Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Stem Cell Investig ; 6: 3, 2019.
Article in English | MEDLINE | ID: mdl-30976600

ABSTRACT

BACKGROUND: The placenta is an abundant source of mesenchymal stem/stromal cells (MSC), but our understanding of their functional properties remains limited. We previously created a placental-derived chorionic MSC (CMSC) cell line to overcome the difficulties associated with conducting extensive ex vivo optimization and experimental work on primary cells. The aim of this study was to characterize the migratory behavior of the CMSC29 cell line in vitro. METHODS: Stimulators of MSC migration, including two cytokines, stromal cell-derived factor-1α (SDF-1α) and hepatocyte growth factor (HGF), and a pharmacological agent, valproic acid (VPA), were tested for their ability to stimulate CMSC29 cell migration. Assessment of cell migration was performed using the xCELLigence Real-Time Cell Analyzer (RTCA). RESULTS: There was no significant increase in CMSC29 cell migration towards serum free medium with increasing concentration gradients of SDF-1α or HGF. In contrast, treating CMSC29 cells with VPA alone significantly increased their migration towards serum free medium. CONCLUSIONS: Immortalized CMSC29 cells retain important properties of primary CMSC, but their migratory properties are altered. CMSC29 cells do not migrate in response to factors that reportedly stimulate primary MSC/CMSC migration. However, CMSC29 increase their migration in response to VPA treatment alone. Further studies are needed to determine the mechanism by which VPA acts alone to stimulate CMSC29 migration. Still, this study provides evidence that VPA pre-treatment may improve the benefits of cell-based therapies that employ certain MSC sub-types.

2.
Stem Cell Investig ; 6: 2, 2019.
Article in English | MEDLINE | ID: mdl-30842969

ABSTRACT

BACKGROUND: To initiate tissue repair, mesenchymal stem/stromal cells (MSCs) must enter the blood stream, migrate to the targeted area, cross the endothelial barrier and home to the damaged tissue. This process is not yet fully understood in humans and thus, the aim of this study was to develop an ex vivo placental vessel perfusion method to examine human MSC movement from a blood vessel into human tissue. This will provide a better understanding of MSC migration, movement through the endothelial barrier and engraftment into target tissue, in a setting that more closely represents the in vivo state, compared with conventional in vitro human cell culture models. Moreover, important similarities and differences to animal experimental model systems may be revealed by this method. METHODS: Human placental hTERT transformed MSC lines were labelled with live-cell fluorescence dyes, and then perfused into term human placental blood vessel. After labelled MSCs were perfused into the vessel, the vessel was dissected from the placenta and incubated at cell growth conditions. Following incubation, the vessel was washed thoroughly to remove unattached, labelled MSCs and then snap frozen for sectioning. After sectioning, immunofluorescence staining of the endothelium was carried out to detect if labelled MSCs crossed the endothelial barrier. RESULTS: Twelve placental vessel perfusions were successfully completed. In eight of the twelve perfused vessels, qualitative assessment of immunofluorescence in sections (n=20, 5 µm sections/vessel) revealed labelled MSCs had crossed the endothelial barrier. CONCLUSIONS: The human placental ex vivo vessel perfusion method could be used to assess human MSC migration into human tissue. Cells of the MSC lines were able to adhere and transmigrate through the endothelial barrier in a manner similar to that of leukocytes. Notably, cells that transmigrated remained in close proximity to the endothelium, which is consistent with the reported MSC vascular niche in placental blood vessels.

3.
Pregnancy Hypertens ; 12: 189-193, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29203340

ABSTRACT

Decidual stromal cells form the largest proportion of maternal cells at the maternal-fetal interface. Our aim was to investigate the role of the pre-eclampsia associated decidual activin receptor, ACVR2A, in regulating trophoblast functions at this interface. St-T1b and HTR-8/SVneo cell lines were used to model decidual stromal and trophoblast cells respectively. St-T1b conditioned medium inhibited HTR-8/SVneo adhesion, proliferation, migration and invasion; all effects that were attenuated by decidual ACVR2A siRNA transfection. These findings suggest that altered decidual ACVR2A expression perturbs the maternal-fetal crosstalk involved in regulating trophoblast function at the interface, which may affect placentation and lead to pre-eclampsia.


