Expression , Placenta/metabolism , Pregnancy Complications, Infectious/genetics , Premature Birth/etiology , Adult , COVID-19/genetics , COVID-19/transmission , Female , Humans , Infant, Newborn , Infectious Disease Transmission, Vertical , Lymphocytes/metabolism , Monocytes/metabolism , Placenta/cytology , Pregnancy , Premature Birth/genetics , SARS-CoV-2/isolation & purification
4.
Hypertension ; 77(2): 475-490, 2021 02.
Article in English | MEDLINE | ID: mdl-33356399

ABSTRACT

Revaluation of the association of the STOX1 (STORKHEAD_BOX1 PROTEIN 1) transcription factor mutation (Y153H, C allele) with the early utero-vascular origins of placental pathology is warranted. To investigate if placental STOX1 Y153H genotype affects utero-vascular remodeling-compromised in both preterm birth and preeclampsia-we utilized extravillous trophoblast (EVT) explant and placental decidual coculture models, transfection of STOX1 wild-type and mutant plasmids into EVT-like trophoblast cell lines, and a cohort of 75 placentas from obstetric pathologies. Primary EVT and HTR8/SVneo cells carrying STOX1 Y153H secreted lower levels of IL (interleukin) 6, and IL-8, and higher CXCL16 (chemokine [C-X-C motif] ligand 16) and TRAIL (tumor necrosis factor-related apoptosis-inducing ligand) than wild-type EVT and Swan71 cells. Media from wild-type EVT or Swan71 cells transfected with wild-type STOX1 stimulated: endothelial chemokine expression, angiogenesis, and decidual natural killer cell and monocyte migration. In contrast, Y153H EVT conditioned medium, Swan71 transfected with the Y153H plasmid, or HTR8/SVneo media had no effect. Genotyping of placental decidual cocultures demonstrated association of the placental STOX1 CC allele with failed vascular remodeling. Decidual GG NODAL R165H increased in failed cocultures carrying the placental CC alleles of STOX1. Multivariate analysis of the placental cohort showed that the STOX1 C allele correlated with premature birth, with or without severe early-onset preeclampsia, and small for gestational age babies. In conclusion, placental STOX1 Y153H is a precipitating factor in preterm birth and placental preeclampsia due to defects in early utero-placental development.


Subject(s)
Carrier Proteins/metabolism , Placenta/metabolism , Placentation/genetics , Pre-Eclampsia/metabolism , Premature Birth/metabolism , Trophoblasts/metabolism , Adolescent , Adult , Alleles , Carrier Proteins/genetics , Cell Line , Female , Humans , Infant, Newborn , Interleukin-6/metabolism , Interleukin-8/metabolism , Pre-Eclampsia/genetics , Pregnancy , Pregnancy Trimester, First/metabolism , Premature Birth/genetics , Young Adult
5.
Am J Obstet Gynecol ; 224(3): 298.e1-298.e8, 2021 03.
Article in English | MEDLINE | ID: mdl-32853537

ABSTRACT

BACKGROUND: Although there is some evidence that severe acute respiratory syndrome coronavirus 2 can invade the human placenta, limited data exist on the gestational age-dependent expression profile of the severe acute respiratory syndrome coronavirus 2 cell entry mediators, angiotensin-converting enzyme 2 and transmembrane protease serine 2, at the human maternal-fetal interface. There is also no information as to whether the expression of these mediators is altered in pregnancies complicated by preeclampsia or preterm birth. This is important because the expression of decidual and placental angiotensin-converting enzyme 2 and transmembrane protease serine 2 across gestation may affect the susceptibility of pregnancies to vertical transmission of severe acute respiratory syndrome coronavirus 2. OBJECTIVE: This study aimed to investigate the expression pattern of specific severe acute respiratory syndrome coronavirus 2 cell entry genes, angiotensin-converting enzyme 2 and transmembrane protease serine 2, in the placenta across human pregnancy and in paired samples of decidua and placenta in pregnancies complicated by preterm birth or preeclampsia compared with those in term uncomplicated pregnancies. STUDY DESIGN: In this study, 2 separate cohorts of patients, totaling 87 pregnancies, were included. The first cohort was composed of placentae from first- (7-9 weeks), second- (16-18 weeks), and third-trimester preterm (26-31 weeks) and third-trimester term (38-41 weeks) pregnancies (n=5/group), whereas the second independent cohort included matched decidua and placentae from pregnancies from term uncomplicated pregnancies (37-41 weeks' gestation; n=14) and pregnancies complicated by preterm birth (26-37 weeks' gestation; n=11) or preeclampsia (25-37 weeks' gestation; n=42). Samples were subjected to quantitative polymerase chain reaction and next-generation sequencing or RNA sequencing for next-generation RNA sequencing for angiotensin-converting enzyme 2 and transmembrane protease serine 2 mRNA expression quantification, respectively. RESULTS: In the first cohort, angiotensin-converting enzyme 2 and transmembrane protease serine 2, exhibited a gestational age-dependent expression profile, that is, angiotensin-converting enzyme 2 and transmembrane protease serine 2 mRNA was higher (P<.05) in the first-trimester placenta than in second-trimester, preterm birth, and term placentae (P<.05) and exhibited a negative correlation with gestational age (P<.05). In the second cohort, RNA sequencing demonstrated very low or undetectable expression levels of angiotensin-converting enzyme 2 in preterm birth, preeclampsia, and term decidua and in placentae from late gestation. In contrast, transmembrane protease serine 2 was expressed in both decidual and placental samples but did not change in pregnancies complicated by either preterm birth or preeclampsia. CONCLUSION: The increased expression of these severe acute respiratory syndrome coronavirus 2 cell entry-associated genes in the placenta in the first trimester of pregnancy compared with those in later stages of pregnancy suggests the possibility of differential susceptibility to placental entry to severe acute respiratory syndrome coronavirus 2 across pregnancy. Even though there is some evidence of increased rates of preterm birth associated with severe acute respiratory syndrome coronavirus 2 infection, we found no increase in mRNA expression of angiotensin-converting enzyme 2 or transmembrane protease serine 2 at the maternal-fetal interface.


