Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
Endocrinology ; 160(3): 684-698, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30715257

RESUMEN

Placental development is particularly altered in trisomy of chromosome 21 (T21)-affected pregnancies. We previously described in T21-affected placentae an abnormal paracrine crosstalk between the villus mesenchymal core and villus trophoblasts. T21-affected placentae are known to be characterized by their hypovascularity. However, the causes of this anomaly remain not fully elucidated. Therefore, the hypothesis of an abnormal paracrine crosstalk between fetal mesenchymal core and placental endothelial cells (PLECs) was evocated. Villus mesenchymal cells from control (CMCs) and T21 placentae (T21MCs) were isolated and grown in culture to allow their characterization and collection of conditioned media for functional analyses (CMC-CM and T21MC-CM, respectively). Interestingly, PLEC proliferation and branching ability were less stimulated by T21MC-CM than by CMC-CM. Protein array analysis identified secreted proangiogenic growth factors in CMC-CM, which were reduced in T21MC-CM. Combined mass spectrometry and biochemical analysis identified spondin-2 as a factor decreased in T21MC-CM compared with CMC-CM. We found that exogenous spondin-2 stimulated PLEC proliferation and established that T21MC-CM supplemented with spondin-2 recovered conditioned media ability to induce PLEC proliferation and angiogenesis. Hence, this study demonstrates a crosstalk between villus mesenchymal and fetal endothelial cells, in which spondin-2 secreted from mesenchymal cells plays a central role in placental vascular functions. Furthermore, our results also suggest that a reduction in spondin-2 secretion may contribute to the pathogenesis of T21 placental hypovascularity.


Asunto(s)
Síndrome de Down/fisiopatología , Proteínas de la Matriz Extracelular/fisiología , Proteínas de Neoplasias/fisiología , Placenta/irrigación sanguínea , Placentación , Estudios de Casos y Controles , Endostatinas/metabolismo , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Neovascularización Fisiológica , Placenta/fisiopatología , Embarazo
2.
Invest New Drugs ; 37(5): 1075-1085, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30367323

RESUMEN

Introduction The use of paclitaxel in pregnant cancer patients is feasible in terms of fetal safety, but little is known about the effects of paclitaxel on the placenta. Using three experimental models, we aimed to assess the effects of paclitaxel on the expression of placental drug transporters. Methods In the in vitro model (human primary trophoblast culture), trophoblasts were isolated from normal term placentas and subsequently exposed to paclitaxel. The transcriptional regulation of 84 genes encoding for drug transporters, and the protein expression of ABCB1/P-gp and ABCG2/BCRP were assessed. In the in vivo model, placental tissues isolated from pregnant cancer patients treated with paclitaxel were analyzed to assess the protein expression of ABCB1/P-gp and ABCG2/BCRP. The same parameters were assessed in extracts from human placental cotyledons perfused ex vivo with paclitaxel. Results In the in vitro model, the expression of twelve drug-transporters genes was found to be significantly down-regulated after exposure to paclitaxel, including ABCC10, SLC28A3, SLC29A2, and ATP7B (involved in the transport of taxanes, antimetabolites, and cisplatin, respectively). The protein expression of ABCB1/P-gp increased by 1.3-fold after paclitaxel administration. Finally, the protein expression of ABCB1/P-gp and ABCG2/BCRP was higher in cotyledons from mothers treated with multiple doses of paclitaxel during pregnancy than in cotyledons perfused with a single dose of paclitaxel. Discussion Paclitaxel modulates the expression of placental drug transporters involved in the disposition of various anticancer agents. Further studies will be needed to assess the impact of repeated or prolonged exposure to paclitaxel on the expression and function of placental drug transporters.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas de Neoplasias/metabolismo , Neoplasias/tratamiento farmacológico , Paclitaxel/farmacología , Complicaciones Neoplásicas del Embarazo/tratamiento farmacológico , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Adulto , Antineoplásicos Fitogénicos/sangre , Antineoplásicos Fitogénicos/farmacología , Femenino , Humanos , Neoplasias/metabolismo , Paclitaxel/sangre , Embarazo , Complicaciones Neoplásicas del Embarazo/metabolismo , Pronóstico
3.
Sci Rep ; 7(1): 4523, 2017 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-28674422

RESUMEN

Homeobox genes regulate embryonic and placental development, and are widely expressed in the human placenta, but their regulatory control by DNA methylation is unclear. DNA methylation analysis was performed on human placentae from first, second and third trimesters to determine methylation patterns of homeobox gene promoters across gestation. Most homeobox genes were hypo-methylated throughout gestation, suggesting that DNA methylation is not the primary mechanism involved in regulating HOX genes expression in the placenta. Nevertheless, several genes showed variable methylation patterns across gestation, with a general trend towards an increase in methylation over gestation. Three genes (TLX1, HOXA10 and DLX5) showed inverse gains of methylation with decreasing mRNA expression throughout pregnancy, supporting a role for DNA methylation in their regulation. Proteins encoded by these genes were primarily localised to the syncytiotrophoblast layer, and showed decreased expression later in gestation. siRNA mediated downregulation of DLX5, TLX1 and HOXA10 in primary term villous cytotrophoblast resulted in decreased proliferation and increased expression of differentiation markers, including ERVW-1. Our data suggest that loss of DLX5, TLX1 and HOXA10 expression in late gestation is required for proper placental differentiation and function.