Subject(s)
Activin Receptors, Type II/metabolism , Cell Adhesion , Cell Movement , Cell Proliferation , Decidua/metabolism , Paracrine Communication , Stromal Cells/metabolism , Trophoblasts/metabolism , Activin Receptors, Type II/genetics , Cell Line , Culture Media, Conditioned/metabolism , Decidua/pathology , Female , Humans , Placentation , Pre-Eclampsia/genetics , Pre-Eclampsia/metabolism , Pre-Eclampsia/pathology , Pregnancy , Signal Transduction , Stromal Cells/pathology , Trophoblasts/pathology
4.
Placenta ; 59: 131-138, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28697979

ABSTRACT

INTRODUCTION: Chorionic mesenchymal stem/stromal cells (CMSC) can be isolated from the placenta in large numbers. Although their functions are yet to be fully elucidated, they have a role in tissue development and repair. To fulfil such a role, CMSC must be able to migrate to the microenvironment of the injury site. This process is not fully understood and the aim of this study therefore, was to examine in vitro CMSC migration in response to tissue inflammation and hypoxic conditioning. METHODS: CMSC were derived from the chorionic villi. A trans-endothelium migration (TEM) assay was used to study CMSC migration through an activated endothelial cell monolayer using the HMEC-1 cell line. A cytokine array was used to identify and compare the cytokine production profile of activated versus non-activated HMEC-1. RESULTS: There were significant changes in cytokine production by HMEC-1 cells following lipopolysaccharide (LPS) treatment and hypoxic conditioning. Despite this, results from the TEM assay showed no significant change in the average number of CMSC that migrated through the LPS activated HMEC-1 layer compared to the untreated control. Furthermore, there was no significant change in the average number of CMSC that migrated through the HMEC-1 monolayer when exposed to hypoxic (1% O2), normoxic (8% O2) or hyperoxic (21% O2) conditions. CONCLUSION: These data suggest that cell functions such as transendothelial migration can vary between MSC derived from different tissues in response to the same biological cues.


Subject(s)
Cell Movement , Endothelial Cells/physiology , Hypoxia/physiopathology , Mesenchymal Stem Cells/physiology , Placenta/cytology , Cell Line , Cell Migration Assays , Cytokines/metabolism , Female , Humans , Lipopolysaccharides , Pregnancy
5.
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
6.
Pregnancy Hypertens ; 5(4): 346-53, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26597752

ABSTRACT

OBJECTIVES: Activin A, a TGFß family member, circulates in the maternal blood at increasing concentrations throughout gestation during a healthy pregnancy. The circulating concentration of activin A is further increased in pre-eclampsia (PE), a hypertensive disorder of pregnancy that is marked by systemic maternal vascular endothelial cell dysfunction. The effect of increasing activin A concentrations on the maternal vascular endothelium is unknown. The study aim was to investigate the effect of physiological and pathological activin A concentrations observed in normotensive and PE pregnancies respectively, on vascular endothelial cell function. METHODS AND RESULTS: Immunostaining demonstrated the presence of the activin A receptor, ACVR2A, in SGHEC-7 cells used to model the vascular endothelium. SGHEC-7 cells were treated with activin A concentrations representative of concentrations throughout gestation in normotensive (0-10ng/ml) and PE (50ng/ml) pregnancies. xCELLigence functional assays revealed that normotensive activin A concentrations increased SGHEC-7 proliferation and migration, which was inhibited by PE concentrations. Additionally, fluorescence based assays showed that PE concentrations increased endothelial permeability. None of the tested activin A concentrations affected cell apoptosis. PE concentrations also resulted in an imbalance of the vasoactive factors eNOS, PTGIS and EDN1, as determined by quantitative real-time polymerase chain reaction and enzyme-linked immunosorbent assays. CONCLUSION: Compared with normotensive activin A concentrations, the higher PE activin A concentrations resulted in abnormal endothelial functions, which may contribute to the systemic maternal vascular endothelial cell dysfunction observed in the disorder.


