Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Placenta ; 35(12): 1027-34, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25449030

ABSTRACT

INTRODUCTION: Decidual leukocytes are critical to the development of the fetomaternal interface, regulating tolerance to the semi-allogeneic fetus and vascular transformation of the uterine spiral arteries. Despite the continuation of these processes beyond the first trimester of pregnancy, the second trimester has largely been unstudied, with investigation focusing on early gestation and term tissues. We sought to characterize changes in decidual leukocyte populations from first to second trimester. METHODS: Multicolor flow cytometry was performed on isolated decidual leukocytes from elective terminations of pregnancy between 6 and 20 weeks of gestation for study of first (6-12 weeks) and second trimesters (13-20 weeks). Specific subpopulations were identified by comparison to isotype and fluorescent-minus-one (FMO) controls. RESULTS: Decidual natural killer cells (CD56(+)CD16(-)CD3(-)) did not change in number, although a population of dNK with decreased CD56 brightness was observed in second trimester decidua. CD14(+)HLA-DR(+) macrophage numbers declined from first to second trimester (p = 0.031), yet a CD163(+)CD206(+) subset designating alternatively activated M2-like macrophages increased during the same period (p = 0.015). Intermediate CD205(+) dendritic cells demonstrated significant decline (p = 0.022), but immature CD209(+) and mature CD83(+) dendritic cells did not differ between trimesters. Total CD3(+) and CD3(+)CD4(+) T lymphocytes increased (p = 0.0079, p = 0.0028); CD3(+)CD8(+) T cells trended towards increase but did not differ significantly. CONCLUSION: Several changes in leukocyte subsets are observed in the second trimester that promote a tolerogenic and angiogenic decidual microenvironment through mid-gestation.


Subject(s)
Decidua/cytology , Leukocytes/cytology , Pregnancy Trimester, First , Pregnancy Trimester, Second , Adult , Antigens, CD , Decidua/immunology , Female , Flow Cytometry , Humans , Leukocyte Count , Leukocytes/immunology , Pregnancy , Young Adult
2.
Placenta ; 35 Suppl: S15-9, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24321780

ABSTRACT

Workshops are an important part of the IFPA annual meeting as they allow for discussion of specialised topics. At IFPA meeting 2013 there were twelve themed workshops, three of which are summarized in this report. These workshops related to various aspects of placental biology but collectively covered areas of placental function, cell turnover and immunology: 1) immunology; 2) novel determinants of placental cell fate; 3) dual perfusion of human placental tissue.


Subject(s)
Placenta/immunology , Placentation , Pregnancy/immunology , Animals , Female , Humans , Perfusion/methods
3.
Placenta ; 34(9): 817-23, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23790363

ABSTRACT

INTRODUCTION: The multidrug resistance proteins, P-glycoprotein (P-gp, encoded by the ABCB1 gene) and breast cancer resistance protein (BCRP, encoded by ABCG2) are highly expressed in the first trimester placenta. These transporters protect the fetus from exposure to maternally derived toxins and xenobiotics. Since oxygen is a regulator of multidrug resistance in various tissues, we hypothesized that changes in oxygen tension alter placental ABCB1/P-gp and ABCG2/BCRP expression in the first trimester. METHODS: Placental specimens were collected from first (n = 7), second (n = 5) and term pregnancies (n = 5). First trimester placental villous explants were incubated (24 or 48 h) in different oxygen tension (3-20%). ABCB1, ABCG2 and VEGFA mRNA expression levels were assessed by RT-PCR and protein was localized by IHC. RESULTS: ABCB1 is expressed most highly in the first trimester placenta (p < 0.05), whereas ABCG2 expression does not change significantly over pregnancy. P-gp and BCRP staining is present in the syncytiotrophoblast and in cytotrophoblasts. ABCG2 mRNA is increased in hyperoxic (20%) conditions after 48 h (p < 0.05). In contrast, hypoxia (3%) did not change ABCB1 mRNA expression but significantly increased VEGFA mRNA (p < 0.05). Hypoxia resulted in increased BCRP staining in cytotrophoblasts and in the microvillous membrane of the syncytium. Whereas, hypoxia resulted in increased P-gp staining in proliferating cytotrophoblasts. CONCLUSION: We conclude that placental multidrug resistance expression, specifically ABCG2, is regulated by oxygen tension in the first trimester. It is possible that changes in placental oxygen supply are capable of altering fetal drug exposure especially during early pregnancy.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP-Binding Cassette Transporters/metabolism , Drug Resistance, Multiple , Gene Expression Regulation, Developmental , Neoplasm Proteins/metabolism , Oxygen/metabolism , Placenta/metabolism , Vascular Endothelial Growth Factor A/metabolism , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/biosynthesis , ATP-Binding Cassette Transporters/genetics , Cell Hypoxia , Female , Giant Cells/cytology , Giant Cells/drug effects , Giant Cells/metabolism , Humans , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Placenta/cytology , Placenta/drug effects , Placentation , Pregnancy , Pregnancy Trimester, First , Pregnancy Trimester, Second , Pregnancy Trimester, Third , RNA, Messenger/metabolism , Tissue Banks , Tissue Culture Techniques , Trophoblasts/cytology , Trophoblasts/drug effects , Trophoblasts/metabolism , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics
4.
Placenta ; 33(4): 285-93, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22264586