Asunto(s)
Diferenciación Celular/genética , Metilación de ADN , Proteínas de Homeodominio/genética , Placenta/metabolismo , Proteínas Proto-Oncogénicas/genética , Factores de Transcripción/genética , Trofoblastos/citología , Trofoblastos/metabolismo , Células Cultivadas , Mapeo Cromosómico , Femenino , Regulación de la Expresión Génica , Genes Homeobox , Edad Gestacional , Proteínas Homeobox A10 , Humanos , Familia de Multigenes , Embarazo
4.
BMC Health Serv Res ; 17(1): 221, 2017 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-28320392

RESUMEN

BACKGROUND: Small for gestational age (SGA) infants are at increased risk for preterm birth morbidities as well as a range of adverse perinatal outcomes that result in part from associated premature birth. We sought to evaluate the costs of SGA versus appropriate for gestational age (AGA) infants in France from pregnancy through the first year of life and separate the contributions of prematurity from the contribution of foetal growth on costs. METHODS: This is a cross-sectional population-based study using national hospital discharge data from French public and private hospitals. SGA infants were defined as newborns with a birth weight below the 10th percentile of French intrauterine growth curves adjusted for foetal sex. AGA infants were defined as newborns with a birth weight between the 25th and the 75th. All births were selected between January 1st, 2011 and December 31st, 2011. Costs were calculated from the hospital perspective for both mothers and children using their diagnostic related group and the French national cost study. Hospital outcomes were extracted from the database and compared by gestational age and mode of delivery. RESULTS: Of 777,720 total births in 2011, 84,688 SGA births (10.9%) and 395,760 AGA births (50.8%) were identified. After adjustment for gestational age, the cost for an SGA infant was €2,783 higher than for an AGA infant. The total maternal and infant hospital cost of SGA in France was estimated at 23% the total cost for deliveries. The high cost is explained by higher complication rates, more frequent hospital readmissions and longer lengths of stay. CONCLUSIONS: Being small for gestational age is an independent contributor to 1-year hospital costs for both mothers and infants.


Asunto(s)
Recien Nacido Prematuro , Recién Nacido Pequeño para la Edad Gestacional , Atención Perinatal/economía , Adulto , Peso al Nacer , Costos y Análisis de Costo , Estudios Transversales , Parto Obstétrico/economía , Femenino , Francia , Edad Gestacional , Humanos , Lactante , Recién Nacido , Enfermedades del Prematuro/economía , Enfermedades del Prematuro/terapia , Tiempo de Internación/economía , Masculino , Morbilidad , Madres/estadística & datos numéricos , Embarazo , Factores Sexuales
5.
FASEB J ; 31(6): 2380-2392, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28223336

RESUMEN

Placental functioning relies on the appropriate differentiation of progenitor villous cytotrophoblasts (CTBs) into extravillous cytotrophoblasts (EVCTs), including invasive EVCTs, and the multinucleated syncytiotrophoblast (ST) layer. This is accompanied by a general move away from a proliferative, immature phenotype. Genome-scale expression studies have provided valuable insight into genes that are associated with the shift to both an invasive EVCT and ST phenotype, whereas genome-scale DNA methylation analysis has shown that differentiation to ST involves widespread methylation shifts, which are counteracted by low oxygen. In the current study, we sought to identify DNA methylation variation that is associated with transition from CTB to ST in vitro and from a noninvasive to invasive EVCT phenotype after culture on Matrigel. Of the several hundred differentially methylated regions that were identified in each comparison, the majority showed a loss of methylation with differentiation. This included a large differentially methylated region (DMR) in the gene body of death domain-associated protein 6 (DAXX ), which lost methylation during both CTB syncytialization to ST and EVCT differentiation to invasive EVCT. Comparison to publicly available methylation array data identified the same DMR as among the most consistently differentially methylated genes in placental samples from preeclampsia pregnancies. Of interest, in vitro culture of CTB or ST in low oxygen increases methylation in the same region, which correlates with delayed differentiation. Analysis of combined epigenomics signatures confirmed DAXX DMR as a likely regulatory element, and direct gene expression analysis identified a positive association between methylation at this site and DAXX expression levels. The widespread dynamic nature of DAXX methylation in association with trophoblast differentiation and placenta-associated pathologies is consistent with an important role for this gene in proper placental development and function.-Novakovic, B., Evain-Brion, D., Murthi, P., Fournier, T., Saffery, R. Variable DAXX gene methylation is a common feature of placental trophoblast differentiation, preeclampsia, and response to hypoxia.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Metilación de ADN/fisiología , Proteínas Nucleares/metabolismo , Oxígeno/administración & dosificación , Preeclampsia/metabolismo , Trofoblastos/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Diferenciación Celular/fisiología , Células Cultivadas , Proteínas Co-Represoras , Epigenómica , Femenino , Regulación de la Expresión Génica/fisiología , Estudio de Asociación del Genoma Completo , Humanos , Chaperonas Moleculares , Proteínas Nucleares/genética , Placenta/citología , Embarazo
6.
Hypertension ; 68(6): 1438-1446, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27698065

