Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Pediatr Res ; 92(6): 1590-1597, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35184136

RESUMEN

BACKGROUND: Maternal cardiovascular risk factors (CVRF) in pregnancy, i.e., obesity and hyperglycemia, transmit to the fetus and affect placental and fetal endothelial function. Moreover, a sex dimorphism in endothelial function and susceptibility towards CVRF exists already in utero. Endothelial colony-forming cells (ECFC) are circulating endothelial progenitors highly present in neonatal cord blood and sensitive to CVRF. This study investigated whether fetal sex or subtle maternal metabolic changes within healthy range alter fetal ECFC outgrowth. METHODS: Outgrowth of ECFC from cord blood of male (n = 31) and female (n = 26) neonates was analyzed after healthy pregnancies and related to fetal sex and maternal metabolic parameters. RESULTS: Male ECFC grew out earlier (-20.57% days; p = 0.031) than female. Although all women were non-diabetic, higher levels of fasting plasma glucose (FPG) at midpregnancy increased the time required for colony outgrowth (OR: 1.019; p = 0.030), which, after stratifying for fetal sex, was significant only in the males. Gestational weight gain and BMI did not affect outgrowth. Colony number was unchanged by all parameters. CONCLUSIONS: Fetal sex and maternal FPG within normal range alter ECFC function in utero. A role of ECFC in postnatal angiogenesis and vasculogenesis has been suggested, which may be affected by altered outgrowth dynamics. IMPACT: This study is the first to report that a sexual dimorphism exists in ECFC function, as cells of female progeny require a longer period of time until colony outgrowth than ECFC of male progeny. Our data show that ECFC function is highly sensitive and affected by maternal glucose levels even in a normal, non-diabetic range. Our data raise the question of whether maternal plasma glucose in pregnancy should be considered to play a critical role even in the non-diabetic setting.


Asunto(s)
Células Progenitoras Endoteliales , Recién Nacido , Humanos , Femenino , Masculino , Embarazo , Glucosa , Sangre Fetal , Glucemia/metabolismo , Células Cultivadas , Placenta , Ayuno
2.
Int J Mol Sci ; 23(10)2022 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-35628180

RESUMEN

As opposed to adults, high-density lipoprotein (HDL) is the main cholesterol carrying lipoprotein in fetal circulation. The major HDL receptor, scavenger receptor class B type I (SR-BI), contributes to local cholesterol homeostasis. Arterial endothelial cells (ECA) from human placenta are enriched with cholesterol compared to venous endothelial cells (ECV). Moreover, umbilical venous and arterial plasma cholesterol levels differ markedly. We tested the hypothesis that the uptake of HDL-cholesteryl esters differs between ECA and ECV because of the differential expression of SR-BI. We aimed to identify the key regulators underlying these differences and the functional consequences. Immunohistochemistry was used for visualization of SR-BI in situ. ECA and ECV were isolated from the chorionic plate of human placenta and used for RT-qPCR, Western Blot, and HDL uptake assays with 3H- and 125I-labeled HDL. DNA was extracted for the methylation profiling of the SR-BI promoter. SR-BI regulation was studied by exposing ECA and ECV to differential oxygen concentrations or shear stress. Our results show elevated SR-BI expression and protein abundance in ECA compared to ECV in situ and in vitro. Immunohistochemistry demonstrated that SR-BI is mainly expressed on the apical side of placental endothelial cells in situ, allowing interaction with mature HDL circulating in the fetal blood. This was functionally linked to a higher increase of selective cholesterol ester uptake from fetal HDL in ECA than in ECV, and resulted in increased cholesterol availability in ECA. SR-BI expression on ECV tended to decrease with shear stress, which, together with heterogeneous immunostaining, suggests that SR-BI expression is locally regulated in the placental vasculature. In addition, hypomethylation of several CpG sites within the SR-BI promoter region might contribute to differential expression of SR-BI between chorionic arteries and veins. Therefore, SR-BI contributes to a local cholesterol homeostasis in ECA and ECV of the human feto-placental vasculature.


Asunto(s)
Antígenos CD36 , Células Endoteliales , Arterias/metabolismo , Antígenos CD36/genética , Antígenos CD36/metabolismo , Colesterol/metabolismo , Células Endoteliales/metabolismo , Femenino , Homeostasis , Humanos , Lipoproteínas HDL/metabolismo , Placenta/metabolismo , Embarazo , Receptores Inmunológicos/metabolismo , Receptores de Lipoproteína , Receptores Depuradores de Clase B/genética , Receptores Depuradores de Clase B/metabolismo
3.
Int J Mol Sci ; 22(23)2021 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-34884524