Subject(s)
Activins/blood , Endothelium, Vascular , Pre-Eclampsia/blood , Pre-Eclampsia/diagnosis , Vascular Endothelial Growth Factor Receptor-1/blood , Activin Receptors, Type II/blood , Adult , Biomarkers/blood , Cell Culture Techniques , Endothelium, Vascular/physiopathology , Female , Humans , Pre-Eclampsia/physiopathology , Predictive Value of Tests , Pregnancy , Sensitivity and Specificity
7.
Placenta ; 36(11): 1300-9, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26343951

ABSTRACT

INTRODUCTION: Arresten and canstatin are endogenous anti-angiogenic factors derived from type IV collagen α-chains COL4A1 and COL4A2 respectively. While their functions are explored in cancer studies, little is known about their role in pregnancy. Pre-eclampsia (PE) is a common, serious hypertensive disorder of pregnancy that is characterised by systemic endothelial dysfunction. COL4A1 and COL4A2 are maternal PE susceptibility genes that have increased mRNA expression in PE decidua. Our study aim was to determine the levels of arresten and canstatin in plasma and decidua from PE and gestational age matched normotensive patients. METHODS: Plasma was collected from normotensive (n = 44) and PE (n = 39) women during the second and third trimesters of pregnancy. Third trimester decidua was collected at delivery from normotensive and PE women (n = 4 each). Levels of arresten and canstatin were determined by Western immunoblotting. RESULTS: Arresten levels were significantly increased in second and third trimester PE plasma, and in third trimester PE decidua (p < 0.05). Third trimester PE plasma arresten levels also significantly correlated with the need for MgSO4 treatment, where a 1.7 fold increase was observed in patients requiring MgSO4 treatment (p < 0.05). No significant change in canstatin levels was observed between normotensive and PE patients. DISCUSSION: This is the first study to report significant increases in the levels of collagen fragment arresten in PE plasma and decidua. Given its significant increase before the onset of clinical disease and associations with clinical severity in the third trimester, arresten may be a useful biomarker for predicting PE and monitoring its severity.


Subject(s)
Collagen Type IV/blood , Pre-Eclampsia/blood , Adult , Case-Control Studies , Female , Humans , Pregnancy , Pregnancy Trimester, Second/blood , Pregnancy Trimester, Third/blood
8.
PLoS One ; 10(5): e0128230, 2015.
Article in English | MEDLINE | ID: mdl-26010865

ABSTRACT

BACKGROUND: Preeclampsia (PE) is a serious hypertensive pregnancy disorder with a significant genetic component. Numerous genetic studies, including our own, have yielded many susceptibility genes from distinct functional groups. Additionally, transcriptome profiling of tissues at the maternal-fetal interface has likewise yielded many differentially expressed genes. Often there is little overlap between these two approaches, although genes identified in both approaches are significantly associated with PE. We have thus taken a novel integrative bioinformatics approach of analysing pathways common to the susceptibility genes and the PE transcriptome. METHODS: Using Illumina Human Ht12v4 and Wg6v3 BeadChips, transcriptome profiling was conducted on n = 65 normotensive and n = 60 PE decidua basalis tissues collected at delivery. The R software package libraries lumi and limma were used to preprocess transcript data for pathway analysis. Pathways were analysed and constructed using Pathway Studio. We examined ten candidate genes, which are from these functional groups: activin/inhibin signalling-ACVR1, ACVR1C, ACVR2A, INHA, INHBB; structural components-COL4A1, COL4A2 and M1 family aminopeptidases-ERAP1, ERAP2 and LNPEP. RESULTS/CONCLUSION: Major common regulators/targets of these susceptibility genes identified were AGT, IFNG, IL6, INHBA, SERPINE1, TGFB1 and VEGFA. The top two categories of pathways associated with the susceptibility genes, which were significantly altered in the PE decidual transcriptome, were apoptosis and cell signaling (p < 0.001). Thus, susceptibility genes from distinct functional groups share similar downstream pathways through common regulators/targets, some of which are altered in PE. This study contributes to a better understanding of how susceptibility genes may interact in the development of PE. With this knowledge, more targeted functional analyses of PE susceptibility genes in these key pathways can be performed to examine their contributions to the pathogenesis and severity of PE.