Subject(s)
Angiotensin-Converting Enzyme 2/genetics , COVID-19/etiology , Placenta/virology , Pre-Eclampsia/metabolism , Premature Birth/metabolism , SARS-CoV-2/physiology , Serine Endopeptidases/genetics , Female , Humans , Placenta/metabolism , Pregnancy , RNA, Messenger/analysis , Virus Internalization
6.
Mol Hum Reprod ; 26(11): 850-865, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32898274

ABSTRACT

A critical component of early human placental development includes migration of extravillous trophoblasts (EVTs) into the decidua. EVTs migrate toward and displace vascular smooth muscle cells (SMCs) surrounding several uterine structures, including spiral arteries. Shallow trophoblast invasion features in several pregnancy complications including preeclampsia. Maternal obesity is a risk factor for placental dysfunction, suggesting that factors within an obese environment may impair early placental development. Herein, we tested the hypothesis that palmitic acid, a saturated fatty acid circulating at high levels in obese women, induces an inflammatory response in EVTs that hinders their capacity to migrate toward SMCs. We found that SMCs and SMC-conditioned media stimulated migration and invasion of an EVT-like cell line, HTR8/SVneo. Palmitic acid impaired EVT migration and invasion toward SMCs, and induced expression of several vasoactive and inflammatory mediators in EVTs, including endothelin, interleukin (IL)-6, IL-8 and PAI1. PAI1 was increased in plasma of women with early-onset preeclampsia, and PAI1-deficient EVTs were protected from the anti-migratory effects of palmitic acid. Using first trimester placental explants, palmitic acid exposure decreased EVT invasion through Matrigel. Our findings reveal that palmitic acid induces an inflammatory response in EVTs and attenuates their migration through a mechanism involving PAI1. High levels of palmitic acid in pathophysiological situations like obesity may impair early placental development and predispose to placental dysfunction.


Subject(s)
Cell Movement , Inflammation , Palmitic Acid/pharmacology , Plasminogen Activator Inhibitor 1/physiology , Trophoblasts/physiology , Adult , Cell Movement/drug effects , Cell Movement/genetics , Cells, Cultured , Decidua/drug effects , Decidua/physiology , Female , HEK293 Cells , Humans , Inflammation/blood , Inflammation/chemically induced , Inflammation/pathology , Inflammation/physiopathology , Inflammation Mediators/blood , Inflammation Mediators/pharmacology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/physiology , Obesity/blood , Obesity/complications , Obesity/physiopathology , Palmitic Acid/blood , Placenta/cytology , Placenta/drug effects , Placentation/drug effects , Placentation/physiology , Plasminogen Activator Inhibitor 1/blood , Pregnancy , Pregnancy Complications/blood , Pregnancy Complications/physiopathology , Trophoblasts/cytology , Trophoblasts/drug effects , Young Adult
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
8.
J Cell Mol Med ; 23(1): 610-618, 2019 01.
Article in English | MEDLINE | ID: mdl-30407748