RESUMEN

The placenta is an endocrine fetal organ, which secretes a plethora of steroid- and proteo-hormones, metabolic proteins, growth factors, and cytokines in order to adapt maternal physiology to pregnancy. Central to the growth of the fetus is the supply with nutrients, foremost with glucose. Therefore, during pregnancy, maternal insulin resistance arises, which elevates maternal blood glucose levels, and consequently ensures an adequate glucose supply for the developing fetus. At the same time, maternal ß-cell mass and function increase to compensate for the higher insulin demand. These adaptations are also regulated by the endocrine function of the placenta. Excessive insulin resistance or the inability to increase insulin production accordingly disrupts physiological modulation of pregnancy mediated glucose metabolism and may cause maternal gestational diabetes (GDM). A growing body of evidence suggests that this adaptation of maternal glucose metabolism differs between pregnancies carrying a girl vs. pregnancies carrying a boy. Moreover, the risk of developing GDM differs depending on the sex of the fetus. Sex differences in placenta derived hormones and bioactive proteins, which adapt and modulate maternal glucose metabolism, are likely to contribute to this sexual dimorphism. This review provides an overview on the adaptation and maladaptation of maternal glucose metabolism by placenta-derived factors, and highlights sex differences in this regulatory network.


Asunto(s)
Adaptación Fisiológica , Diabetes Gestacional/patología , Sistema Endocrino/fisiopatología , Feto/fisiopatología , Glucosa/metabolismo , Resistencia a la Insulina , Placenta/fisiopatología , Femenino , Humanos , Insulina/metabolismo , Masculino , Embarazo , Factores Sexuales
4.
Clin Sci (Lond) ; 134(1): 39-51, 2020 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-31825070

RESUMEN

Endothelial function and the risk for endothelial dysfunction differ between males and females. Besides the action of estrogen, sex chromosome gene expression and programming effects also provoke this sexual dimorphism. MicroRNAs (miRNAs) have emerged as regulators of endothelial cell function and dysfunction. We here hypothesized distinct miRNA expression patterns in male versus female human endothelial cells that contribute to the functional differences. We used our well-established model of fetal endothelial cells isolated from placenta (fpEC) and analyzed sexual dimorphic miRNA expression and potentially affected biological functions. Next-generation miRNA sequencing of fpEC isolated after pregnancies with male and female neonates identified sex-dependent miRNA expression patterns. Potential biological pathways regulated by the altered set of miRNAs were determined using mirPath and mirSystem softwares, and suggested differences in barrier function and actin organization. The identified pathways were further investigated by monolayer impedance measurements (ECIS) and analysis of F-actin organization (Phalloidin). Nine miRNAs were differentially expressed in fpEC of male versus female neonates. Functional pathways most significantly regulated by these miRNAs included 'Adherens junction', 'ECM receptor interaction' and 'Focal adhesion'. These pathways control monolayer barrier function and may be paralleled by altered cytoskeletal organization. In fact, monolayer impedance was higher in fpEC of male progeny, and F-actin staining revealed more pronounced peripheral stress fibers in male versus female fpEC. Our data highlight that endothelial cell function differs between males and females already in utero, and that altered miRNAs are associated with sex dependent differences in barrier function and actin organization.


Asunto(s)
Actinas/metabolismo , Células Endoteliales/metabolismo , MicroARNs/genética , Caracteres Sexuales , Diabetes Gestacional/genética , Estrógenos/metabolismo , Femenino , Feto/metabolismo , Humanos , Masculino , Placenta/metabolismo , Embarazo
5.
Int J Mol Sci ; 21(3)2020 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-32012940

RESUMEN

Maternal overweight in pregnancy alters the metabolic environment and generates chronic low-grade inflammation. This affects fetal development and programs the offspring's health for developing cardiovascular and metabolic disease later in life. MME (membrane-metalloendopeptidase, neprilysin) cleaves various peptides regulating vascular tone. Endothelial cells express membrane-bound and soluble MME. In adults, the metabolic environment of overweight and obesity upregulates endothelial and circulating MME. We here hypothesized that maternal overweight increases MME in the feto-placental endothelium. We used primary feto-placental endothelial cells (fpEC) isolated from placentas after normal vs. overweight pregnancies and determined MME mRNA, protein, and release. Additionally, soluble cord blood MME was analyzed. The effect of oxygen and tumor necrosis factor α (TNFα) on MME protein in fpEC was investigated in vitro. Maternal overweight reduced MME mRNA (-39.9%, p < 0.05), protein (-42.5%, p = 0.02), and MME release from fpEC (-64.7%, p = 0.02). Both cellular and released MME protein negatively correlated with maternal pre-pregnancy BMI. Similarly, cord blood MME was negatively associated with pre-pregnancy BMI (r = -0.42, p = 0.02). However, hypoxia and TNFα, potential negative regulators of MME expression, did not affect MME protein. Reduction of MME protein in fpEC and in cord blood may alter the balance of vasoactive peptides. Our study highlights the fetal susceptibility to maternal metabolism and inflammatory state.