ABSTRACT

The placental microvasculature is essential for efficient transfer of gases, nutrients and waste between the mother and fetus. Microvascular hypoplasia of the terminal villi is a common pathology in severe Intra Uterine Growth Restriction (IUGR). We used novel methods to obtain placental micro-vascular endothelial cells (PlMEC) from preterm control placentas (n = 3) and placentas from pregnancies with severe IUGR (n = 6) with absent or reversed end-diastolic velocity in the umbilical artery. Distal placental villous tissue was collected to enrich for intermediate and terminal villi. Tissue was digested and PlMEC positively selected using tocosylated magnetic Dynabeads labeled with Human Endothelial Antigen lectin. The purity of the PlMEC (94 ± 2 SD %) was assessed by CD31 and vimentin immunocytochemistry. RNA was extracted from the PlMEC samples and subjected to Affymetrix microarray analysis (U133Plus2 array chips). Comparison of preterm and IUGR PlMEC gene expression profiles identified BTNL9 and NTRK2 transcripts to be upregulated and SAA1 and SLAMF1 transcripts to be downregulated in all 6 IUGR cases relative to preterm controls. A third downregulated gene GNAS was identified to be near significance. Changes were demonstrated to be significant at the mRNA level by Real Time PCR in the PlMEC samples. Changes in the IUGR endothelium were confirmed at the protein level by immunohistochemistry for the 3 with available antibodies. We used a tissue microarray constructed from an independent cohort of placental samples from severe IUGR (n = 7), preeclamptic (n = 7), preterm control (n = 6) and term control (n = 6) pregnancies. Results confirmed differential endothelial expression of BTNL9, NTRK2 and SLAMF1 in IUGR versus preterm and term samples. These studies are the first to characterize PlMEC gene expression profiles thus we have advanced our understanding of the molecular basis of placental micro-vascular pathophysiology in fetal growth restriction.


Subject(s)
Endothelium, Vascular/metabolism , Fetal Growth Retardation/metabolism , Gene Expression Regulation, Developmental , Microvessels/metabolism , Placenta/blood supply , Adult , Antigens, CD/genetics , Antigens, CD/metabolism , Butyrophilins , Cohort Studies , Endothelium, Vascular/pathology , Female , Fetal Growth Retardation/pathology , Gene Expression Profiling , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Microvessels/pathology , Oligonucleotide Array Sequence Analysis , Placenta/pathology , Pre-Eclampsia/metabolism , Pre-Eclampsia/pathology , Pregnancy , Premature Birth/metabolism , Premature Birth/pathology , RNA, Messenger/metabolism , Receptor, trkB/genetics , Receptor, trkB/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Serum Amyloid A Protein/genetics , Serum Amyloid A Protein/metabolism , Severity of Illness Index , Signaling Lymphocytic Activation Molecule Family Member 1 , Young Adult
5.
Placenta ; 33(3): 143-50, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22230251

ABSTRACT

The placenta is a highly vascularized organ thus angiogenesis is a key process in placental development. The contribution that different cells in the villous stroma play in placental angiogenesis is largely unknown. In this study we identified a novel stromal cell type in sections of term placenta which is morphologically fibroblastic and expressing the fibroblast marker TE-7 but also positive for the monocytic markers CD115 and CD14 and designated these cells as fibrocyte-like cells. Populations of fibrocyte-like cells from the placenta were isolated by two methods: culture of adherence-selected placental cells and, for higher purity, by CD45 fluorescence activated cell sorting (FACS). Fibrocyte-like cell conditioned medium increased endothelial tubule-like structure formation 2-fold versus control medium. Both pro-angiogenic growth factors vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (b-FGF) and the anti-angiogenic factor soluble-Flt were found in the conditioned medium. Neutralizing antibodies against VEGF and b-FGF reduced endothelial cell tubule-like structures to control levels. These data suggests that fibrocyte-like cells, a previously unidentified cell of the villous stroma, may play an important role in the regulation of placental angiogenesis.


Subject(s)
Fibroblasts/cytology , Fibroblasts/physiology , Placenta/cytology , Cell Separation/methods , Cells, Cultured , Chorionic Villi/metabolism , Chorionic Villi/physiology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Female , Flow Cytometry , Fluorescent Antibody Technique, Indirect , Humans , Macrophages/metabolism , Macrophages/physiology , Monocytes/metabolism , Monocytes/physiology , Neovascularization, Physiologic/physiology , Placenta/blood supply , Placenta/physiology , Pregnancy , Primary Cell Culture
6.
Placenta ; 32 Suppl 2: S81-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21227506

ABSTRACT

Workshops are an important part of the IFPA annual meeting. At IFPA Meeting 2010 there were twelve themed workshops, six of which are summarized in this report. 1. The immunology workshop focused on normal and pathological functions of the maternal immune system in pregnancy. 2. The transport workshop dealt with regulation of ion and water transport across the syncytiotrophoblast of human placenta. 3. The epigenetics workshop covered DNA methylation and its potential role in regulating gene expression in placental development and disease. 4. The vascular reactivity workshop concentrated on methodological approaches used to study placental vascular function. 5. The workshop on epitheliochorial placentation covered current advances from in vivo and in vitro studies of different domestic species. 6. The proteomics workshop focused on a variety of techniques and procedures necessary for proteomic analysis and how they may be implemented for placental research.