RESUMEN

The effects of fluid shear stress (FSS) on the human syncytiotrophoblast and its biological functions have never been studied. During pregnancy, the syncytiotrophoblast is the main source of placental growth factor (PlGF), a proangiogenic factor involved in the placental angiogenesis and the vascular adaptation to pregnancy. The role of FSS in regulating PlGF expression in syncytiotrophoblasts is unknown. We investigated the impact of FSS on the production and secretion of the PlGF by the human syncytiotrophoblasts in primary cell culture. Laminar and continuous FSS (1 dyn cm-2) was applied to human syncytiotrophoblasts cultured in a parallel-plate flow chambers. Secreted levels of PlGF, sFlt-1 (soluble fms-like tyrosin kinase-1), and prostaglandin E2 were tested by immunologic assay. PlGF levels of mRNA and intracellular protein were examined by RT-PCR and Western blot, respectively. Intracellular cAMP levels were examined by time-resolved fluorescence resonance energy transfer cAMP accumulation assay. Production of cAMP and PlGF secretion was significantly increased in FSS conditions compared with static conditions. Western blot analysis of cell extracts exposed to FSS showed an increased phosphorylation of protein kinase A substrates and cAMP response element-binding protein on serine 133. FSS-induced phosphorylation of cAMP response element-binding protein and upregulation of PlGF were prevented by inhibition of protein kinase A with H89 (3 µmol/L). FSS also triggers intracellular calcium flux, which increases the synthesis and release of prostaglandin E2. The enhanced intracellular cAMP in FSS conditions was blocked by COX1/COX2 (cyclooxygenase) inhibitors, suggesting that the increase in prostaglandin E2 production could activate the cAMP/protein kinase A pathway in an autocrine/paracrine fashion. FSS activates the cAMP/protein kinase A pathway leading to upregulation of PlGF in human syncytiotrophoblast.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas de la Membrana/genética , Factor de Crecimiento Placentario/genética , Análisis de Varianza , Células Cultivadas , Femenino , Humanos , Immunoblotting , Inmunohistoquímica , Placenta/metabolismo , Placenta/fisiopatología , Embarazo , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Transducción de Señal/genética , Estrés Mecánico , Trofoblastos/metabolismo , Regulación hacia Arriba
7.
Placenta ; 44: 83-90, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27452442

RESUMEN

During pregnancy, the placenta ensures multiple functions, which are directly involved in the initiation, fetal growth and outcome of gestation. The placental tissue involved in maternal-fetal exchanges and in synthesis of pregnancy hormones is the mononucleated villous cytotrophoblast (VCT) which aggregates and fuses to form and renew the syncytiotrophoblast (ST). Knowledge of the gene expression pattern specific to this endocrine and exchanges tissue of human placenta is of major importance to understand functions of this heterogeneous and complex tissue. Therefore, we undertook a global analysis of the gene expression profiles of primary cultured-VCT (n = 6) and in vitro-differentiated-ST (n = 5) in comparison with whole term placental tissue from which mononucleated VCT were isolated. A total of 880 differentially expressed genes (DEG) were observed between VCT/ST compared to whole placenta, and a total of 37 and 137 genes were significantly up and down-regulated, respectively, in VCT compared to ST. The 37 VCT-genes were involved in cellular processes (assembly, organization, and maintenance), whereas the 137 ST-genes were associated with lipid metabolism and cell morphology. In silico, all networks were linked to 3 transcriptional regulators (PPARγ, RARα and NR2F1) which are known to be essential for trophoblast differentiation. A subset of six DEG was validated by RT-qPCR and four by immunohistochemistry. To conclude, recognition of these pathways is fundamental to increase our understanding of the molecular basis of human trophoblast differentiation. The present study provides for the first time a gene expression signature of the VCT and ST compared to their originated term human placental tissue.