ABSTRACT

The ATP-binding cassette (ABC) transporters control placental transfer of several nutrients, steroids, immunological factors, chemicals, and drugs at the maternal-fetal interface. We and others have demonstrated a gestational age-dependent expression pattern of two ABC transporters, P-glycoprotein and breast cancer resistance protein throughout pregnancy. However, no reports have comprehensively elucidated the expression pattern of all 50 ABC proteins, comparing first trimester and term human placentae. We hypothesized that placental ABC transporters are expressed in a gestational-age dependent manner in normal human pregnancy. Using the TaqMan® Human ABC Transporter Array, we assessed the mRNA expression of all 50 ABC transporters in first (first trimester, n = 8) and third trimester (term, n = 12) human placentae and validated the resulting expression of selected ABC transporters using qPCR, Western blot and immunohistochemistry. A distinct gene expression profile of 30 ABC transporters was observed comparing first trimester vs. term placentae. Using individual qPCR in selected genes, we validated the increased expression of ABCA1 (P < 0.01), ABCA6 (P < 0.001), ABCA9 (P < 0.001) and ABCC3 (P < 0.001), as well as the decreased expression of ABCB11 (P < 0.001) and ABCG4 (P < 0.01) with advancing gestation. One important lipid transporter, ABCA6, was selected to correlate protein abundance and characterize tissue localization. ABCA6 exhibited increased protein expression towards term and was predominantly localized to syncytiotrophoblast cells. In conclusion, expression patterns of placental ABC transporters change as a function of gestational age. These changes are likely fundamental to a healthy pregnancy given the critical role that these transporters play in the regulation of steroidogenesis, immunological responses, and placental barrier function and integrity.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Placenta/metabolism , Transcriptome/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Adult , Female , Gene Expression Profiling/methods , Gestational Age , Humans , Neoplasm Proteins/genetics , Pregnancy , Trophoblasts/metabolism
9.
Am J Reprod Immunol ; 81(1): e13054, 2019 01.
Article in English | MEDLINE | ID: mdl-30267451

ABSTRACT

PROBLEM: During pregnancy, the decidual spiral arterioles (SpAs) that supply maternal blood to the placenta undergo a series of changes to optimise the transfer of nutrients and oxygen to the developing foetus. Recent studies have shown that initiation of SpA transformation coincides with decidual leucocyte infiltration. Leucocytes are known to be a source of matrix metalloproteinases (MMPs); however, the complete profile of MMPs expressed by decidual NK cells (dNK) and macrophages has not been characterised. We hypothesised that leucocyte-derived MMPs contribute to SpA remodelling. METHODS: Decidual NK cells and macrophages were isolated from first trimester decidua and their MMP repertoire profiled by qRT-PCR (n = 10; 5-11 weeks). Dual immunofluorescence was used to localise MMP expression in situ (n = 3; 5-12 weeks). Gelatin zymography was carried out to assess whether leucocyte-derived MMPs can degrade ECM. In situ zymography and immunofluorescence identified MMP activity in tissue-resident dNK and macrophages. RESULTS: Decidual NK cells cells and macrophages expressed MMP2, -7, -9, -11, -16, -19 and tissue inhibitors of metalloproteinase-1, -2, and -3. Both cell types degraded gelatin using MMP2 and MMP9 and broke down collagen in an in vitro model of the SpA. Extravillous trophoblasts (EVTs) expressed a similar repertoire of MMPs. CONCLUSION: We suggest that matrix remodelling in SpA is initiated by infiltrating leucocytes, while EVTs become involved at later stages.


Subject(s)
Arterioles/physiology , Decidua/pathology , Killer Cells, Natural/physiology , Macrophages/physiology , Placenta/physiology , Trophoblasts/physiology , Cell Differentiation , Cell Movement , Cells, Cultured , Extracellular Matrix/metabolism , Female , Humans , Matrix Metalloproteinases/metabolism , Pregnancy , Proteolysis
10.
EBioMedicine ; 39: 531-539, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30579870

ABSTRACT

BACKGROUND: Decidual natural killer (dNK) cells are the predominant lymphocytes accumulated at the maternal-fetal interface. Regulatory mechanism of dNK cells in preeclampsia, a gestational complication characterized by high blood pressure and increased proteinuria occurring after 20 weeks pregnancy, is not completely understood. METHODS: Multi-parameter flow cytometry is applied to investigate the phenotype and function of dNK cells freshly isolated from decidual samples or conditionally cultured by TGFb stimulation. FINDINGS: In preeclampsia, we documented elevated numbers of CD56+ CD3- dNK cells in close proximity to Foxp3+ regulatory T (Treg) cells within the decidua. In vitro experiments using dNK cells from early gestation showed that dNK activation (IFNG, IL-8 and CD107a) can be downregulated by Treg cells. The expression of these markers by dNK cells was significantly lower in preeclampsia. We also observed a positive correlation between the expression of dNK activation receptors (NKp30 and NKG2D) and the expression of IFNG in specific dNK subsets. TGFb levels are increased in the decidua of preeclamptic pregnancies. We analyzed co-expression of activation (IFNG/IL-8/CD107a) and angiogenic (VEGF) markers in dNK cells. TGFb treatment reduced while blockade of TGFb increased co-expression of these markers. INTERPRETATION: Our findings suggest that elevated decidual TGFb1 supresses the activation of specific subsets of dNK which in turn contributes to the uteroplacental pathology associated with the onset of preeclampsia.