Subject(s)
Genetic Predisposition to Disease , Genome-Wide Association Study , Pre-Eclampsia/genetics , Signal Transduction , Transcriptome , Adult , Female , Humans , Pre-Eclampsia/metabolism , Pregnancy
9.
Angiogenesis ; 18(2): 115-23, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25433512

ABSTRACT

Placental angiogenesis is critical to the success of human pregnancy. Angiogenesis is defined as the formation of new blood vessels from existing vasculature. Angiogenesis is necessary for the establishment of adequate placental perfusion, which is important for providing the optimum in utero environment to support fetal development. Defective placental angiogenesis is associated with several pregnancy complications, the most clinically important of which is preeclampsia; the multisystem disorder is characterized by maternal hypertension, proteinuria, and endothelial dysfunction. Here, we review our current understanding of several key angiogenic factors that are associated with placental angiogenesis. We also discuss their importance with respect to preeclampsia, where aberrant expression and release of these factors into the maternal circulation is thought to contribute to the pathogenesis and pathophysiology of preeclampsia.


Subject(s)
Angiogenic Proteins/physiology , Placenta/physiology , Pre-Eclampsia/etiology , Female , Humans , Pregnancy
10.
Mol Hum Reprod ; 19(10): 665-75, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23761267

ABSTRACT

Abnormal trophoblast function is associated with human fetal growth restriction (FGR). Targeted disruption of homeobox gene transforming growth ß-induced factor (TGIF-1) results in placental dysfunction in the mouse. The role of human TGIF-1 in placental cell function is unknown. The aims of this study were to determine the expression of TGIF-1 in human idiopathic FGR-affected placentae compared with gestation-matched controls (GMC), to elucidate the functional role of TGIF-1 in trophoblasts and to identify its downstream targets. Real-time PCR and immunoblotting revealed that TGIF-1 mRNA and protein expression was significantly increased in FGR-affected placentae compared with GMC (n = 25 in each group P < 0.05). Immunoreactive TGIF-1 was localized to the villous cytotrophoblasts, syncytiotrophoblast, microvascular endothelial cells and in scattered stromal cells in both FGR and GMC. TGIF-1 inactivation in BeWo cells using two independent siRNA resulted in significantly decreased mRNA and protein of trophoblast differentiation markers, human chorionic gonadotrophin (CGB/hCG), syncytin and 3ß-hydroxysteroid dehydrogenase/3ß-honest significant difference expression. Our data demonstrate that homeobox gene TGIF-1 is a potential up-stream regulator of trophoblast differentiation and the altered TGIF-1 expression may contribute to aberrant villous trophoblast differentiation in FGR.


Subject(s)
Fetal Growth Retardation/metabolism , Homeodomain Proteins/metabolism , Repressor Proteins/metabolism , Trophoblasts/cytology , Trophoblasts/metabolism , 3-Hydroxysteroid Dehydrogenases/genetics , 3-Hydroxysteroid Dehydrogenases/metabolism , Adult , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Chorionic Gonadotropin/genetics , Chorionic Gonadotropin/metabolism , Female , Fetal Growth Retardation/genetics , Gene Products, env/genetics , Gene Products, env/metabolism , Homeodomain Proteins/genetics , Humans , Mice , Pregnancy , Pregnancy Proteins/genetics , Pregnancy Proteins/metabolism , Repressor Proteins/genetics
11.
Hum Reprod ; 28(6): 1497-507, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23477905

ABSTRACT

STUDY QUESTION: What are the effects of the eotaxin group of chemokines (CCL11, CCL24 and CCL26) on extravillous trophoblast (EVT) functions important during uterine decidual vessel remodelling? SUMMARY ANSWER: CCL11, CCL24 and CCL26 can regulate EVT migration, invasion and adhesion, highlighting a potential regulatory role for these chemokines during uterine decidual spiral arteriole remodelling in the first trimester of human pregnancy. WHAT IS KNOWN ALREADY: A successful human pregnancy depends on adequate remodelling of the uterine decidual spiral arterioles, a process carried out by EVT which invade from the placenta. The invasion by EVT into the maternal uterine decidual vessels is regulated by the interaction of many factors including members of the chemokine subfamily of cytokines. STUDY DESIGN, SIZE, DURATION: This study used the HTR8/SVneo cell line as a model for invasive EVT. All experiments were repeated on at least three separate occasions. PARTICIPANTS/MATERIALS, SETTING, METHODS: The effect of recombinant human CCL11, CCL24 and CCL26 on EVT migration and invasive potential was measured using the xCELLigence real-time system, wound-healing and Matrigel invasion assays, zymography to measure MMP activity and reverse zymography to measure TIMP activity. A commercially available adhesion assay was used to assess EVT adhesion to extracellular matrix proteins. MAIN RESULTS AND THE ROLE OF CHANCE: All the three eotaxins were found to significantly stimulate migration of the EVT-derived cell line HTR8/SVneo (P < 0.05) with no significant changes in cell number following treatment with each chemokine (P > 0.05). All the three eotaxins significantly increased HTR8/SVneo invasion (P < 0.05) and MMP2 activity (P < 0.05) without any effects on TIMP2 activity (P > 0.05). All the three eotaxins significantly increased HTR8/SVneo cell binding to collagen IV (P < 0.05) and fibronectin (P < 0.05). LIMITATIONS, REASONS FOR CAUTION: This work has been conducted in vitro with a commonly used cell line model of EVT, HTR8/SVneo. WIDER IMPLICATIONS OF THE FINDINGS: This study is the first to comprehensively examine the effects of the eotaxin group of chemokines on EVT functions and demonstrates that all the three eotaxins have the ability to regulate EVT functions critical to their role in vessel remodelling. This identifies a new role for the eotaxin group of chemokines during placentation.