Asunto(s)
Regulación hacia Abajo , Células Endoteliales/enzimología , Sangre Fetal/enzimología , Regulación del Desarrollo de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Neprilisina/biosíntesis , Obesidad Materna/enzimología , Placenta/enzimología , Adulto , Línea Celular , Células Endoteliales/patología , Femenino , Humanos , Obesidad Materna/patología , Placenta/patología , Embarazo
6.
Histochem Cell Biol ; 152(5): 377-390, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31541300

RESUMEN

Function and dysfunction of endothelial cells are regulated by a multitude of factors. Endothelial cell research often requires in vitro cell culture experiments. Hence, various culture media specifically designed to promote endothelial cell growth are available. These strikingly differ in their composition: complex media contain endothelial cell growth supplement (ECGS), an extract produced of bovine brain with undefined amounts of biologically active compounds, whilst defined media contain selected growth factors in defined concentrations. We here compared the effect of seven purchasable endothelial cell culture media on colony outgrowth, proliferation, viability, in vitro angiogenesis and phenotype of mature primary human endothelial cells using feto-placental endothelial cells isolated from chorionic arteries (fpEC). The effect of media on colony outgrowth was additionally tested on umbilical cord blood-derived endothelial progenitor cells (ECFCs). Outgrowth, purity, proliferation and viability differed between media. Outgrowth of fpEC and ECFCs was best in a defined medium containing EGF, FGF2 and VEGF. By contrast, established fpEC isolations proliferated best in complex media containing ECGS, heparin and ascorbic acid. Also viability of cells was higher in complex media. In vitro angiogenesis was most intense in a defined medium containing the highest number of individual growth factors. FACS analysis of surface markers for endothelial cell subtypes revealed that endothelial phenotype of fpEC was unaffected by media composition. Our data demonstrate the fundamental effect of endothelial cell culture media on primary cell isolation success and behaviour. Whether the composition of supplements is suitable also for individual experiments needs to be tested specifically.


Asunto(s)
Medios de Cultivo Condicionados/farmacología , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Animales , Bovinos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Humanos , Fenotipo
7.
Diabetologia ; 61(11): 2398-2411, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30091044

RESUMEN

AIMS/HYPOTHESIS: An adverse intrauterine environment can result in permanent changes in the physiology of the offspring and predispose to diseases in adulthood. One such exposure, gestational diabetes mellitus (GDM), has been linked to development of metabolic disorders and cardiovascular disease in offspring. Epigenetic variation, including DNA methylation, is recognised as a leading mechanism underpinning fetal programming and we hypothesised that this plays a key role in fetoplacental endothelial dysfunction following exposure to GDM. Thus, we conducted a pilot epigenetic study to analyse concordant DNA methylation and gene expression changes in GDM-exposed fetoplacental endothelial cells. METHODS: Genome-wide methylation analysis of primary fetoplacental arterial endothelial cells (AEC) and venous endothelial cells (VEC) from healthy pregnancies and GDM-complicated pregnancies in parallel with transcriptome analysis identified methylation and expression changes. Most-affected pathways and functions were identified by Ingenuity Pathway Analysis and validated using functional assays. RESULTS: Transcriptome and methylation analyses identified variation in gene expression linked to GDM-associated DNA methylation in 408 genes in AEC and 159 genes in VEC, implying a direct functional link. Pathway analysis found that genes altered by exposure to GDM clustered to functions associated with 'cell morphology' and 'cellular movement' in healthy AEC and VEC. Further functional analysis demonstrated that GDM-exposed cells had altered actin organisation and barrier function. CONCLUSIONS/INTERPRETATION: Our data indicate that exposure to GDM programs atypical morphology and barrier function in fetoplacental endothelial cells by DNA methylation and gene expression change. The effects differ between AEC and VEC, indicating a stringent cell-specific sensitivity to adverse exposures associated with developmental programming in utero. DATA AVAILABILITY: DNA methylation and gene expression datasets generated and analysed during the current study are available at the National Center for Biotechnology Information (NCBI) Gene Expression Omnibus (GEO) database ( http://www.ncbi.nlm.nih.gov/geo ) under accession numbers GSE106099 and GSE103552, respectively.


Asunto(s)
Diabetes Gestacional/metabolismo , Células Endoteliales/metabolismo , Feto/irrigación sanguínea , Placenta/irrigación sanguínea , Metilación de ADN/genética , Diabetes Gestacional/genética , Epigénesis Genética/genética , Femenino , Desarrollo Fetal/genética , Humanos , Embarazo
8.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1863(9): 968-979, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29778664