Subject(s)
Decidua/immunology , Killer Cells, Natural/immunology , Pre-Eclampsia/immunology , Transforming Growth Factor beta1/metabolism , Cells, Cultured , Culture Media, Conditioned/chemistry , Decidua/metabolism , Down-Regulation , Female , Flow Cytometry , Humans , Interferon-gamma/metabolism , Interleukin-8/metabolism , Lysosomal-Associated Membrane Protein 1/metabolism , Pregnancy , Signal Transduction , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/pharmacology
11.
J Cell Mol Med ; 22(11): 5378-5393, 2018 11.
Article in English | MEDLINE | ID: mdl-30256530

ABSTRACT

Dysregulation of trophoblast differentiation is implicated in the placental pathologies of intrauterine growth restriction and pre-eclampsia. P-glycoprotein (P-gp encoded by ABCB1) is an ATP-binding cassette transporter present in the syncytiotrophoblast layer of the placenta where it acts as a molecular sieve. In this study, we show that P-gp is also expressed in the proliferating cytotrophoblast (CT), the syncytiotrophoblast (ST) and the extravillous trophoblast (EVT), suggesting our hypothesis of a functional role for P-gp in placental development. Silencing of ABCB1, via siRNA duplex, results in dramatically reduced invasion and migration, and increased tube formation and fusion in the EVT-like HTR8/SVneo cell line. In both EVT and CT explant differentiation experiments, silencing of ABCB1 leads to induction of the fusion markers human hCG, ERVW-1 and GJA1 and terminal differentiation of both trophoblast subtypes. Moreover, P-gp protein levels are decreased in both the villous and the EVT of severe early-onset pre-eclamptic placentas. We conclude that, in addition to its role as a syncytial transporter, P-gp is a key factor in the maintenance of both CT and EVT lineages and that its decrease in severe pre-eclampsia may contribute to the syncytial and EVT placental pathologies associated with this disease.


Subject(s)
Placentation/genetics , Pre-Eclampsia/genetics , Trophoblasts/metabolism , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B/genetics , Cell Differentiation/genetics , Cell Line , Cell Movement/genetics , Cell Proliferation/genetics , Female , Humans , Placenta/metabolism , Placenta/pathology , Pre-Eclampsia/metabolism , Pre-Eclampsia/pathology , Pregnancy , RNA, Small Interfering/genetics , Trophoblasts/pathology
12.
J Immunol ; 198(10): 4115-4128, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28396316

ABSTRACT

Decidual spiral arteriole (SpA) remodeling is essential to ensure optimal uteroplacental blood flow during human pregnancy, yet very little is known about the regulatory mechanisms. Uterine decidual NK (dNK) cells and macrophages infiltrate the SpAs and are proposed to initiate remodeling before colonization by extravillous trophoblasts (EVTs); however, the trigger for their infiltration is unknown. Using human first trimester placenta, decidua, primary dNK cells, and macrophages, we tested the hypothesis that EVTs activate SpA endothelial cells to secrete chemokines that have the potential to recruit maternal immune cells into SpAs. Gene array, real-time PCR, and ELISA analyses showed that treatment of endothelial cells with EVT conditioned medium significantly increased production of two chemokines, CCL14 and CXCL6. CCL14 induced chemotaxis of both dNK cells and decidual macrophages, whereas CXCL6 also induced dNK cell migration. Analysis of the decidua basalis from early pregnancy demonstrated expression of CCL14 and CXCL6 by endothelial cells in remodeling SpAs, and their cognate receptors are present in both dNK cells and macrophages. Neutralization studies identified IL-6 and CXCL8 as factors secreted by EVTs that induce endothelial cell CCL14 and CXCL6 expression. This study has identified intricate crosstalk between EVTs, SpA cells, and decidual immune cells that governs their recruitment to SpAs in the early stages of remodeling and has identified potential key candidate factors involved. This provides a new understanding of the interactions between maternal and fetal cells during early placentation and highlights novel avenues for research to understand defective SpA remodeling and consequent pregnancy pathology.


Subject(s)
Arterioles/physiology , Decidua/physiology , Endothelial Cells/metabolism , Killer Cells, Natural/physiology , Macrophages/physiology , Trophoblasts/metabolism , Arterioles/cytology , Arterioles/immunology , Cell Movement/immunology , Cells, Cultured , Chemokine CXCL6/biosynthesis , Chemokine CXCL6/immunology , Chemokines, CC/biosynthesis , Chemokines, CC/immunology , Culture Media/chemistry , Decidua/immunology , Endothelial Cells/drug effects , Endothelial Cells/immunology , Female , Humans , Interleukin-6/immunology , Interleukin-6/metabolism , Interleukin-8/immunology , Interleukin-8/metabolism , Macrophages/immunology , Placenta/cytology , Placenta/immunology , Pregnancy , Pregnancy Trimester, First , Trophoblasts/immunology
13.
Am J Reprod Immunol ; 77(6)2017 06.
Article in English | MEDLINE | ID: mdl-28185362