Subject(s)
Chemokine CCL11/pharmacology , Chemokine CCL24/pharmacology , Chemokines, CC/pharmacology , Decidua/drug effects , Trophoblasts/drug effects , Arterioles/drug effects , Cell Adhesion/drug effects , Cell Line , Cell Movement/drug effects , Chemokine CCL11/physiology , Chemokine CCL24/physiology , Chemokine CCL26 , Chemokines, CC/physiology , Collagen , Decidua/blood supply , Drug Combinations , Female , Humans , Laminin , Proteoglycans , Trophoblasts/cytology
12.
Fetal Diagn Ther ; 32(4): 225-30, 2012.
Article in English | MEDLINE | ID: mdl-22906990

ABSTRACT

Intrauterine growth restriction (IUGR) is an adverse pregnancy outcome associated with significant perinatal and pediatric morbidity and mortality, and an increased risk of chronic disease later in adult life. While a number of maternal, fetal and environmental factors are known causes of IUGR, the majority of IUGR cases are of unknown cause. These IUGR cases are frequently associated with placental insufficiency, possibly as a result of placental maldevelopment. Understanding the molecular mechanisms of abnormal placental development in IUGR associated with placental insufficiency is therefore of increasing importance. Here, we review our understanding of transcriptional control of normal placental development as well as human IUGR associated with placental insufficiency. We also assess the potential for understanding transcriptional control as a means for revealing new molecular targets for the detection, diagnosis and clinical management of IUGR associated with placental insufficiency.


Subject(s)
Genes, Homeobox , Placental Insufficiency/genetics , Animals , Female , Fetal Growth Retardation/etiology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Placenta/metabolism , Placental Insufficiency/metabolism , Placental Insufficiency/physiopathology , Placentation , Pregnancy
13.
Mol Cell Endocrinol ; 349(2): 138-44, 2012 Feb 26.
Article in English | MEDLINE | ID: mdl-22051447

ABSTRACT

During early human pregnancy, extravillous trophoblast (EVT) cells from the placenta invade the uterine decidual spiral arterioles and mediate the remodelling of these vessels such that a low pressure, high blood flow can be supplied to the placenta. This is essential to facilitate normal growth and development of the foetus. Defects in remodelling can manifest as the serious pregnancy complication pre-eclampsia. During the period of vessel remodelling three key pregnancy-associated hormones, human chorionic gonadotrophin (hCG), progesterone (P(4)) and oestradiol (E(2)), are found in high concentrations at the maternal-foetal interface. Potentially these hormones may control EVT movement and thus act as regulators of vessel remodelling. This review will discuss what is known about how these hormones affect EVT proliferation, migration and invasion during vascular remodelling and the potential relationship between hCG, P(4), E(2) and the development of pre-eclampsia.