Asunto(s)
Vellosidades Coriónicas/metabolismo , Placenta/metabolismo , Transcriptoma , Trofoblastos/metabolismo , Femenino , Perfilación de la Expresión Génica , Humanos , Embarazo , Nacimiento a Término
8.
Toxicol In Vitro ; 32: 76-85, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26657896

RESUMEN

Our aim was to study the toxicity of benzo(a)pyrene (BaP), an environmental pollutant that can reach placenta, on two human placental models in order to propose biomarkers in risk assessment for pregnancy. Ex vivo human placental cells isolated from term placenta and JEG-3 cancer cell line were incubated with BaP at 0.1-10 µM for 48 h or 72 h. BaP induced neither loss of cell viability nor apoptosis in ex vivo placental cells. To go further, we performed experiments on JEG-3 cell line that provides near-unlimited cells. The results we obtained in JEG-3 cells confirmed that BaP, in our experimental conditions, is neither necrotic nor apoptotic for placental cells. BaP toxicity on placental cells resulted in cell cycle arrest (G2/M phase) associated with inhibition of cell proliferation. Besides, we observed that BaP remodeled the protein content of membrane microdomains via increased expression of ZO-1, caveolin-1 and P2X7 cell degenerescence receptor. In conclusion, we identified nuclear and membrane potential biomarkers of risks for placenta and then pregnancy. These potential biomarkers detected on placental cell lines could represent useful tools for toxicological studies.


Asunto(s)
Benzo(a)pireno/toxicidad , Caveolina 1/metabolismo , Contaminantes Ambientales/toxicidad , Placenta/citología , Receptores Purinérgicos P2X7/metabolismo , Proteína de la Zonula Occludens-1/metabolismo , Apoptosis/efectos de los fármacos , Biomarcadores/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Citocromo P-450 CYP1A1/metabolismo , Femenino , Humanos , Embarazo
9.
PLoS One ; 10(7): e0133506, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26186596

RESUMEN

The chorionic villus of the human placenta is the source of specific endocrine functions and nutrient exchanges. These activities are ensured by the syncytiotrophobast (ST), which bathes in maternal blood. The ST arises and regenerates throughout pregnancy by fusion of underlying cytotrophoblasts (CT). Any anomaly of ST formation or regeneration can affect pregnancy outcome and fetal growth. Because of its direct interaction with maternal blood, the ST is sensitive to drugs, pollutants and xenohormones. Ex vivo assays of perfused cotyledon show that formaldehyde, a common pollutant present in furniture, paint and plastics, can accumulate in the human placenta and cross to the fetal compartment. By means of RT-qPCR, immunoblot and immunocytochemistry experiments, we demonstrate in vitro that formaldehyde exerts endocrine toxicity on human trophoblasts, including a decrease in the production of protein hormones of pregnancy. In addition, formaldehyde exposure triggered human trophoblast fusion by upregulating syncitin-1 receptor expression (ASC-type amino-acid transporter 2: ASCT2). Moreover, we show that formaldehyde-exposed trophoblasts present an altered redox status associated with oxidative stress, and an increase in ASCT2 expression intended to compensate for this stress. Finally, we demonstrate that the adverse effects of formaldehyde on trophoblast differentiation and fusion are reversed by N-acetyl-L-cysteine (Nac), an antioxidant.


Asunto(s)
Diferenciación Celular , Formaldehído/toxicidad , Hormonas Placentarias/metabolismo , Trofoblastos/efectos de los fármacos , Adulto , Sistema de Transporte de Aminoácidos ASC/genética , Sistema de Transporte de Aminoácidos ASC/metabolismo , Células Cultivadas , Femenino , Humanos , Intercambio Materno-Fetal , Antígenos de Histocompatibilidad Menor , Estrés Oxidativo , Circulación Placentaria , Embarazo , Trofoblastos/citología , Trofoblastos/metabolismo
10.
PLoS One ; 10(6): e0127330, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26070137

RESUMEN

In addition to nourishing the embryo, extra-embryonic tissues (EETs) contribute to early embryonic patterning, primitive hematopoiesis, and fetal health. These tissues are of major importance for human medicine, as well as for efforts to improve livestock efficiency, but they remain incompletely understood. In bovines, EETs are accessible easily, in large amounts, and prior to implantation. We took advantage of this system to describe, in vitro and in vivo, the cell types present in bovine EETs at Day 18 of development. Specifically, we characterized the gene expression patterns and phenotypes of bovine extra-embryonic ectoderm (or trophoblast; bTC), endoderm (bXEC), and mesoderm (bXMC) cells in culture and compared them to their respective in vivo micro-dissected cells. After a week of culture, certain characteristics (e.g., gene expression) of the in vitro cells were altered with respect to the in vivo cells, but we were able to identify "cores" of cell-type-specific (and substrate-independent) genes that were shared between in vitro and in vivo samples. In addition, many cellular phenotypes were cell-type-specific with regard to extracellular adhesion. We evaluated the ability of individual bXMCs to migrate and spread on micro-patterns, and observed that they easily adapted to diverse environments, similar to in vivo EE mesoderm cells, which encounter different EE epithelia to form chorion, yolk sac, and allantois. With these tissue interactions, different functions arose that were detected in silico and corroborated in vivo at D21-D25. Moreover, analysis of bXMCs allowed us to identify the EE cell ring surrounding the embryonic disc (ED) at D14-15 as mesoderm cells, which had been hypothesized but not shown prior to this study. We envision these data will serve as a major resource for the future in the analysis of peri-implanting phenotypes in response to the maternal metabolism and contribute to subsequent studies of placental/fetal development in eutherians.