ABSTRACT

PROBLEM: The extravillous trophoblasts (EVT) express HLA-C and HLA-G, but HLA-E and HLA-F are the subject of conflicting reports. In this study, we define the HLA expression profile during active EVT placental implantation, pregnancy development, and parturition. METHOD OF STUDY: Immunohistochemistry, q-PCR, and Western blot were used to investigate HLA-C, HLA-E, and HLA-F placental expression across gestation from the early first trimester, late first trimester, second trimester (n=10 in each), preterm gestation (n=6) to elective term cesarean section and term vaginal deliveries (n=12, 38-41 weeks). EVT explants and Swan71 cells were used to assess HLA-C and HLA-F during active EVT migration. RESULTS: HLA-G, HLA-C, and HLA-F were expressed by 1st-trimester EVT and became intracellular and weaker as gestation progressed. HLA-E was only expressed in 1st-trimester placenta. HLA-F and HLA-C mRNA and protein expression levels showed a significant increase in the fetal villous mesenchyme across gestation. HLA-C levels increased with labor. We detected a 100-kDa HLA-F band in early pregnancy suggesting dimer formation on the EVT surface. These results were confirmed in EVT outgrowths and Swan71 trophoblast which showed that HLA-F and HLA-G are increased on the cell surface of migrating EVT, while HLA-C was internalized. CONCLUSION: Expression of HLA-F and HLA-G on the cell surface of actively migrating EVT supports their specific role in early EVT invasion and interactions with uterine natural killer cells. HLA-C's limited expression to the proliferative EVT suggests a protective role in the earliest events of implantation but not in active EVT invasion. We also show for the first time that HLA-C may be involved in parturition.


Subject(s)
Histocompatibility Antigens Class I/metabolism , Placenta/metabolism , Pregnancy/metabolism , Trophoblasts/metabolism , Adult , Cell Movement , Female , Histocompatibility Antigens Class I/genetics , Humans , Infant, Newborn , Male , Parturition/metabolism , Placentation , Pregnancy Trimesters/metabolism , RNA, Messenger/metabolism , Young Adult
14.
Cell Mol Immunol ; 14(2): 203-213, 2017 Feb.
Article in English | MEDLINE | ID: mdl-26277900

ABSTRACT

Decidual natural killer (dNK) cells express an array of activation receptors to regulate placental immunity and development during early pregnancy. We investigated the functional character of human dNK cells during the first and second trimester of gestation and the interaction between dNK and trophoblast cells. Although the frequency of CD56+CD16- dNK among the total CD45+ leukocytes did not change over this period, the expression of the activating receptors, NKp80 and NKG2D, was greatly upregulated. We observed a significantly higher number of extravillous trophoblast cells in proximity to the dNK cells in the first trimester in comparison with the second trimester decidua. NKG2D expression by first trimester dNK cells was decreased when co-cultured with the HTR-8 trophoblast cell line. In the second trimester, functional markers of dNK activation, i.e., angiogenic factor production (e.g., vascular endothelial growth factor, interleukin-8, interferon-gamma), remained stable despite an increase in NKp80 or NKG2D surface expression. Furthermore, the degranulation capacity of dNK cells, as assessed by CD107a, was decreased in the second trimester. We suggest that in the first trimester, trophoblast-dNK interactions generate a population of dNK cells with a suppressed activating phenotype. In the second trimester, the loss of trophoblast-dNK interactions led to the inhibition of dNK cell function, although their activating receptor expression was increased. We speculate that during pregnancy, two mechanisms operate to modulate the dNK cell activation:suppression of activating receptor levels in the first trimester by trophoblasts and disengagement of receptor-ligand coupling in the second trimester.


Subject(s)
Killer Cells, Natural/immunology , Lectins, C-Type/metabolism , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Pregnancy Trimester, First/immunology , Pregnancy Trimester, Second/immunology , Receptors, Natural Killer Cell/metabolism , Cell Communication , Decidua/cytology , Female , Humans , Interleukin-8/metabolism , Lymphocyte Count , Neovascularization, Physiologic , Phenotype , Pregnancy , Trophoblasts/metabolism , Up-Regulation , Vascular Endothelial Growth Factor A/metabolism
15.
Reproduction ; 152(5): 457-65, 2016 11.
Article in English | MEDLINE | ID: mdl-27539603

ABSTRACT

Aberrant placental angiogenesis is associated with fetal growth restriction (FGR). In mice, targeted disruption of the homeobox gene, transforming growth ß-induced factor (Tgif-1), which is also a transcription factor, causes defective placental vascularisation. Nevertheless, the role of TGIF-1 in human placental angiogenesis is unclear. We have previously reported increased TGIF-1 expression in human FGR placentae and demonstrated localisation of TGIF-1 protein in placental endothelial cells (ECs). However, its functional role remains to be investigated. In this study, we aimed to specifically compare TGIF-1 mRNA expression in placental ECs isolated from human FGR-affected pregnancies with gestation-matched control pregnancies in two independent cohorts from Australia and Canada and to identify the functional role of TGIF-1 in placental angiogenesis using the human umbilical vein endothelial cell-derived cell line, SGHEC-7, and primary human umbilical vein ECs. Real-time PCR revealed that TGIF-1 mRNA expression was significantly increased in ECs isolated from FGR-affected placentae compared with that of controls. The functional roles of TGIF-1 were determined in ECs after TGIF-1 siRNA transfection. TGIF-1 inactivation in ECs significantly reduced TGIF-1 at both the mRNA and protein levels, as well as the proliferative and invasive potential, but significantly increased the angiogenic potential. Using angiogenesis PCR screening arrays, we identified ITGAV, NRP-1, ANPGT-1 and ANPGT-2 as novel downstream targets of TGIF-1, after TGIF-1 inactivation in ECs. Collectively, these results show that TGIF-1 regulates EC function and the expression of angiogenic molecules; and when abnormally expressed, may contribute to the aberrant placental angiogenesis observed in FGR.