Subject(s)
Arterioles/physiology , Chorionic Gonadotropin/metabolism , Chorionic Villi/blood supply , Decidua/blood supply , Estradiol/metabolism , Pre-Eclampsia/metabolism , Progesterone/metabolism , Trophoblasts/physiology , Arterioles/pathology , Chorionic Villi/physiology , Decidua/physiology , Female , Fetus , Humans , Maternal-Fetal Exchange , Pre-Eclampsia/physiopathology , Pregnancy
14.
Rev. bras. ginecol. obstet ; 33(11): 367-375, nov. 2011. tab
Article in English | LILACS | ID: lil-611360

ABSTRACT

Preeclampsia, which affects about 3 to 5 percent of pregnant women, is the most frequent medical complication in pregnancy and the most important cause of maternal and perinatal morbidity and mortality. During the past three decades, numerous clinical, biophysical, and biochemical screening tests have been proposed for the early detection of preeclampsia. Literature shows large discrepancies in the sensitivity and predictive value of several of these tests. No single screening test used for preeclampsia prediction has gained widespread acceptance into clinical practice. Instead, its value seems to be in increasing the predictive value of panels of tests, which include other clinical measurements. The aim of this review was to examine the combination of maternal risk factors, mean arterial blood pressure, and uterine artery Doppler, together with biomarkers in the preeclampsia prediction.


A pré-eclâmpsia, que afeta cerca de 3 a 5 por cento das mulheres grávidas, é a mais frequente complicação médica durante a gestação e a mais importante causa de morbidade e mortalidade maternal e perinatal. Durante as últimas três décadas, numerosos testes de rastreamento clínicos, biofísicos e bioquímicos foram propostos para a detecção precoce da pré-eclâmpsia. A literatura mostra grandes discrepâncias na sensibilidade e no valor preditivo de muitos desses testes. Nenhum teste de rastreamento isolado usado para a predição da pré-eclâmpsia tem ganhado ampla aceitação na prática clínica. Ao contrário, parece que o valor preditivo aumenta com a inclusão de um painel de testes, os quais incluem outros parâmetros clínicos. O objetivo desta revisão foi examinar a combinação dos fatores de risco maternos, a pressão arterial média, o Doppler das artérias uterinas com os marcadores séricos, na predição da pré-eclâmpsia.


Subject(s)
Female , Humans , Pregnancy , Pre-Eclampsia/diagnosis , Biomarkers/analysis , Early Diagnosis , Ultrasonography, Prenatal
15.
Mol Cell Endocrinol ; 342(1-2): 73-80, 2011 Aug 06.
Article in English | MEDLINE | ID: mdl-21664947

ABSTRACT

Remodelling of the uterine vasculature during the first trimester of human pregnancy requires invasion of trophoblast from the placenta into decidual spiral arterioles. The pregnancy-associated hormones human chorionic gonadotropin (hCG), progesterone (P(4)) and oestradiol (E(2)) are present at high concentrations at the maternal-fetal interface during the remodelling period and thus may contribute to the regulation of trophoblast movement. This study examined the effects of these hormones on trophoblast functions. HTR8/SVneo cells were treated with hCG (5-100mIU/mL), P(4) (20nM-20µM) or E(2) (0.07-734nM). hCG significantly stimulated migration and MMP-9 activity but did not affect cell numbers. P(4) significantly inhibited migration, MMP-2 and -9 activity and reduced cell numbers. E(2) had no effect on migration, MMP activity or cell numbers. We conclude that hCG and P(4), but not E(2), play direct roles in controlling trophoblast invasion, acting as positive and negative stimuli respectively to regulate trophoblast movement during vascular remodelling in early pregnancy.


Subject(s)
Chorionic Gonadotropin/pharmacology , Estradiol/pharmacology , Progesterone/pharmacology , Trophoblasts/drug effects , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Pregnancy , Recombinant Proteins/pharmacology , Trophoblasts/cytology , Trophoblasts/physiology , Uterus/blood supply
16.
Rev Bras Ginecol Obstet ; 33(11): 367-75, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22267116

ABSTRACT

Preeclampsia, which affects about 3 to 5% of pregnant women, is the most frequent medical complication in pregnancy and the most important cause of maternal and perinatal morbidity and mortality. During the past three decades, numerous clinical, biophysical, and biochemical screening tests have been proposed for the early detection of preeclampsia. Literature shows large discrepancies in the sensitivity and predictive value of several of these tests. No single screening test used for preeclampsia prediction has gained widespread acceptance into clinical practice. Instead, its value seems to be in increasing the predictive value of panels of tests, which include other clinical measurements. The aim of this review was to examine the combination of maternal risk factors, mean arterial blood pressure, and uterine artery Doppler, together with biomarkers in the preeclampsia prediction.