RESUMEN

Gestational diabetes mellitus (GDM) is associated with excessive oxidative stress which may affect placental vascular function. Cholesterol homeostasis is crucial for maintaining fetoplacental endothelial function. We aimed to investigate whether and how GDM affects cholesterol metabolism in human fetoplacental endothelial cells (HPEC). HPEC were isolated from fetal term placental arterial vessels of GDM or control subjects. Cellular reactive oxygen species (ROS) were detected by H2DCFDA fluorescent dye. Oxysterols were quantified by gas chromatography-mass spectrometry analysis. Genes and proteins involved in cholesterol homeostasis were detected by real-time PCR and immunoblotting, respectively. Cholesterol efflux was determined from [3H]-cholesterol labeled HPEC and [14C]-acetate was used as cholesterol precursor to measure cholesterol biosynthesis and esterification. We detected enhanced formation of ROS and of specific, ROS-derived oxysterols in HPEC isolated from GDM versus control pregnancies. ROS-generated oxysterols were simultaneously elevated in cord blood of GDM neonates. Liver-X receptor activation in control HPEC by synthetic agonist TO901319, 7-ketocholesterol, or 7ß-hydroxycholesterol upregulated ATP-binding cassette transporters (ABC)A1 and ABCG1 expression, accompanied by increased cellular cholesterol efflux. Upregulation of ABCA1 and ABCG1 and increased cholesterol release to apoA-I and HDL3 (78 ±â€¯17%, 40 ±â€¯9%, respectively) were also observed in GDM versus control HPEC. The LXR antagonist GGPP reversed ABCA1 and ABCG1 upregulation and reduced the increased cholesterol efflux in GDM HPEC. Similar total cellular cholesterol levels were detected in control and GDM HPEC, while GDM enhanced cholesterol biosynthesis along with upregulated 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) and sterol O-acyltransferase 1 (SOAT1) mRNA and protein levels. Our results suggest that in GDM cellular cholesterol homeostasis in the fetoplacental endothelium is modulated via LXR activation and helps to maintain its proper functionality.


Asunto(s)
Colesterol/metabolismo , Diabetes Gestacional/metabolismo , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Homeostasis/genética , Receptores X del Hígado/genética , Transportador 1 de Casete de Unión a ATP/genética , Transportador 1 de Casete de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/metabolismo , Adulto , Estudios de Casos y Controles , Colesterol/farmacología , Diabetes Gestacional/genética , Diabetes Gestacional/patología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Femenino , Feto/irrigación sanguínea , Feto/metabolismo , Feto/patología , Regulación de la Expresión Génica , Humanos , Hidroxicolesteroles/metabolismo , Hidroxicolesteroles/farmacología , Hidroximetilglutaril-CoA Reductasas/genética , Hidroximetilglutaril-CoA Reductasas/metabolismo , Cetocolesteroles/metabolismo , Cetocolesteroles/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Receptores X del Hígado/metabolismo , Estrés Oxidativo , Placenta/irrigación sanguínea , Placenta/metabolismo , Placenta/patología , Embarazo , Cultivo Primario de Células , Esterol O-Aciltransferasa/genética , Esterol O-Aciltransferasa/metabolismo
9.
Clin Sci (Lond) ; 132(22): 2437-2449, 2018 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-30389858

RESUMEN

MicroRNAs (miRNAs), small non-coding RNAs, have emerged as important, epigenetic regulators of endothelial function. Metabolic disturbances such as diabetes alter miRNA expression. In adults, the miRNA transcriptome as well as endothelial function differ between the sexes. Here, we hypothesized that metabolic disturbances associated with gestational diabetes (GDM) alter miRNA signatures in feto-placental endothelial cells (fpEC), dependent on fetal sex. We isolated human primary fpEC after normal and GDM-complicated pregnancies with male and female neonates and screened for differential miRNA expression using next-generation miRNA sequencing. To test for miRNAs commonly regulated in fpEC of female and male progeny, data were stratified for fetal sex and maternal body mass index (BMI). Analyses were also performed separately for female and male fpEC, again accounting for maternal BMI as covariate. Potential biological pathways regulated by the altered set of miRNAs were determined using mirPath software. Maternal GDM altered 26 miRNA signatures when male and female fpEC were analyzed together. Separate analysis of male versus female fpEC revealed 22 GDM affected miRNAs in the females and only 4 in the males, without overlap. Biological functions potentially modulated by the affected miRNAs related to 'Protein Processing in Endoplasmic Reticulum' and 'Proteoglycans in Cancer'. Maternal GDM alters miRNA signatures in fpEC, and biological functions affected by these miRNAs relate to well-known adverse functional consequences of diabetes on endothelium. GDM effects were highly dependent on fetal sex with miRNA signatures in female fpEC being more susceptible to metabolic derangements of GDM than miRNAs in male fpEC.


Asunto(s)
Diabetes Gestacional/genética , Células Endoteliales/metabolismo , MicroARNs/genética , Placenta/irrigación sanguínea , Circulación Placentaria , Caracteres Sexuales , Transcriptoma , Adulto , Estudios de Casos y Controles , Células Cultivadas , Diabetes Gestacional/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Masculino , MicroARNs/metabolismo , Embarazo , Factores de Riesgo , Análisis para Determinación del Sexo , Factores Sexuales , Adulto Joven
10.
Lab Invest ; 97(4): 409-418, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28112754