Asunto(s)
Mesodermo/citología , Mesodermo/embriología , Cultivo Primario de Células , Animales , Bovinos , Células Cultivadas , Análisis por Conglomerados , Implantación del Embrión , Desarrollo Embrionario , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Mesodermo/metabolismo , Fenotipo , Transcriptoma
11.
Biochim Biophys Acta ; 1853(9): 2033-44, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25595530

RESUMEN

Annexin-A5 (AnxA5) is the smallest member of the annexins, a group of soluble proteins that bind to membranes containing negatively-charged phospholipids, principally phosphatidylserine, in a Ca(2+)-dependent manner. AnxA5 presents unique properties of binding and self-assembling on membrane surfaces, forming highly ordered two-dimensional (2D) arrays. We showed previously that AnxA5 plays a central role in the machinery of cell membrane repair of murine perivascular cells, promoting the resealing of membrane damages via the formation of 2D protein arrays at membrane disrupted sites and preventing the extension of membrane ruptures. As the placenta is one of the richest source of AnxA5 in humans, we investigated whether AnxA5 was involved in membrane repair in this organ. We addressed this question at the level of human trophoblasts, either mononucleated cytotrophoblasts or multinucleated syncytiotrophoblasts, in choriocarcinoma cells and primary trophoblasts. Using established procedure of laser irradiation and fluorescence microscopy, we observed that both human cytotrophoblasts and syncytiotrophoblasts repair efficiently a µm²-size disruption. Compared to wild-type cells, AnxA5-deficient trophoblasts exhibit severe defect of membrane repair. Through specifically binding to the disrupted site as early as a few seconds after membrane wounding, AnxA5 promotes membrane resealing of injured human trophoblasts. In addition, we observed that a large membrane area containing the disrupted site was released in the extracellular milieu. We propose mechanisms ensuring membrane resealing and subsequent lesion removal in human trophoblasts. This article is part of a Special Issue entitled: 13th European Symposium on Calcium.


Asunto(s)
Anexina A5/metabolismo , Membrana Celular/metabolismo , Trofoblastos/metabolismo , Anexina A5/genética , Línea Celular Tumoral , Membrana Celular/patología , Femenino , Humanos , Embarazo , Trofoblastos/patología
12.
Mol Biosyst ; 10(11): 2974-83, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25167371

RESUMEN

While acting upon chromatin compaction, histone post-translational modifications (PTMs) are involved in modulating gene expression through histone-DNA affinity and protein-protein interactions. These dynamic and environment-sensitive modifications are constitutive of the histone code that reflects the transient transcriptional state of the chromatin. Here we describe a global screening approach for revealing epigenetic disruption at the histone level. This original approach enables fast and reliable relative abundance comparison of histone PTMs and variants in human cells within a single LC-MS experiment. As a proof of concept, we exposed BeWo human choriocarcinoma cells to sodium butyrate (SB), a universal histone deacetylase (HDAC) inhibitor. Histone acid-extracts (n = 45) equally representing 3 distinct classes, Control, 1 mM and 2.5 mM SB, were analysed using ultra-performance liquid chromatography coupled with a hybrid quadrupole time-of-flight mass spectrometer (UPLC-QTOF-MS). Multivariate statistics allowed us to discriminate control from treated samples based on differences in their mass spectral profiles. Several acetylated and methylated forms of core histones emerged as markers of sodium butyrate treatment. Indeed, this untargeted histonomic approach could be a useful exploratory tool in many cases of xenobiotic exposure when histone code disruption is suspected.