Subject(s)
Pre-Eclampsia/diagnosis , Biomarkers/analysis , Early Diagnosis , Female , Humans , Pregnancy , Ultrasonography, Prenatal
17.
Am J Pathol ; 177(4): 2103-15, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20802175

ABSTRACT

During the first trimester of pregnancy, the uterine spiral arteries are remodeled, creating heavily dilated conduits that lack maternal vasomotor control but allow the placenta to meet an increasing requirement for nutrients and oxygen. To effect permanent vasodilatation, the internal elastic lamina and medial elastin fibers must be degraded. In this study, we sought to identify the elastolytic proteases involved in this process. Primary first-trimester cytotrophoblasts (CTBs) derived from the placenta exhibited intracellular and membrane-associated elastase activity; membrane-associated activity was primarily attributable to matrix metalloproteinases (MMP). Indeed, Affymetrix microarray analysis and immunocytochemistry implicated MMP-12 (macrophage metalloelastase) as a key mediator of elastolysis. Cultured human aortic smooth muscle cells (HASMCs) exhibited constitutive membrane-associated elastase activity and inducible intracellular elastase activity; these cells also expressed MMP-12 protein. Moreover, a specific inhibitor of MMP-12 significantly reduced CTB- and HASMC-mediated elastolysis in vitro, to 31.7 ± 10.9% and 23.3 ± 8.7% of control levels, respectively. MMP-12 is expressed by both interstitial and endovascular trophoblasts in the first-trimester placental bed and by vascular SMCs (VSMCs) in remodeling spiral arteries. Perfusion of isolated spiral artery segments with CTB-conditioned medium stimulated MMP-12 expression in medial VSMCs. Our data support a model in which trophoblasts and VSMCs use MMP-12 cooperatively to degrade elastin during vascular remodeling in pregnancy, with the localized release of elastin peptides and CTB-derived factors amplifying elastin catabolism.


Subject(s)
Elastin/metabolism , Matrix Metalloproteinase 12/metabolism , Muscle, Smooth, Vascular/enzymology , Trophoblasts/enzymology , Uterine Artery/metabolism , Uterus/blood supply , Aorta/cytology , Aorta/metabolism , Biomarkers/metabolism , Blotting, Western , Cells, Cultured , Decidua/metabolism , Decidua/pathology , Female , Gene Expression Profiling , Humans , Immunoenzyme Techniques , Muscle, Smooth, Vascular/pathology , Oligonucleotide Array Sequence Analysis , Pancreatic Elastase/metabolism , Placenta/metabolism , Placenta/pathology , Pregnancy , Pregnancy Trimester, First/metabolism , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Trophoblasts/pathology , Uterine Artery/pathology , Uterus/metabolism , Uterus/pathology
18.
Circ Res ; 100(6): 834-41, 2007 Mar 30.
Article in English | MEDLINE | ID: mdl-17322170

ABSTRACT

Remodeling of the uterine spiral arteries during pregnancy transforms them from high to low resistance vessels that lack vasoconstrictive properties. This process is essential to meet the demand for increased blood flow imposed by the growing fetus. Loss of endothelial and smooth muscle cells (SMC) is evident in remodeled arteries but the mechanisms underlying this transformation remain unknown. This study investigated the hypothesis that fetal trophoblast invading from the placenta instigate remodeling by triggering cell death in vascular SMC. Specifically, a role for trophoblast-derived death inducing cytokine tumor necrosis factor-alpha-related apoptosis-inducing ligand (TRAIL) was investigated. Expression of the activating TRAIL receptors R1 and R2 was detected by flow cytometry on human aortic SMC and by immunohistochemistry on spiral artery SMC. Recombinant human TRAIL induced human aortic SMC apoptosis, which was inhibited by antibodies against TRAIL-R1 or -R2. Perfusion of denuded spiral artery segments with recombinant human TRAIL also induced SMC apoptosis. Trophoblasts isolated from first trimester placenta expressed membrane-associated TRAIL and induced apoptosis of human aortic SMC; apoptosis was significantly inhibited by a recombinant human TRAIL-R1:Fc construct. Trophoblast within the first trimester placental bed also expressed TRAIL. These data show that: 1) TRAIL causes SMC death; 2) trophoblast produce the apoptotic cytokine TRAIL; and 3) trophoblast induce SMC apoptosis via a TRAIL-dependent mechanism. We conclude that TRAIL produced by trophoblast causes apoptosis of SMC and thus may contribute to SMC loss during spiral artery remodeling in pregnancy.