RESUMEN

Feto-placental angiogenesis and vascular development are tightly regulated by pro- and anti-angiogenic factors. Villous trophoblast may be a major source of these factors. It forms the classical placental barrier between mother and fetus, and is thus exposed to maternal influences as well. Metabolic and hormonal derangements in gestational diabetes mellitus (GDM) affect feto-placental angiogenesis and vascular growth. Here we hypothesized that GDM alters the trophoblast secretome, which will modulate the paracrine regulation of feto-placental angiogenesis. Primary term trophoblasts were isolated from normal (n=6) and GDM (n=6) pregnancies. Trophoblast conditioned medium (CM) was used to investigate paracrine effects of normal and GDM-exposed trophoblasts on feto-placental endothelial cells (fpECs; n=7), using functional assays for 2D network formation, wound healing, chemotaxis, and proliferation. Gene expression of 23 pro- and anti-angiogenic factors was analyzed. Four trophoblast-derived paracrine regulators of angiogenesis were specifically measured in CM. CM from GDM trophoblasts increased 2D network formation of fpEC by 2.4-fold (P<0.001), whereas wound healing was attenuated by 1.8-fold (P=0.02) and chemo-attraction to the CM was reduced by 33±9% (P=0.02). The effect of CM on proliferation was unchanged between normal and GDM trophoblasts. Expression analysis of pro- and anti-angiogenic molecules in normal and GDM trophoblasts revealed significant differences in ANGPT2, HGF, KISS1 and PLGF expression. Analysis of secreted proteins demonstrated reduced pigment epithelium derived factor and tumor necrosis factor-α secretion by GDM trophoblasts. GDM alters the balance of trophoblast derived, angiogenesis modulating paracrine factors. This may contribute to GDM-associated changes in placental angiogenesis and vascular structure.


Asunto(s)
Diabetes Gestacional/metabolismo , Neovascularización Fisiológica , Comunicación Paracrina , Trofoblastos/metabolismo , Adulto , Angiopoyetina 2/genética , Angiopoyetina 2/metabolismo , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Medios de Cultivo Condicionados/farmacología , Diabetes Gestacional/genética , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Ensayo de Inmunoadsorción Enzimática , Femenino , Feto/irrigación sanguínea , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Expresión Génica/efectos de los fármacos , Factor de Crecimiento de Hepatocito/genética , Humanos , Kisspeptinas/genética , Placenta/irrigación sanguínea , Factor de Crecimiento Placentario/genética , Embarazo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Necrosis Tumoral alfa/metabolismo
11.
Reproduction ; 154(6): 735-744, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29089453

RESUMEN

We assessed the response of primary cultures of placental villous mononucleated trophoblasts and multinucleated syncytiotrophoblast to calcitriol, the most biologically active form of vitamin D. Whole-genome microarray data showed that calcitriol modulates the expression of many genes in trophoblasts within 6 hours of exposure and RT-qPCR revealed similar responses in cytotrophoblasts, syncytiotrophoblasts and villous explants. Both cytotrophoblasts and syncytiotrophoblasts expressed genes for the vitamin D receptor, for LRP2 and CUBN that mediate internalization of calcidiol, for CYP27B1 that encodes the enzyme that converts calcidiol into active calcitriol, and for CYP24A1 that encodes the enzyme that modifies calcitriol and calcidiol to inactive calcitetrol. Notably, we found an inverse effect of calcitriol on expression of CD14 and CD180/RP105, proteins that differentially regulate toll-like receptor 4-mediated immune responses. Supported by gene ontology analysis, we tested the hypothesis that CD14 and CD180 modulate the inflammatory response of syncytiotrophoblast to bacterial lipopolysaccharide (LPS). These cells showed a robust response to a wide range of LPS concentrations, with induction of active NF-κB and increased secretion of IL-6 and IL-8. SiRNA-mediated knockdown of CD14 reduced the secretion of IL-6 and IL-8 in response to LPS. Collectively, our data showed that calcitriol has a rapid and widespread effect on villous trophoblast gene expression in general, and a specific effect on the innate immune response by syncytiotrophoblast.


Asunto(s)
Antígenos CD/metabolismo , Calcitriol/farmacología , Receptores de Lipopolisacáridos/metabolismo , Lipopolisacáridos/farmacología , Trofoblastos/efectos de los fármacos , 25-Hidroxivitamina D3 1-alfa-Hidroxilasa/genética , 25-Hidroxivitamina D3 1-alfa-Hidroxilasa/metabolismo , Antígenos CD/genética , Antígenos CD/inmunología , Calcitriol/metabolismo , Células Cultivadas , Femenino , Regulación de la Expresión Génica , Humanos , Inmunidad Innata/efectos de los fármacos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Receptores de Lipopolisacáridos/genética , Receptores de Lipopolisacáridos/inmunología , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , FN-kappa B/metabolismo , Embarazo , Cultivo Primario de Células , Interferencia de ARN , Receptores de Calcitriol/genética , Receptores de Calcitriol/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Factores de Tiempo , Transcriptoma , Transfección , Trofoblastos/inmunología , Trofoblastos/metabolismo , Vitamina D3 24-Hidroxilasa/genética , Vitamina D3 24-Hidroxilasa/metabolismo
12.
Angiogenesis ; 19(3): 373-88, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27278471