Subject(s)
Fetus/physiology , Placenta/physiology , Trophoblasts/physiology , Animals , Education , Epigenesis, Genetic/physiology , Female , Fetus/blood supply , Fetus/cytology , Fetus/immunology , Humans , Ion Transport/physiology , Maternal-Fetal Exchange/physiology , Placenta/blood supply , Placenta/cytology , Placenta/immunology , Placentation/physiology , Pregnancy , Proteomics/methods , Trophoblasts/cytology , Trophoblasts/immunology
7.
Biol Reprod ; 83(6): 1036-45, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20739666

ABSTRACT

This study examines the role of HER1 signaling in the differentiation of proliferative extravillous trophoblast (EVT) into invasive EVT. Using the JAR choriocarcinoma cell line and placental villous explants as experimental models and immunohistochemical assessment of protein markers of EVT differentiation (downregulation of HER1 and Cx40 and upregulation of HER2 and alpha1 integrin), we show that the ability of decidual conditioned medium (DCM) to induce HER1/2 switching was abrogated in the presence of the HER1 antagonist, AG1478. Similarly, epidermal growth factor (EGF) treatment resulted in the downregulation of HER1 and an upregulation of HER2 expression, whereas co-incubation of EGF with AG1478 inhibited this response. However, EGF did not downregulate Cx40 or induce migration of EVT. In contrast, heparin-binding epidermal-like growth factor (HBEGF) stimulated dose-dependent JAR cell migration, which was inhibited by both AG1478 and AG825 (HER2 antagonist). Western blot analysis of HER1 activation demonstrated that HBEGF-mediated phosphorylation of the HER1 Tyr992 and Tyr1068 sites, while EGF activated the Tyr1045 site. Moreover, HBEGF induced a stronger and more sustained activation of both the mitogen-activated protein kinase and phosphoinositol 3 kinase (PIK3) signaling pathways. Migration assays using a panel of signaling pathway inhibitors demonstrated that the HBEGF-mediated migration was dependent on the PIK3 pathway. These results demonstrate that HBEGF-mediated HER1 signaling through PIK3 is an important component of EVT invasion.


Subject(s)
Cell Differentiation , ErbB Receptors/metabolism , Signal Transduction , Trophoblasts/metabolism , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Line , Cell Movement/drug effects , Culture Media, Conditioned , Decidua/metabolism , Enzyme Inhibitors/pharmacology , Epidermal Growth Factor/metabolism , ErbB Receptors/antagonists & inhibitors , Female , Heparin-binding EGF-like Growth Factor , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Ligands , Phosphatidylinositol 3-Kinase/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Placentation , Pregnancy , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Signal Transduction/drug effects , Tissue Culture Techniques , Trophoblasts/cytology , Trophoblasts/drug effects
8.
Placenta ; 31(10): 910-8, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20709391

ABSTRACT

OBJECTIVES: The Liver X receptors (LXR) alpha and beta and their target genes such as the ATP-binding cassette (ABC) transporters have been shown to be crucially involved in the regulation of cellular cholesterol homeostasis. The aim of this study was to characterize the role of LXR alpha/beta in the human placenta under normal physiological circumstances and in preeclampsia. STUDY DESIGN: We investigated the expression pattern of the LXRs and their target genes in the human placenta during normal pregnancy and in preeclampsia. Placental explants and cell lines were studied under different oxygen levels and pharmacological LXR agonists. MAIN OUTCOME MEASURES: Gene expressions (Taqman PCR) and protein levels (Western Blot) were combined with immunohistochemistry to analyze the expression of LXR and its target genes. RESULTS: In the human placenta, LXRA and LXRB expression increased during normal pregnancy. This was paralleled by the expression of their prototypical target genes, e.g., the cholesterol transporter ABCA1. Interestingly, early-onset preeclamptic placentae revealed a significant upregulation of ABCA1. Culture of JAr trophoblast cells and human first trimester placental explants under low oxygen lead to increased expression of LXRA and ABCA1 which was further enhanced by the LXR agonist T0901317. CONCLUSIONS: LXRA together with ABCA1 are specifically expressed in the human placenta and can be regulated by hypoxia. Deregulation of this system in early preeclampsia might be the result of placental hypoxia and hence might have consequences for maternal-fetal cholesterol transport.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Hypoxia/metabolism , Orphan Nuclear Receptors/metabolism , Oxygen/metabolism , Placenta/metabolism , Pre-Eclampsia/metabolism , Trophoblasts/metabolism , ATP-Binding Cassette Transporters/genetics , Anticholesteremic Agents/pharmacology , Cell Line, Tumor , Cholesterol/metabolism , Female , Gene Expression Regulation, Developmental , Humans , Hydrocarbons, Fluorinated/pharmacology , Immunoblotting , Immunohistochemistry , In Vitro Techniques , Liver X Receptors , Orphan Nuclear Receptors/agonists , Orphan Nuclear Receptors/genetics , Oxygen/administration & dosage , Placenta/cytology , Pre-Eclampsia/pathology , Pregnancy , RNA/chemistry , RNA/genetics , Reverse Transcriptase Polymerase Chain Reaction , Statistics, Nonparametric , Sulfonamides/pharmacology , Trophoblasts/cytology
9.
Cell Death Differ ; 16(5): 719-27, 2009 May.
Article in English | MEDLINE | ID: mdl-19219068