Asunto(s)
Ácido Butírico/farmacología , Cromatografía Liquida/métodos , Histonas/análisis , Espectrometría de Masas/métodos , Acetilación , Línea Celular Tumoral , Inhibidores de Histona Desacetilasas/farmacología , Histonas/metabolismo , Humanos , Metilación , Análisis Multivariante , Procesamiento Proteico-Postraduccional
13.
Biomed Res Int ; 2014: 781632, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25093183

RESUMEN

The placenta is a transient organ essential for fetal development. During human placental development, chorionic villi grow in coordination with a large capillary network resulting from both vasculogenesis and angiogenesis. Angiogenin is one of the most potent inducers of neovascularisation in experimental models in vivo. We and others have previously mapped angiogenin expression in the human term placenta. Here, we explored angiogenin involvement in early human placental development. We studied, angiogenin expression by in situ hybridisation and/or by RT-PCR in tissues and primary cultured trophoblastic cells and angiogenin cellular distribution by coimmunolabelling with cell markers: CD31 (PECAM-1), vascular endothelial cadherin (VE-cadherin), vascular endothelial growth factor receptor-2 (VEGF-R2), Tie-2, von Willebrand factor, CD34, erythropoeitin receptor (Epo-R), alpha-smooth muscle actin, CD45, cytokeratin 7, and Ki-67. Extravillous and villous cytotrophoblasts, isolated and differentiated in vitro, expressed and secreted angiogenin. Angiogenin was detected in villous trophoblastic layers, and structured and nascent fetal vessels. In decidua, it was expressed by glandular epithelial cells, vascular cells and macrophages. The observed pattern of angiogenin expression is compatible with a role in blood vessel formation and in cross-talk between trophoblasts and endothelial cells. In view of angiogenin properties, we suggest that angiogenin may participate in placental vasculogenesis and organogenesis.


Asunto(s)
Vasos Sanguíneos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Placentación , Ribonucleasa Pancreática/biosíntesis , Vasos Sanguíneos/crecimiento & desarrollo , Vellosidades Coriónicas/crecimiento & desarrollo , Vellosidades Coriónicas/metabolismo , Células Endoteliales/metabolismo , Femenino , Humanos , Hibridación in Situ , Placenta/metabolismo , Embarazo , Cultivo Primario de Células , Ribonucleasa Pancreática/metabolismo , Trofoblastos/metabolismo
14.
J Cell Sci ; 127(Pt 19): 4172-85, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25052094

RESUMEN

Cell fusion occurs as part of the differentiation of some cell types, including myotubes in muscle and osteoclasts in remodeling bone. In the human placenta, mononuclear cytotrophoblasts in a human chorionic gonadotropin (hCG)-driven process fuse to form multinucleated syncytia that allow the exchange of nutrients and gases between the maternal and fetal circulation. Experiments in which protein kinase A (PKA) is displaced from A-kinase anchoring proteins (AKAPs), or in which specific AKAPs are depleted by siRNA-mediated knockdown, point to ezrin as a scaffold required for hCG-, cAMP- and PKA-mediated regulation of the fusion process. By a variety of immunoprecipitation and immunolocalization experiments, we show that ezrin directs PKA to a molecular complex of connexin 43 (Cx43, also known as GJA1) and zona occludens-1 (ZO-1, also known as TJP1). A combination of knockdown experiments and reconstitution with ezrin or Cx43 with or without the ability to bind to its interaction partner or to PKA demonstrate that ezrin-mediated coordination of the localization of PKA and Cx43 is necessary for discrete control of Cx43 phosphorylation and hCG-stimulated gap junction communication that triggers cell fusion in cytotrophoblasts.


Asunto(s)
Conexina 43/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas del Citoesqueleto/metabolismo , Uniones Comunicantes/metabolismo , Trofoblastos/metabolismo , Comunicación Celular/fisiología , Diferenciación Celular , Fusión Celular , Femenino , Humanos , Proteínas de la Membrana/metabolismo , Embarazo , Transducción de Señal , Trofoblastos/citología
15.
Therapie ; 69(1): 3-11, 2014.
Artículo en Francés | MEDLINE | ID: mdl-24698183

RESUMEN

With more than 830,000 live births in France, a great number of pregnant women are concerned by a treatment during pregnancy and many questions revolve around appreciating medication-related risks during pregnancy. The human placenta is the interface between mother and fetus and remains difficult to study for ethical reasons. Placental transfer of drugs from mother to fetus is dependent on their physicochemical properties, maternal and fetal factors and placental factors. The human placental perfusion model is the only experimental model to study human placental transfer of drugs in organized placental tissue. In vitro models utilizing cell cultures are mostly limited to the investigation of cellular toxicity along pregnancy or specific transfer mechanisms, such as their interaction with transporters. Taking advantage of the complementarity of these models, it will be possible to develop a rational use of drugs during this period.