RESUMEN

The rapidly expanding feto-placental vasculature needs tight control by paracrine and endocrine mechanisms. Here, we focused on paracrine influence by trophoblast, the placental epithelium. We aimed to identify differences in regulation of feto-placental angiogenesis in early versus late pregnancy. To this end, the effect of conditioned media (CM) from early and late pregnancy human trophoblast was tested on network formation, migration and proliferation of human feto-placental endothelial cells. Only CM of late pregnancy trophoblast reduced network formation and migration. Screening of trophoblast transcriptome for anti-angiogenic candidates identified pigment epithelium-derived factor (PEDF) with higher expression and protein secretion in late pregnancy trophoblast. Addition of a PEDF-neutralizing antibody restored the anti-angiogenic effect of CM from late pregnancy trophoblast. Notably, human recombinant PEDF reduced network formation only in combination with VEGF. Also in the CAM assay, the combination of PEDF with VEGF reduced branching of vessels below control levels. Analysis of phosphorylation of ERK1/2 and FAK, two key players in VEGF-induced proliferation and migration, revealed that PEDF altered VEGF signaling, while PEDF alone did not affect phosphorylation of ERK1/2 and FAK. These data suggest that the trophoblast-derived anti-angiogenic molecule PEDF is involved in restricting growth and expansion of the feto-placental endothelium predominantly in late pregnancy and targets to modulate the intracellular effect of VEGF.


Asunto(s)
Proteínas del Ojo/fisiología , Feto/irrigación sanguínea , Feto/fisiología , Neovascularización Fisiológica , Factores de Crecimiento Nervioso/fisiología , Placenta/irrigación sanguínea , Placenta/fisiología , Serpinas/fisiología , Inhibidores de la Angiogénesis/fisiología , Animales , Anticuerpos Neutralizantes/administración & dosificación , Células Cultivadas , Embrión de Pollo , Medios de Cultivo Condicionados , Proteínas del Ojo/antagonistas & inhibidores , Proteínas del Ojo/inmunología , Femenino , Humanos , Factores de Crecimiento Nervioso/antagonistas & inhibidores , Factores de Crecimiento Nervioso/inmunología , Comunicación Paracrina , Embarazo , Primer Trimestre del Embarazo , Tercer Trimestre del Embarazo , Serpinas/inmunología , Trofoblastos/fisiología , Factor A de Crecimiento Endotelial Vascular/fisiología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/fisiología
13.
Reproduction ; 151(6): R91-R102, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26908920

RESUMEN

The phospholipase A2 (PLA2) family is a very diverse group of enzymes, all serving in the cleavage of phospholipids, thereby releasing high amounts of arachidonic acid (AA) and lysophospholipids. AA serves as a substrate for prostaglandin production, which is of special importance in pregnancy for the onset of parturition. Novel research demonstrates that PLA2 action affects the immune response of the mother toward the child and is therefore probably implied in the tolerance of the fetus and prevention of miscarriage. This review presents data on the biochemical and enzymatic properties of PLA2 during gestation with a special emphasis on its role for the placental function and development of the fetus. We also critically discuss the possible pathophysiological significance of PLA2 alterations and its possible functional consequences. These alterations are often associated with pregnancy pathologies such as preeclampsia and villitis or pregnancy complications such as obesity and diabetes in the mother as well as preterm onset of labor.


Asunto(s)
Trabajo de Parto/metabolismo , Fosfolipasas A2/metabolismo , Femenino , Humanos , Embarazo
14.
Am J Obstet Gynecol ; 213(4 Suppl): S182-96, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26428498

RESUMEN

Epigenetic mechanisms, often defined as regulating gene activity independently of underlying DNA sequence, are crucial for healthy development. The sum total of epigenetic marks within a cell or tissue (the epigenome) is sensitive to environmental influence, and disruption of the epigenome in utero has been associated with adverse pregnancy outcomes. Not surprisingly, given its multifaceted functions and important role in regulating pregnancy outcome, the placenta shows unique epigenetic features. Interestingly however, many of these are only otherwise seen in human malignancy (the pseudomalignant placental epigenome). Epigenetic variation in the placenta is now emerging as a candidate mediator of environmental influence on placental functioning and a key regulator of pregnancy outcome. However, replication of findings is generally lacking, most likely due to small sample sizes and a lack of standardization of analytical approaches. Defining DNA methylation "signatures" in the placenta associated with maternal and fetal outcomes offers tremendous potential to improve pregnancy outcomes, but care must be taken in interpretation of findings. Future placental epigenetic research would do well to address the issues present in epigenetic epidemiology more generally, including careful consideration of sample size, potentially confounding factors, issues of tissue heterogeneity, reverse causation, and the role of genetics in modulating epigenetic profile. The importance of animal or in vitro models in establishing a functional role of epigenetic variation identified in human beings, which is key to establishing causation, should not be underestimated.