ABSTRACT

Mammalian placentation is a highly regulated process and is dependent on the proper development of specific trophoblast cell lineages. The two major types of trophoblast, villous and extravillous, show mitotic arrest during differentiation. In mice, the transcription factor, glial cell missing-1 (Gcm1), blocks mitosis and is required for syncytiotrophoblast formation and morphogenesis of the labyrinth, the murine equivalent of the villous placenta. The human homolog GCM1 has an analogous expression pattern, but its function is presently unknown. We studied GCM1 function in the human-derived BeWo choriocarcinoma cell line and in first trimester human placental villous and extravillous explants. The GCM1 expression was either inhibited by siRNA and antisense oligonucleotides methods or upregulated by forskolin treatment. Inhibition of GCM1 resulted in an increased rate of proliferation, but prevented de novo syncytiotrophoblast formation in syncytially denuded floating villous explants. GCM1 inhibition prevented extravillous differentiation along the invasive pathway in extravillous explants on matrigel. By contrast, forskolin-induced expression of GCM1 reduced the rate of proliferation and increased the rate of syncytialization in the floating villous explant model. Our studies show that GCM1 has a distinct role in the maintenance, development and turnover of the human trophoblast. Alterations in GCM1 expression or regulation may explain several aspects of two divergent severe placental insufficiency syndromes, namely preeclampsia and intrauterine growth restriction, which cause extreme preterm birth.


Subject(s)
Cell Differentiation , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Trophoblasts/metabolism , Cell Fusion , Cell Line, Tumor , Colforsin/pharmacology , DNA-Binding Proteins , Female , Humans , Nuclear Proteins/antagonists & inhibitors , Pregnancy , Pregnancy Trimester, First , RNA, Small Interfering/metabolism , RNA, Small Interfering/pharmacology , Transcription Factors/antagonists & inhibitors , beta Catenin/metabolism
10.
Placenta ; 30(1): 96-104, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19007985

ABSTRACT

BACKGROUND: In mice the exchange of oxygen and nutrients between mother and fetus occurs in the chorioallantoic placenta where fetal capillaries come in close proximity with maternal blood perfusing trophoblast-lined sinusoids. Despite its critical importance, quantitative in vivo gene expression over the initial stages of chorioallantoic placental development has not been described, nor are there in vitro systems recapitulating the critical syncytiotrophoblast differentiation step in its formation. Here we describe molecular events that occur during the onset of chorioallantoic morphogenesis in mice in vivo, and in placental explant and whole conceptus cultures in vitro. RESULTS: Chorioallantoic morphogenesis began immediately following allantoic fusion with the chorion in vivo, and was associated with significant upregulation of syncytiotrophoblast associated mRNA (Gcm1 and Syncytin A). However mouse placentas with chorioallantoic point attachment cultured with the allantois or as whole conceptuses did not upregulate Gcm1 and/or Syncytin A, suggesting that syncytiotrophoblast differentiation did not occur in vitro. Failure of morphogenesis appeared to be due to failure to sustain in vitro the chorionic trophoblast cells from which the syncytiotrophoblast cells are derived. In vitro culture conditions did support the upregulation of ectoplacental cone marker Tpbpalpha, maintenance of giant cell marker Pl1, and maintenance of Fgfr2 expression; all of which mimicked in vivo events observed over this developmental interval. CONCLUSIONS: We conclude that chorionic trophoblast maintenance and the early events that occur in vivo between chorioallantoic point attachment and primary villous formation are dependent on undefined intrauterine factors that were not present in the in vitro culture system. Nevertheless, in vitro culture conditions were appropriate to reproduce in vivo expression levels of Fgfr2, Pl1, and Tpbpalpha in placental explants.


Subject(s)
Allantois/metabolism , Chorion/metabolism , Embryo, Mammalian/embryology , Gene Expression Regulation, Developmental , Transcription, Genetic , Allantois/embryology , Animals , Biomarkers/metabolism , Chorion/embryology , Culture Techniques , DNA-Binding Proteins , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Mice, Inbred ICR , Neuropeptides/genetics , Neuropeptides/metabolism , Pregnancy , Pregnancy Proteins/genetics , Pregnancy Proteins/metabolism , RNA, Messenger/metabolism , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Transcription Factors , Trophoblasts/metabolism , Trophoblasts/physiology
11.
Placenta ; 29 Suppl A: S17-9, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18155293

ABSTRACT

Stem cells that can be derived from fetal membranes represent an exciting field of research that bears tremendous potential for developmental biology and regenerative medicine. In this report we summarize contributions to a workshop in which newest insights into the characteristics, subtypes and molecular determinants of stem cells from trophoblast and endometrial tissues were presented.