Subject(s)
Endothelial Cells/pathology , Fetal Growth Retardation/diagnosis , Fetal Growth Retardation/metabolism , Homeodomain Proteins/metabolism , Placenta/pathology , Repressor Proteins/metabolism , Trophoblasts/pathology , Adult , Cell Proliferation , Cells, Cultured , Endothelial Cells/metabolism , Female , Humans , Infant , Male , Placenta/metabolism , Pregnancy , Trophoblasts/metabolism
16.
Histol Histopathol ; 31(5): 557-71, 2016 May.
Article in English | MEDLINE | ID: mdl-26602431

ABSTRACT

Uterine spiral arteriole (SA) remodeling in early pregnancy involves a coordinated series of events including decidual immune cell recruitment, vascular cell disruption and loss, and colonization by placental-derived extravillous trophoblast (EVT). During this process, decidual SA are converted from narrow, muscular vessels into dilated channels lacking vasomotor control. We hypothesized that this extensive alteration in SA architecture must require significant reorganization and/or breakdown of the vascular extracellular matrix (ECM). First trimester decidua basalis (30 specimens) was immunostained to identify spiral arterioles undergoing trophoblast-independent and -dependent phases of remodeling. Serial sections were then immunostained for a panel of ECM markers, to examine changes in vascular ECM during the remodeling process. The initial stages of SA remodeling were characterized by loss of laminin, elastin, fibrillin, collagen types III, IV and VI from the basement membrane, vascular media and/or adventitia, and surrounding decidual stromal cells. Loss of ECM correlated with disruption and disorganization of vascular smooth muscle cells, and the majority of changes occurred prior to extensive colonization of the vessel wall by EVT. The final stages of SA remodeling, characterized by the arrival of EVT, were associated with the increased mural deposition of fibronectin and fibrinoid. This study provides the first detailed analysis of the spatial and temporal loss of ECM from the walls of remodeling decidual SA in early pregnancy.


Subject(s)
Arterioles/metabolism , Extracellular Matrix/metabolism , Placenta/blood supply , Pregnancy , Decidua/blood supply , Decidua/metabolism , Extracellular Matrix/chemistry , Female , Humans , Immunohistochemistry , Placenta/metabolism , Pregnancy Trimester, First
17.
Reprod Biol Endocrinol ; 12: 123, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-25475528

ABSTRACT

BACKGROUND: Adverse gestational outcomes such as preeclampsia (PE) and intrauterine growth restriction (IUGR) are associated with placental insufficiency. Normal placental development relies on the insulin-like growth factors -I and -II (IGF-I and -II), in part to stimulate trophoblast proliferation and extravillous trophoblast (EVT) migration. The insulin-like growth factor binding proteins (IGFBPs) modulate the bioavailability of IGFs in various ways, including sequestration, potentiation, and/or increase in half-life. The roles of IGFBP-4 and -5 in the placenta are unknown, despite consistent associations between pregnancy complications and the levels of two IGFBP-4 and/or -5 proteases, pregnancy-associated plasma protein -A and -A2 (PAPP-A and PAPP-A2). The primary objective of this study was to elucidate the effects of IGFBP-4 and -5 on IGF-I and IGF-II in a model of EVT migration. A related objective was to determine the timing and location of IGFBP-4 and -5 expression in the placental villi. METHODS: We used wound healing assays to examine the effects of IGFBP-4 and -5 on the migration of HTR-8/SVneo cells following 4 hours of serum starvation and 24 hours of treatment. Localization of IGFBP-4, -5 and PAPP-A2 was assessed by immunohistochemical staining of first trimester placental sections. RESULTS: 2 nM IGF-I and -II each increased HTR-8/SVneo cell migration with IGF-I increasing migration significantly more than IGF-II. IGFBP-4 and -5 showed different levels of inhibition against IGF-I. 20 nM IGFBP-4 completely blocked the effects of 2 nM IGF-I, while 20 nM IGFBP-5 significantly reduced the effects of 2 nM IGF-I, but not to control levels. Either 20 nM IGFBP-4 or 20 nM IGFBP-5 completely blocked the effects of 2 nM IGF-II. Immunohistochemistry revealed co-localization of IGFBP-4, IGFBP-5 and PAPP-A2 in the syncytiotrophoblast layer of first trimester placental villi as early as 5 weeks of gestational age. CONCLUSIONS: IGFBP-4 and -5 show different levels of inhibition on the migration-stimulating effects of IGF-I and IGF-II, suggesting different roles for PAPP-A and PAPP-A2. Moreover, co-localization of the pappalysins and their substrates within placental villi suggests undescribed roles of these molecules in early placental development.