Asunto(s)
Intercambio Materno-Fetal , Preparaciones Farmacéuticas/metabolismo , Placenta/metabolismo , Técnicas de Cultivo de Célula , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/prevención & control , Femenino , Francia , Humanos , Modelos Biológicos , Embarazo , Complicaciones del Embarazo/tratamiento farmacológico
16.
PLoS One ; 8(11): e79413, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24265769

RESUMEN

Human placental development is characterized by invasion of extravillous cytotrophoblasts (EVCTs) into the uterine wall during the first trimester of pregnancy. Peroxisome proliferator-activated receptor γ (PPARγ) plays a major role in placental development, and activation of PPARγ by its agonists results in inhibition of EVCT invasion in vitro. To identify PPARγ target genes, microarray analysis was performed using GeneChip technology on EVCT primary cultures obtained from first-trimester human placentas. Gene expression was compared in EVCTs treated with the PPARγ agonist rosiglitazone versus control. A total of 139 differentially regulated genes were identified, and changes in the expression of the following 8 genes were confirmed by reverse transcription-quantitative polymerase chain reaction: a disintegrin and metalloproteinase domain12 (ADAM12), connexin 43 (CX43), deleted in liver cancer 1 (DLC1), dipeptidyl peptidase 4 (DPP4), heme oxygenase 1 (HMOX-1), lysyl oxidase (LOX), plasminogen activator inhibitor 1 (PAI-1) and PPARγ. Among the upregulated genes, lysyl oxidase (LOX) was further analyzed. In the LOX family, only LOX, LOXL1 and LOXL2 mRNA expression was significantly upregulated in rosiglitazone-treated EVCTs. RNA and protein expression of the subfamily members LOX, LOXL1 and LOXL2 were analyzed by absolute RT-qPCR and western blotting, and localized by immunohistochemistry and immunofluorescence-confocal microscopy. LOX protein was immunodetected in the EVCT cytoplasm, while LOXL1 was found in the nucleus and nucleolus. No signal was detected for LOXL2 protein. Specific inhibition of LOX activity by ß-aminopropionitrile in cell invasion assays led to an increase in EVCT invasiveness. These results suggest that LOX, LOXL1 and LOXL2 are downstream PPARγ targets and that LOX activity is a negative regulator of trophoblastic cell invasion.


Asunto(s)
Perfilación de la Expresión Génica , PPAR gamma/metabolismo , Placentación , Proteína-Lisina 6-Oxidasa/genética , Proteína-Lisina 6-Oxidasa/metabolismo , Trofoblastos/citología , Aminoácido Oxidorreductasas/genética , Aminoácido Oxidorreductasas/metabolismo , Aminopropionitrilo/farmacología , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Placenta/efectos de los fármacos , Placenta/enzimología , Embarazo , Primer Trimestre del Embarazo/genética , Primer Trimestre del Embarazo/fisiología , Transporte de Proteínas/efectos de los fármacos , Proteína-Lisina 6-Oxidasa/antagonistas & inhibidores , Rosiglitazona , Tiazolidinedionas/farmacología , Trofoblastos/efectos de los fármacos
17.
PLoS One ; 8(11): e78326, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24278109

RESUMEN

Intrauterine growth restriction (IUGR) in preterm infants increases the risk of bronchopulmonary dysplasia, characterized by arrested alveolarization. We evaluated the impact of two different rat models (nitric oxide synthase inhibition or protein deprivation) of IUGR on alveolarization, before, during, and at the end of this postnatal process. We studied IUGR rat pups of dams fed either a low protein (LPD) or a normal diet throughout gestation and pups of dams treated by continuous infusion of Nω-nitro-L-arginine methyl ester (L-NAME) or its diluent on the last four days of gestation. Morphometric parameters, alveolar surface (Svap), mean linear intercept (MLI) and radial alveolar count (RAC) and transcriptomic analysis were determined with special focus on genes involved in alveolarization. IUGR pups regained normal weight at day 21 in the two treated groups. In the LPD group, Svap, MLI and RAC were not different from those of controls at day 4, but were significantly decreased at day 21, indicating alveolarization arrest. In the L-NAME group, Svap and RAC were significantly decreased and MLI was increased at day 4 with complete correction at day 21. In the L-NAME model, several factors involved in alveolarization, VEGF, VEGF-R1 and -R2, MMP14, MMP16, FGFR3 and 4, FGF18 and 7, were significantly decreased at day 4 and/or day 10, while the various factors studied were not modified in the LPD group. These results demonstrate that only maternal protein deprivation leads to sustained impairment of alveolarization in rat pups, whereas L-NAME impairs lung development before alveolarization. Known growth factors involved in lung development do not seem to be involved in LPD-induced alveolarization disorders, raising the question of a possible programming of altered alveolarization.