Subject(s)
Extraembryonic Membranes/cytology , Extraembryonic Membranes/physiology , Stem Cells/cytology , Stem Cells/physiology , Animals , Education , Humans
12.
Placenta ; 27 Suppl A: S141-3, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16542721

ABSTRACT

Challenge lies ahead in unravelling the role played by trophoblast and its repertoire of expressed genes in normal human placental development, growth and pathology. Specific technical advances will clearly be required for characterisation of function. In particular, improvements in our repertoire of in vitro models are needed before many of the key questions can be answered. Recent advances in the study of human trophoblast differentiation are discussed.


Subject(s)
Cell Communication , Cell Differentiation , Gene Expression Regulation, Developmental , Stem Cells/physiology , Trophoblasts/physiology , Cell Fusion , Cell Lineage , Cell Movement , Humans
13.
Placenta ; 27 Suppl A: S114-21, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16545451

ABSTRACT

In this study we show that decidua conditioned media (DCM) downregulate Connexin 40 (C x 40) expression in extravillous trophoblast (EVT) outgrowths and can promote EVT differentiation to the invasive phenotype resulting in switching of integrin and EGF receptor expression. This suggests that growth factors secreted by the decidua, such as EGF, mediate trophoblast migration/invasion and may do so by modulating C x 40 expression and function. To test this hypothesis we have utilized migration assays using cell lines expressing C x 40. Migration assays were performed with Jeg-3, Jeg-3 overexpressing C x 40 (JpUHD) and JAR cells seeded on fibronectin-coated inserts with 8 microm pores and incubated in the absence or presence of serum-starved decidual cells. Cell migration was only observed in the presence of DCM. Conversely overexpression of C x 40 in Jeg-3 cells resulted in inhibition of cell migration as compared to wild-type control. Addition of DCM to cultured JAR cells resulted in the downregulation of C x 40 protein. EGF is known to stimulate trophoblast migration/invasion and was detected in DCM; therefore, we investigated the action of EGF on C x 40. EGF (10 ng/mL) resulted in the downregulation of C x 40 in the JAR cell line. However, EGF had no effect on JAR cell migration. We conclude that decidual secretion of growth factors, such as EGF, may act to prime trophoblast for migration/invasion through modulation of connexin expression and function.


Subject(s)
Cell Movement/drug effects , Chorionic Villi/metabolism , Connexins/metabolism , Epidermal Growth Factor/physiology , Trophoblasts/metabolism , Trophoblasts/physiology , Biomarkers, Tumor/analysis , Cell Differentiation , Cell Line, Tumor , Cell Membrane/metabolism , Culture Media, Conditioned/pharmacology , Decidua/metabolism , Female , Humans , Pregnancy , Pregnancy Trimester, First , Gap Junction alpha-5 Protein
14.
Placenta ; 27(4-5): 367-74, 2006.
Article in English | MEDLINE | ID: mdl-15950280

ABSTRACT

Murine trophoblast stem (TS) cells express fibroblast growth factor receptor 2 (FGFR2) and are maintained in their proliferative state by fibroblast growth factor 4 (FGF4). We show in this report that in the first trimester human placenta FGFR2 expression is similarly found in a subset of villous cytotrophoblast and in proximal anchoring columns. Western analysis demonstrated declining FGFR2 protein expression as gestation advanced, suggesting a similar role for FGF in early human trophoblast proliferation. Mouse TS cell differentiation is known to occur along two distinct transcriptionally-regulated pathways; extravillous trophoblast (EVT) cells invade the uterine wall to promote maternal blood flow whilst syncytiotrophoblast lines chorionic villi in the labyrinth. Similar differentiation steps occur in the human placenta though the fate of human trophoblast stem cells is presently unknown. To investigate the mechanisms underlying human cytotrophoblast differentiation we have developed a novel cultured floating first trimester villous explant model in which denuded first trimester villi spontaneously regenerate syncytiotrophoblast following 48 h of culture. Addition of FGF4 and heparin inhibited syncytiotrophoblast regeneration in favor of forming clumps of cytotrophoblast. Proximal cells in these clumps were FGFR2 immuno-reactive and proliferative, intermediate parts expressed alpha5beta1-integrin, while the distal portion expressed HLA-G and the invasive integrin alpha1beta1 indicating differentiation to the EVT phenotype. In contrast, non-denuded villi exposed to FGF4 exhibited similar proliferation of the cytotrophoblast; however, these cells did not express any of the invasive EVT markers. We conclude that FGFR2-positive chorionic cytotrophoblasts exhibit bi-potential behaviour, being capable of forming either syncytiotrophoblast or EVT. We suggest bipotential trophoblast progenitor cells persist during first trimester human placental development.