Asunto(s)
Epigénesis Genética , Placenta/fisiología , Complicaciones del Embarazo/genética , Resultado del Embarazo/genética , Blastocisto , Metilación de ADN , Femenino , Regulación de la Expresión Génica , Interacción Gen-Ambiente , Variación Genética , Impresión Genómica , Humanos , Embarazo , Factores Sexuales
15.
Cells Tissues Organs ; 200(3-4): 181-94, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-26068777

RESUMEN

Proteases are required for a multitude of cellular processes including homeostatic tissue remodelling, invasion and angiogenesis. The physiological function of a cell or tissue is reflected by the set of proteases expressed, also termed degradome. The role of proteases in invasion and angiogenesis has been studied intensively, mostly in cancer. We aimed to compare the set of proteases required for physiological invasion versus physiological angiogenesis from cells deriving from the same organ, and to identify the proteases specific for each process. The human placenta comprises trophoblasts that invade the maternal uterus in a regulated, physiological manner, and it is the source of primary endothelial cells. We isolated the trophoblasts and endothelial cells and verified their invasive phenotype and angiogenic properties, respectively. We then performed gene expression analysis of the degradome, e.g. cysteine, metallo, serine, threonine and aspartic proteases, identified the differentially expressed proteases among the trophoblasts and endothelial cells, and clustered them hierarchically. The results revealed that the set of proteases in trophoblasts versus in endothelial cells overlaps, with a total of 69% in common. Nevertheless, 42% of the studied degradomes differed, with a fold change ≥2. For instance, metalloproteinases were predominantly expressed in trophoblasts, and 31% of the proteases were exclusively expressed in either trophoblasts or endothelial cells; this suggests particular roles for these proteases in either invasion or angiogenesis. Our data identify common and distinct proteases in cells capable of performing invasion and angiogenesis, and may provide basic information for the design of techniques to specifically investigate invasion or angiogenesis.


Asunto(s)
Movimiento Celular , Neovascularización Fisiológica , Proteolisis , Recuento de Células , Separación Celular , Células Endoteliales/citología , Células Endoteliales/enzimología , Femenino , Gelatina/metabolismo , Perfilación de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Péptido Hidrolasas/genética , Péptido Hidrolasas/metabolismo , Fenotipo , Embarazo , Control de Calidad , Reproducibilidad de los Resultados , Trofoblastos/citología , Trofoblastos/enzimología
16.
Epigenetics ; 17(13): 2188-2208, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35950598

RESUMEN

Maternal diabetes and/or obesity in pregnancy are undoubtedly associated with later disease-risk in the offspring. The placenta, interposed between the mother and the foetus, is a potential mediator of this risk through epigenetic mechanisms, including DNA methylation. In recent years, multiple studies have identified differentially methylated CpG sites in the placental tissue DNA in pregnancies complicated by diabetes and obesity. We reviewed all published original research relevant to this topic and analysed our findings with the focus of identifying overlaps, contradictions, and gaps. Most studies focused on the association of gestational diabetes and/or hyperglycaemia in pregnancy and DNA methylation in placental tissue at term. We identified overlaps in results related to specific candidate genes, but also observed a large research gap of pregnancies affected by type 1 diabetes. Other unanswered questions relate to analysis of specific placental cell types and the timing of DNA methylation change in response to diabetes and obesity during pregnancy. Maternal metabolism is altered already in the first trimester involving structural and functional changes in the placenta, but studies into its effects on placental DNA methylation during this period are lacking and urgently needed. Foetal sex is also an important determinant of pregnancy outcome, but only few studies have taken this into account. Collectively, we provide a reference work for researchers working in this large and evolving field. Based on the results of the literature review, we formulate suggestions for future focus of placental DNA methylation studies in pregnancies complicated by diabetes and obesity.


Asunto(s)
Diabetes Gestacional , Embarazo , Femenino , Humanos , Diabetes Gestacional/genética , Diabetes Gestacional/metabolismo , Metilación de ADN , Placenta/metabolismo , Factores Sexuales , Obesidad/complicaciones , Obesidad/genética , Obesidad/metabolismo , Resultado del Embarazo/genética
17.
Placenta ; 97: 68-70, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32792067

RESUMEN

Fetal sex influences placental function as well as maternal and fetal health, being an important factor to consider in pregnancy studies. However, fetal sex determination in the first trimester of pregnancy still faces some technical limitations. Here we describe an RT-qPCR technique to determine fetal sex based on X-inactive specific transcript (XIST) and DEAD-Box helicase 3 Y-linked (DDX3Y) gene expression. This method is straightforward, reliable, fast and applicable on both, placental tissue and primary cells.