Asunto(s)
Retardo del Crecimiento Fetal/fisiopatología , Pulmón/patología , Alveolos Pulmonares/fisiopatología , Animales , Animales Recién Nacidos , Arginina/análogos & derivados , Arginina/farmacología , Dieta con Restricción de Proteínas/efectos adversos , Femenino , Retardo del Crecimiento Fetal/etiología , Pulmón/efectos de los fármacos , Masculino , Embarazo , Alveolos Pulmonares/patología , Ratas
18.
Mol Cell Endocrinol ; 377(1-2): 75-83, 2013 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-23831639

RESUMEN

Human idiopathic fetal growth restriction (FGR) is associated with placental insufficiency. Previously, we reported that the expression of homeobox gene Distal-less 3 (DLX3) is increased in idiopathic FGR placentae and is a regulator of villous trophoblast differentiation. Here, we identify the downstream targets of DLX3 in trophoblast-derived cell lines. We modelled the high levels of DLX3 in FGR using an over-expression plasmid construct and complemented this using short-interference RNA (siRNA) for inactivation in cultured cells. Using a real-time PCR-based gene profiling, candidate target genes of DLX3 over-expression and inactivation were identified as regulators of trophoblast differentiation; GATA2 and PPARγ. The expression of GATA2 and PPARγ were further assessed in placental tissues and showed increased mRNA and protein levels in FGR-affected tissues compared with gestation-matched controls. We conclude that DLX3 orchestrates the expression of multiple regulators of trophoblast differentiation and that expression of these regulatory genes is abnormal in FGR.


Asunto(s)
Retardo del Crecimiento Fetal/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Placenta/metabolismo , Transducción de Señal/genética , Factores de Transcripción/metabolismo , Adulto , Línea Celular , Femenino , Factor de Transcripción GATA2/metabolismo , Estudios de Asociación Genética , Proteínas de Homeodominio/genética , Humanos , PPAR gamma/metabolismo , Placenta/patología , Plásmidos/metabolismo , Embarazo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Reproducibilidad de los Resultados , Factores de Transcripción/genética , Trofoblastos/metabolismo , Trofoblastos/patología
19.
Mol Hum Reprod ; 19(10): 665-75, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23761267

RESUMEN

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.


Asunto(s)
Retardo del Crecimiento Fetal/metabolismo , Proteínas de Homeodominio/metabolismo , Proteínas Represoras/metabolismo , Trofoblastos/citología , Trofoblastos/metabolismo , 3-Hidroxiesteroide Deshidrogenasas/genética , 3-Hidroxiesteroide Deshidrogenasas/metabolismo , Adulto , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Gonadotropina Coriónica/genética , Gonadotropina Coriónica/metabolismo , Femenino , Retardo del Crecimiento Fetal/genética , Productos del Gen env/genética , Productos del Gen env/metabolismo , Proteínas de Homeodominio/genética , Humanos , Ratones , Embarazo , Proteínas Gestacionales/genética , Proteínas Gestacionales/metabolismo , Proteínas Represoras/genética
20.
FASEB J ; 27(4): 1309-21, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23233533

RESUMEN

Embryo implantation requires extensive angiogenesis at the maternal-fetal interface. Hyperglycosylated human chorionic gonadotropin (hCG-H), a trophoblast invasive signal produced by extravillous cytotrophoblasts and by choriocarcinoma, was evaluated for its angiogenic role. hCG-H was purified by HPLC from choriocarcinoma supernatant, and the glycosylation pattern was determined by 2D gel analysis. Angiogenesis models used were aortic ring assay with wild-type and LHCGR-knockout mice, endothelial and mural cell proliferation, and migration assays. The TGF-ß signaling pathway was studied by coimmunoprecipitation, competitive binding, TGF-ß reporter gene assays, and Smad immunoblotting. hCG-H displayed a potent angiogenic effect [3.2-fold increase of number of vessel intersections in wild-type aortic rings (11.406 to 36.964)]. hCG-H-induced angiostimulation was independent of the classic hCG signaling pathway since it persisted in LHCGR-knockout mice [4.73-fold increase of number of vessel intersections (10.826 to 51.288)]. Using TGF-ß signaling inhibitors, Tß-RII was identified as the hCG-H receptor responsible for its angiogenic switch. hCG-H exposure enhanced phosphorylation of Smad 2 in endothelial and mural cells and genomic activation of Smad-responsive elements. Interaction between hCG-H and Tß-RII was demonstrated by coimmunoprecipitation and binding competition with (125)I-TGF-ß. This new paracrine interaction between trophoblast and endothelial cells through the hCG-H and the TGF-ß receptor complex plays a key role in angiogenesis associated with placental development and tumorigenesis.


Asunto(s)
Gonadotropina Coriónica/metabolismo , Células Endoteliales/metabolismo , Neovascularización Fisiológica , Placenta/metabolismo , Receptores de HL/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Inductores de la Angiogénesis/metabolismo , Animales , Células Cultivadas , Implantación del Embrión/fisiología , Femenino , Glicosilación , Humanos , Ratones , Ratones Endogámicos C57BL , Embarazo , Ratas , Ratas Wistar , Transducción de Señal/fisiología , Trofoblastos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...