Subject(s)
Cell Differentiation , Chorionic Villi/physiology , Fibroblast Growth Factor 4/physiology , Pregnancy Trimester, First/physiology , Trophoblasts/cytology , Animals , Female , Giant Cells/cytology , Humans , In Vitro Techniques , Mice , Pregnancy , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Trophoblasts/metabolism
15.
Placenta ; 25(7): 595-607, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15193866

ABSTRACT

Little is known about the role of gap junctional intercellular communication (GJIC) in human trophoblast differentiation, particularly during the formation of extravillous trophoblast (EVT) cell columns and their subsequent differentiation into invasive cells. We have identified transcripts for five connexin gap junction proteins in the early human placenta (Cx32, Cx37, Cx40, Cx43 and Cx45). Of these, Cx40 and Cx45 proteins immunolocalize to EVT in anchoring cell columns. Cx40 expression is prominent in the anchoring column throughout the first trimester of pregnancy (6-14 weeks gestation). We used first trimester placental villous explant cultures to determine the functional significance of the inhibition of GJIC in EVT cell proliferation and differentiation using two known GJIC inhibitors, carbenoxolone (CBX) and heptanol. The morphology of EVT outgrowths changed dramatically upon GJIC-blockade, from compact and organized outgrowths into a scattered group of rounded individual trophoblast cells, reminiscent of an early invasive phenotype. Furthermore, the inhibition of GJIC in placental explants by CBX or heptanol induced a switch away from the proliferative and towards an invasive EVT phenotype, as evident from (a) the loss of the proliferation marker Ki67 and (b) an increase in the invasive marker alpha1 integrin. We also utilized antisense oligonucleotides to inhibit Cx40 protein expression in placental explants. Cx40 antisense treatment also resulted in the abolishment of outgrowth EVT cell proliferation (as determined by Ki67 immunostaining). Together, these results suggest that gap junctions composed particularly of Cx40 channels are required for the proliferation of EVT cells in anchoring cell columns, and that a loss of GJIC contributes to differentiation to the invasive EVT phenotype.


Subject(s)
Cell Division , Gap Junctions/physiology , Placentation , Trophoblasts/ultrastructure , Carbenoxolone/pharmacology , Cell Communication/drug effects , Cell Differentiation , Cells, Cultured , Connexin 43/analysis , Connexin 43/genetics , Connexins/analysis , Connexins/genetics , Female , Fluorescent Antibody Technique , Gap Junctions/drug effects , Humans , Immunohistochemistry , In Situ Hybridization , Pregnancy , Pregnancy Trimester, First , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Tissue Culture Techniques , Gap Junction beta-1 Protein , Gap Junction alpha-5 Protein , Gap Junction alpha-4 Protein
16.
Am J Pathol ; 159(3): 993-1008, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11549592

ABSTRACT

The known responses of vascular endothelial growth factor (VEGF) are mediated through VEGF receptor-2 (VEGFR-2/KDR) in endothelial cells. However, it is unknown whether VEGFR-1 (Flt-1) is an inert decoy or a signaling receptor for VEGF during physiological or pathological angiogenesis. Here we report that VEGF-stimulated nitric oxide (NO) release is inhibited by blockade of VEGFR-1 and that VEGFR-1 via NO negatively regulates of VEGFR-2-mediated proliferation and promotes formation of capillary networks in human umbilical vein endothelial cells (HUVECs). Inhibition of VEGFR-1 in a murine Matrigel angiogenesis assay induced large aneurysm-like structures. VEGF-induced capillary growth over 14 days was inhibited by anti-VEGFR-2-blocking antibody as determined by reduced tube length between capillary connections (P < 0.0001) in an in vitro angiogenesis assay. In contrast, loss of VEGFR-1 activity with a neutralizing anti-VEGFR-1 antibody resulted in an increase in the accumulation of endothelial cells (P < 0.0001) and a dramatic decrease in the number of capillary connections that were restored by the addition of NO donor. Porcine aortic endothelial (PAE) cells expressing human VEGFR-1 but not VEGFR-2 plated on growth factor-reduced Matrigel rearranged into tube-like structures that were prevented by anti-VEGFR-1 antibody or a cGMP inhibitor. VEGF stimulated NO release from VEGFR-1- but not VEGFR-2-transfected endothelial cells and placenta growth factor-1 stimulated NO release in HUVECs. Blockade of VEGFR-1 increased VEGF-mediated HUVEC proliferation that was inhibited by NO donors, and potentiated by NO synthase inhibitors. These data indicate that VEGFR-1 is a signaling receptor that promotes endothelial cell differentiation into vascular tubes, in part by limiting VEGFR-2-mediated endothelial cell proliferation via NO, which seems to be a molecular switch for endothelial cell differentiation.


Subject(s)
Endothelial Growth Factors/physiology , Endothelium, Vascular/physiology , Lymphokines/physiology , Neovascularization, Physiologic/physiology , Nitric Oxide/physiology , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Cell Division/drug effects , Cell Division/physiology , Cells, Cultured , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/cytology , Humans , Lymphokines/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type III , Nitroso Compounds/pharmacology , Penicillamine/analogs & derivatives , Penicillamine/pharmacology , Protein Isoforms/immunology , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/immunology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/immunology , Receptors, Growth Factor/genetics , Receptors, Growth Factor/physiology , Receptors, Vascular Endothelial Growth Factor , Transfection , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-1 , Vascular Endothelial Growth Factors
17.
Histol Histopathol ; 16(2): 359-75, 2001 04.
Article in English | MEDLINE | ID: mdl-11332691