Subject(s)
Cell Movement/physiology , Insulin-Like Growth Factor Binding Protein 4/biosynthesis , Insulin-Like Growth Factor Binding Protein 5/biosynthesis , Placenta/metabolism , Trophoblasts/metabolism , Cell Line , Cell Movement/drug effects , Dose-Response Relationship, Drug , Female , Gestational Age , Humans , Immunohistochemistry , Insulin-Like Growth Factor Binding Protein 4/pharmacology , Insulin-Like Growth Factor Binding Protein 5/pharmacology , Insulin-Like Growth Factor I/pharmacology , Insulin-Like Growth Factor II/pharmacology , Pregnancy , Pregnancy Trimester, First , Pregnancy-Associated Plasma Protein-A/biosynthesis , Trophoblasts/cytology , Trophoblasts/drug effects
18.
J Immunol ; 193(6): 3070-9, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25135830

ABSTRACT

The maternal leukocytes of the first-trimester decidua play a fundamental role in implantation and early development of the fetus and placenta, yet little is known regarding the second-trimester decidual environment. Our multicolor flow cytometric analyses of human decidual leukocytes detected an elevation in tissue resident neutrophils in the second trimester. These cells in both human and murine samples were spatially restricted to decidua basalis. In comparison with peripheral blood neutrophils (PMNs), the decidual neutrophils expressed high levels of neutrophil activation markers and the angiogenesis-related proteins: vascular endothelial growth factor-A, Arginase-1, and CCL2, similarly shown in tumor-associated neutrophils. Functional in vitro assays showed that second-trimester human decidua conditioned medium stimulated transendothelial PMN invasion, upregulated VEGFA, ARG1, CCL2, and ICAM1 mRNA levels, and increased PMN-driven in vitro angiogenesis in a CXCL8-dependent manner. This study identified a novel neutrophil population with a physiological, angiogenic role in human decidua.


Subject(s)
Decidua/cytology , Interleukin-8/immunology , Neovascularization, Physiologic/immunology , Neutrophils/cytology , Pregnancy Trimester, Second/immunology , Animals , Antibodies/immunology , Arginase/biosynthesis , Arginase/genetics , B-Lymphocytes/immunology , Cells, Cultured , Chemokine CCL2/biosynthesis , Chemokine CCL2/genetics , Culture Media, Conditioned/pharmacology , DNA-Binding Proteins/genetics , Decidua/immunology , Female , Granulocytes/cytology , Granulocytes/immunology , Humans , Intercellular Adhesion Molecule-1/genetics , Interleukin Receptor Common gamma Subunit/genetics , Interleukin-8/metabolism , Killer Cells, Natural/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Neutrophils/immunology , Pregnancy , RNA, Messenger/biosynthesis , Receptors, Chemokine/biosynthesis , T-Lymphocytes/immunology , Transendothelial and Transepithelial Migration , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics
19.
PLoS One ; 9(1): e87621, 2014.
Article in English | MEDLINE | ID: mdl-24498154

ABSTRACT

BACKGROUND: Preeclampsia (PE) is characterized by exaggerated apoptosis of the villous trophoblast of placental villi. Since p53 is a critical regulator of apoptosis we hypothesized that excessive apoptosis in PE is mediated by abnormal expression of proteins participating in the p53 pathway and that modulation of the p53 pathway alters trophoblast apoptosis in vitro. METHODS: Fresh placental villous tissue was collected from normal pregnancies and pregnancies complicated by PE; Western blotting and real-time PCR were performed on tissue lysate for protein and mRNA expression of p53 and downstream effector proteins, p21, Bax and caspases 3 and 8. To further assess the ability of p53 to modulate apoptosis within trophoblast, BeWo cells and placental villous tissue were exposed to the p53-activator, Nutlin-3, alone or in combination with the p53-inhibitor, Pifithrin-α (PFT-α). Equally, Mdm2 was knocked-down with siRNA. RESULTS: Protein expression of p53, p21 and Bax was significantly increased in pregnancies complicated by PE. Conversely, Mdm2 protein levels were significantly depleted in PE; immunohistochemistry showed these changes to be confined to trophoblast. Reduction in the negative feedback of p53 by Mdm2, using siRNA and Nutlin-3, caused an imbalance between p53 and Mdm2 that triggered apoptosis in term villous explants. In the case of Nutlin, this was attenuated by Pifithrin-α. CONCLUSIONS: These data illustrate the potential for an imbalance in p53 and Mdm2 expression to promote excessive apoptosis in villous trophoblast. The upstream regulation of p53 and Mdm2, with regard to exaggerated apoptosis and autophagy in PE, merits further investigation.