Subject(s)
Apoptosis/physiology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , TNF-Related Apoptosis-Inducing Ligand/physiology , Trophoblasts/metabolism , Apoptosis/drug effects , Arteries/cytology , Cells, Cultured , Decidua/blood supply , Decidua/cytology , Female , Fetus , Humans , Microscopy, Video , Muscle, Smooth, Vascular/cytology , Myometrium/blood supply , Myometrium/cytology , Pregnancy , Pregnancy Trimester, First , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Receptors, Tumor Necrosis Factor/metabolism , TNF-Related Apoptosis-Inducing Ligand/biosynthesis , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Time Factors , Trophoblasts/cytology
19.
Adv Exp Med Biol ; 618: 113-26, 2007.
Article in English | MEDLINE | ID: mdl-18269192

ABSTRACT

In the first trimester of pregnancy fetal trophoblast cells invade the maternal uterine spiral arteries leading to loss of the vascular cells from the vessel wall and remodelling of the extracellular matrix. This is crucial to ensure that sufficient blood can reach the developing fetus. Impaired arterial remodelling is a feature of the major pregnancy pathologies pre-eclampsia and fetal growth restriction. Despite its importance, little is known about the regulation of this process. We have shown, using in vitro culture models and ex vivo explant models, that trophoblast cells play an active role in remodelling spiral arteries, and have implicated apoptotic events in this process. Further we have shown that trophoblast-derived factors such as Fas-ligand, tumor necrosis factor-related apoptosis inducing ligand (TRAIL) are important regulators of this process. The oxygen tension within the uteroplacental environment will vary with gestational age and will depend on the extent of trophoblast invasion and artery remodelling. Fluctuations in oxygen tension may be an important determinant of cellular events both during invasion towards uterine vessels and during the remodelling process. The components of this process known to be regulated by oxygen are reviewed, including lessons that can be learned from pregnancies at high altitude. In addition, data on the effect of varying oxygen tension on trophoblast production of pro-apoptotic factors and susceptibility of vascular smooth muscle cells to induction of apoptosis are described.


Subject(s)
Hypoxia/pathology , Placenta/blood supply , Adaptation, Physiological , Altitude , Blood Vessels/pathology , Female , Humans , Placenta/pathology , Placenta/physiopathology , Pregnancy
20.
Am J Pathol ; 169(5): 1863-74, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17071607

ABSTRACT

During pregnancy, trophoblasts migrate from the placenta into uterine spiral arteries, transforming them into wide channels that lack vasoconstrictive properties. In pathological pregnancies, this process is incomplete. To define the fundamental events involved in spiral artery remodeling, we have studied the effect of trophoblasts on vascular smooth muscle cells (SMCs). Here we demonstrate for the first time that apoptosis of SMCs can be initiated by invading trophoblasts. When trophoblasts isolated from normal placenta (primary trophoblasts) or conditioned medium was perfused into spiral or umbilical artery segments, apoptosis of SMCs resulted. Culture of human aortic SMCs (HASMCs) with primary trophoblasts, primary trophoblast-conditioned medium, or a trophoblast-derived cell line (SGHPL-4) also significantly increased SMC apoptosis. Fas is expressed by spiral artery SMCs, and a Fas-activating antibody triggered HASMC apoptosis. Furthermore, a Fas ligand (FasL)-blocking antibody significantly inhibited HASMC apoptosis induced by primary trophoblasts, SGHPL-4, or trophoblast-conditioned medium. Depleting primary trophoblast-conditioned medium of FasL also abrogated SMC apoptosis in vessels in situ. These results suggest that apoptosis triggered by the release of soluble FasL from invading trophoblasts contributes to the loss of smooth muscle from the walls of spiral arteries during pregnancy.


Subject(s)
Apoptosis , Fas Ligand Protein/metabolism , Muscle, Smooth, Vascular/cytology , Trophoblasts/pathology , Arteries/cytology , Culture Media, Conditioned , Female , Humans , Pregnancy , Time Factors , Trophoblasts/cytology , fas Receptor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...