ABSTRACT

Placental villous development requires the co-ordinated action of angiogenic factors on both endothelial and trophoblast cells. Like vascular endothelial growth factor (VEGF), VEGF-C increases vascular permeability, stimulates endothelial cell proliferation and migration. In the present study, we investigated the expression of VEGF-C and its receptors VEGFR-3 and VEGFR-2 in normal and intrauterine growth-restricted (IUGR) placenta. Immunolocalisation studies showed that like VEGF and VEGFR-1, VEGF-C, VEGFR-3 and VEGFR-2 co-localised to the syncytiotrophoblast, to cells in the maternal decidua, as well as to the endothelium of the large placental blood vessels. Western blot analysis demonstrated a significant decrease in placental VEGF-C and VEGFR-3 protein expression in severe IUGR as compared to gestationally-matched third trimester pregnancies. Conditioned medium from VEGF-C producing pancreatic carcinoma (Suit-2) and endometrial epithelial (Hec-1B) cell lines caused an increased association of the phosphorylated extracellular signal regulated kinase (ERK) in VEGFR-3 immunoprecipitates from spontaneously transformed first trimester trophoblast cells. VEGF121 caused dose-dependant phosphorylation of VEGFR-2 in trophoblast cells as well as stimulating DNA synthesis. In addition, premixing VEGF165 with heparin sulphate proteoglycan potentiated trophoblast proliferation and the association of phospho-ERK with the VEGFR-2 receptor. VEGF165-mediated DNA synthesis was inhibited by anti-VEGFR-2 neutralising antibody. The results demonstrate functional VEGFR-2 and VEGFR-3 receptors on trophoblast and suggest that the decreased expression of VEGF-C and VEGFR-3 may contribute to the abnormal villous development observed in IUGR placenta.


Subject(s)
Endothelial Growth Factors/metabolism , Endothelium, Vascular/metabolism , Fetal Growth Retardation/metabolism , Placenta/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Trophoblasts/metabolism , Blotting, Western , Carcinoma, Pancreatic Ductal/pathology , Culture Media, Conditioned , Endometrium/cytology , Endothelium, Vascular/cytology , Female , Humans , Immunohistochemistry , Infant, Newborn , Lymphokines , Matched-Pair Analysis , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Pregnancy , Pregnancy Proteins/metabolism , Protein Isoforms , Receptors, Vascular Endothelial Growth Factor , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor C , Vascular Endothelial Growth Factor Receptor-3 , Vascular Endothelial Growth Factors
18.
Am J Pathol ; 158(1): 265-73, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11141500

ABSTRACT

Vascular endothelial growth factor (VEGF) receptors are present on nonendothelial cells suggesting that VEGF may mediate nonendothelial effects during organogenesis and tumorigenesis. Here we show that VEGF receptor-1 (VEGFR-1) negatively regulates VEGFR-2-mediated proliferation via nitric oxide (NO) in an epithelial cancer cell line ECV304. Cell proliferation was assessed by [(3)H]thymidine incorporation, fluorescent-activated cell-sorting analysis, and cell number using a Coulter Counter. Total NO generated by the action of nitric oxide synthase was measured by Seivers NOA 280 Nitric Oxide Chemiluminescence Analyser. VEGF (1 ng/ml) stimulated DNA synthesis and increased ECV304 cell number in a manner that was inhibited by a neutralizing anti-VEGFR-2 antibody. In contrast, VEGF (50 ng/ml) stimulated NO release in a manner that was inhibited by functionally neutralizing anti-VEGFR-1 antibody. Blockage of the VEGFR-1 receptor signal with anti-VEGFR-1 stimulated DNA synthesis and increased cell number. Cell-cycle analysis showed that inhibition of VEGFR-1 increased the transition from G(1) to S phase whereas inhibition of VEGFR-2 blocked the VEGF-mediated transition from G(1) to S phase. Finally, the addition of NO donors suppressed both VEGF-mediated proliferation and the increase in growth after blockade of VEGFR-1. Conversely, inhibition of VEGF mediated NO release by nitric oxide synthase inhibitor, L-monomethyl-L-arginine, restored the mitogenic effect of VEGF. These findings identify a dose-dependent reciprocal regulatory mechanism for VEGF via its two receptors. It shows that VEGFR-1 induces cell cytostasis via NO and as such is a suitable target for molecular strategies suppressing tumorigenesis.


Subject(s)
Mitogens/physiology , Penicillamine/analogs & derivatives , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Receptors, Growth Factor/physiology , Antibodies, Monoclonal/pharmacology , Cell Cycle/drug effects , Cell Division/drug effects , Cell Line , DNA/biosynthesis , DNA/drug effects , Dose-Response Relationship, Drug , Endothelial Growth Factors/pharmacology , Enzyme Inhibitors/pharmacology , Humans , Lymphokines/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitroprusside/pharmacology , Penicillamine/pharmacology , Proto-Oncogene Proteins/immunology , Receptor Protein-Tyrosine Kinases/immunology , Receptors, Growth Factor/immunology , Receptors, Vascular Endothelial Growth Factor , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-1 , Vascular Endothelial Growth Factors , omega-N-Methylarginine/pharmacology
19.
Placenta ; 21 Suppl A: S16-24, 2000.
Article in English | MEDLINE | ID: mdl-10831117