Subject(s)
Pre-Eclampsia/genetics , Pre-Eclampsia/metabolism , Trophoblasts/metabolism , Trophoblasts/pathology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Caspase 3/genetics , Caspase 3/metabolism , Caspase 8/genetics , Caspase 8/metabolism , Chorionic Villi/metabolism , Chorionic Villi/pathology , Female , Humans , Placenta/metabolism , Placenta/pathology , Pregnancy , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
20.
Hum Reprod ; 28(11): 3026-37, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24001716

ABSTRACT

STUDY QUESTION: Is sphingosine-1-phosphate (S1P) signalling involved in the regulation of the angiogenic function of decidual (d)NK cells during human pregnancy? SUMMARY ANSWER: Human dNK cells, characterized by S1P receptor 5 (S1PR5) expression, are reactive to microenvironmental S1P to modify their VEGF expression and to regulate trophoblast migration and endothelial angiogenesis. WHAT IS KNOWN ALREADY: S1P signalling can modulate peripheral (p)NK cells migration and function. As a unique NK population, human dNK can produce multiple cytokines and angiogenic growth factors to mediate extravillous trophoblast (EVT) invasion and spiral artery remodelling during pregnancy. STUDY DESIGN, SIZE, DURATION: The study was designed to examine S1PR expression and function by freshly isolated human dNK cells in response to different S1P scenarios, created by FTY720, an S1P analogue and S1PR modulator. Ex vivo and in vitro experiments were performed to evaluate the functions of dNK cells. The study was performed between September 2011 and June 2013. PARTICIPANTS/MATERIALS, SETTING, METHODS: Human peripheral blood and decidual samples were collected and the S1PR expression by the decidual leukocytes population was examined. FTY720-induced dNK phenotypic and functional changes (including VEGF and IL-8 expression) were evaluated by multi-colour flow cytometric assays and transwell migration studies. Human placental explant culture and wound healing assays were performed to investigate whether S1P-activated dNK mediated trophoblast migration while angiogenesis was assessed by human umbilical vein endothelial cells (HUVEC) tube formation assays. Both first and second trimester dNK cells were studied to compare the difference in S1PR expression over time at the fetal-maternal interface. MAIN RESULTS AND THE ROLE OF CHANCE: Freshly isolated NK cells (CD45(+)CD56(+)CD16(-)) from blood (pNK) and decidua (dNK) had low S1PR1 reactivity while S1PR5 was prominently expressed by dNK (40%) and, to a lesser extent, by pNK (18%; P < 0.05) cells. S1PR5 expression by dNK was significantly down-regulated by FTY720 treatment, which also impaired decidual leukocyte mobility and cellular contact with invasive EVT. FTY720 significantly reduced VEGF expression by dNK, both in the numbers of VEGF(+) cells and in fluorescence intensity (P < 0.05). IL-8 expression by dNK was not changed by FTY720 and remained low at 8% positivity. Trophoblast migration and HUVEC tube formation were stimulated by control leukocytes, enriched CD56(+) dNK or their conditioned medium, respectively, but this effect was markedly abrogated once they were pretreated with FTY720 (P < 0.05). There was a significant decrease in S1PR5 expression in second trimester dNK cells, compared with those from first trimester (P < 0.05). No significant differences in the levels of angiogenic factors (VEGF or IL-8) were detected between first and second trimester dNK cells. LIMITATIONS, REASONS FOR CAUTION: Our ex vivo and in vitro experimental samples were from healthy women undergoing elective pregnancy termination. FTY720 is a chemical ligand for the S1PRs; little is known regarding the levels or actions of the naturally occurring ligand S1P in human gestational tissues. The in vivo function of S1PR5(+) dNK may be further investigated by using a genetically modified animal model. WIDER IMPLICATIONS OF THE FINDINGS: This is the first study to investigate the role of S1PR and S1P interaction on dNK cell physiology and their downstream effects on trophoblast migration. We suggest that S1PR5 may represent a potential target for cellular targeted treatments for gestational diseases such as pre-eclampsia and intrauterine growth restriction that are characterized by inadequate dNK/trophoblast-coordinated uterine spiral artery transformation. STUDY FUNDING/COMPETING INTEREST(S): This study was supported by Canadian Institutes of Health Research (CIHR), MOP82811 to Dr S.J.L.


Subject(s)
Cell Movement , Killer Cells, Natural/physiology , Lysophospholipids/physiology , Neovascularization, Physiologic , Sphingosine/analogs & derivatives , Trophoblasts/physiology , Cells, Cultured , Decidua/cytology , Decidua/metabolism , Down-Regulation/drug effects , Female , Fingolimod Hydrochloride , Flow Cytometry , Human Umbilical Vein Endothelial Cells , Humans , Immunohistochemistry , Interleukin-8/metabolism , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Lysophospholipids/metabolism , Propylene Glycols/pharmacology , Receptors, Lysosphingolipid/metabolism , Signal Transduction , Sphingosine/metabolism , Sphingosine/pharmacology , Sphingosine/physiology , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...