ABSTRACT

Morphological studies show poor placental vascular development and an increase in the mitotic index of cytotrophoblast cells in intrauterine growth restriction (IUGR). We hypothesized that the reported relatively high oxygen level in the intervillous space in contact with IUGR placental villi will limit angiogenesis by changes in vascular endothelial growth factor (VEGF) and placenta growth factor (PIGF) expression and function. Western immunoblot analysis demonstrates a diametric expression of PIGF and VEGF proteins throughout pregnancy, with P1GF levels increasing and VEGF levels decreasing, consistent with placental oxygenation. PIGF mRNA and protein is increased in IUGR as compared to gestationally matched normal placentae. Increasing oxygen tension upregulates P1GF protein in term placental villous explants, whereas hypoxia downregulates P1GF and VEGFR-1 (Flt-1) autophosphorylation in term trophoblast choriocarcinoma cell line (BeWo). Levels of soluble Flt-1 (sFlt-1) protein in supernatant of term villous explants were upregulated by 1 per cent hypoxia, whereas hyperoxia (40 per cent) decreased sFlt-1 levels, indicating that under conditions of increasing oxygen tension, PlGF function may remain unopposed. The addition of PlGF-1 to a spontaneously transformed first trimester cytotrophoblast cell line (ED27) stimulated cell proliferation while PlGF-2 had little effect. In contrast, the addition of PlGF-1 had little effect on endothelial cell proliferation while this was inhibited by PIGF-2. Taken together these changes provide a molecular explanation for the observed poor angiogenesis in the pathogenesis of IUGR.


Subject(s)
Endothelial Growth Factors/physiology , Lymphokines/physiology , Oxygen/physiology , Placenta/blood supply , Placenta/physiology , Pregnancy Proteins/physiology , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Animals , Endothelial Growth Factors/genetics , Female , Fetal Hypoxia/pathology , Fetal Hypoxia/physiopathology , Humans , Lymphokines/genetics , Neovascularization, Physiologic , Placenta Growth Factor , Pregnancy , Pregnancy Proteins/genetics , Proto-Oncogene Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Growth Factor/genetics , Receptors, Growth Factor/physiology , Receptors, Vascular Endothelial Growth Factor , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-1 , Vascular Endothelial Growth Factors
20.
Am J Pathol ; 156(6): 2185-99, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10854239

ABSTRACT

Human placental development involves coordinated angiogenesis and trophoblast outgrowth that are compromised in intrauterine growth restriction (IUGR). As Tie-2((-/-)) mice exhibit growth retardation and vascular network malformation, the expression of Tie-2 and its ligands, angiopoietin-1 (Ang-1) and angiopoietin-2 (Ang-2), were investigated in human placenta from normal pregnancies and those complicated by severe IUGR. Ribonucleotide protection assays showed no significant change in the expression of Ang-2 mRNA between gestationally matched normal and IUGR placentas; however, immunoblots revealed that Ang-2 protein was significantly decreased in IUGR, suggesting that this may contribute to the abnormal development of the villous vasculature. In situ hybridization studies showed that Ang-1 and Tie-2 were detected in the cyto/syncytiotrophoblast bilayer in first-trimester placenta, whereas Ang-2 mRNA was restricted to the cytotrophoblast, suggesting their role in trophoblast function. At term, Ang-1 mRNA and immunoreactive protein were restricted to the paravascular tissues of the primary stem villi, supporting its role in vessel maturation. In contrast, Ang-2 was expressed throughout the term villous core, perhaps to permit the developing placental vascular network to remain in a state of fluidity. As these studies also revealed that trophoblast, in addition to endothelial cells, expressed Tie-2 receptors, we investigated the potential role of Ang-1/Ang-2 on trophoblast proliferation, migration, and the release of NO. Using spontaneously transformed first-trimester trophoblast cell lines that exhibit cytotrophoblast-like (ED(27)) and extravillous trophoblast-like (ED(77)) properties, we show that the addition of Ang-2 (250 ng/ml) stimulated DNA synthesis in ED(27) trophoblast cells and triggered the release of NO. Ang-1 stimulated trophoblast (ED(77)) migration in a dose-dependent manner that was inhibited by recombinant Tie-2-FC. These data thus imply, for the first time, a specific role for angiopoietins as regulators of trophoblast behavior in the development of the utero/fetoplacental circulation, an action independent of their well-established roles in vascular endothelium.


Subject(s)
Membrane Glycoproteins/pharmacology , Neoplasm Proteins/physiology , Placenta/physiology , Proteins/pharmacology , Proto-Oncogene Proteins , Trophoblasts/drug effects , Angiopoietin-1 , Angiopoietin-2 , Cell Division/drug effects , Cell Movement/drug effects , Female , Fetal Growth Retardation/metabolism , Humans , Immunologic Techniques , In Situ Hybridization , Membrane Glycoproteins/genetics , Nitric Oxide/metabolism , Placenta/metabolism , Pregnancy , Proteins/genetics , RNA, Messenger/metabolism , Receptor, TIE-2 , Reference Values , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution , Trophoblasts/cytology , Trophoblasts/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...