Asunto(s)
ARN Helicasas DEAD-box/genética , Expresión Génica , Antígenos de Histocompatibilidad Menor/genética , Placenta/metabolismo , Primer Trimestre del Embarazo/genética , ARN Largo no Codificante/genética , Análisis para Determinación del Sexo/métodos , Femenino , Humanos , Embarazo , Primer Trimestre del Embarazo/metabolismo , Trofoblastos/metabolismo
18.
Cell Adh Migr ; 10(1-2): 18-27, 2016 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-26761204

RESUMEN

Maternal gestational diabetes (GDM) is associated with hyperglycaemia and hyperinsulinemia in the fetal circulation which consequently may induce endothelial dysfunction in the feto-placental vasculature. In fact, feto-placental vasculature reveals various morphological changes in response to GDM. The cell adhesion molecules (CAMs) ICAM-1, VCAM-1 and E-selectin promote attachment and trans-endothelial migration of leukocytes, and are up regulated in inflammation and endothelial dysfunction. Thus, we hypothesized that the GDM environment upregulates ICAM-1, VCAM-1 and E-selectin in the feto-placental endothelium. We isolated primary feto-placental endothelial cells (fpEC) after normal (n=18) and GDM pregnancy (n=11) and analyzed mRNA (RT-qPCR) and protein expression (Immunoblot) of ICAM-1, VCAM-1 and E-selectin. While other CAMs were unchanged on mRNA and protein levels, ICAM-1 protein was decreased by GDM. Further analysis revealed also a decrease in the release of soluble ICAM-1 (sICAM-1), whose levels correlated negatively with maternal BMI. We conclude that this reduction of ICAM-1 protein species is the result of post-translational regulation, since ICAM-1 mRNA expression was unchanged. In fact, miRNAs targeting ICAM-1 were upregulated in GDM fpEC. Immunohistochemistry showed weaker ICAM-1 staining in the placental endothelium after GDM pregnancies, and demonstrated ICAM-1 binding partners CD11a and CD18 expressed on leukocytes in fetal circulation and on placental tissue macrophages. This study identified reduction of ICAM-1 protein in fpEC in GDM pregnancy, which was regulated post-transcriptionally. Low ICAM-1 protein production may represent a protective, placenta-specific mechanism to avoid leukocyte transmigration into the placenta in response to GDM.


Asunto(s)
Diabetes Gestacional/genética , Regulación hacia Abajo/genética , Endotelio Vascular/metabolismo , Feto/metabolismo , Molécula 1 de Adhesión Intercelular/genética , Placenta/metabolismo , Transcripción Genética , Antígenos CD/metabolismo , Biomarcadores/metabolismo , Células Endoteliales/metabolismo , Femenino , Sangre Fetal/metabolismo , Humanos , Inmunohistoquímica , Molécula 1 de Adhesión Intercelular/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Embarazo , Unión Proteica , Subunidades de Proteína/metabolismo , Suero/metabolismo , Solubilidad
19.
Biomed Res Int ; 2014: 145846, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25258707

RESUMEN

The placental vasculature rapidly expands during the course of pregnancy in order to sustain the growing needs of the fetus. Angiogenesis and vascular growth are stimulated and regulated by a variety of growth factors expressed in the placenta or present in the fetal circulation. Like in tumors, hypoxia is a major regulator of angiogenesis because of its ability to stimulate expression of various proangiogenic factors. Chronic fetal hypoxia is often found in pregnancies complicated by maternal diabetes as a result of fetal hyperglycaemia and hyperinsulinemia. Both are associated with altered levels of hormones, growth factors, and proinflammatory cytokines, which may act in a proangiogenic manner and, hence, affect placental angiogenesis and vascular development. Indeed, the placenta in diabetes is characterized by hypervascularisation, demonstrating high placental plasticity in response to diabetic metabolic derangements. This review describes the major regulators of placental angiogenesis and how the diabetic environment in utero alters their expression. In the light of hypervascularized diabetic placenta, the focus was placed on proangiogenic factors.


Asunto(s)
Diabetes Gestacional/metabolismo , Glucosa/metabolismo , Insulina/metabolismo , Oxígeno/metabolismo , Placenta/irrigación sanguínea , Femenino , Humanos , Neovascularización Fisiológica , Embarazo
20.
PLoS One ; 8(10): e79233, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24205377

RESUMEN

Molecular mechanisms underlying sexual dimorphism in mammals, fetal sex influences on intrauterine development, and the sex-biased susceptibility for selected diseases in adulthood are novel areas of current research. As importantly, two decades of multifaceted research has established that susceptibility to many adult disorders originates in utero, commonly secondary to the effects of placental dysfunction. We hypothesized that fetal sex influences gene expression and produces functional differences in human placentas. We thus extended previous studies on sexual dimorphism in mammals, which used RNA isolated from whole tissues, to investigate the effects of sex on four cell-phenotypes within a single key tissue, human placental villi. The cells studied included cytotrophoblasts, syncytiotrophoblast, arterial and venous endothelial cells. The cells were isolated from placentas of male or female fetuses and subjected to microarray analysis. We found that fetal sex differentially affected gene expression in a cell-phenotype dependent manner among all four cell-phenotypes. The markedly enriched pathways in males were identified to be signaling pathways for graft-versus-host disease as well as the immune and inflammatory systems that parallel the reported poorer outcome of male fetuses. Our study is the first to compare global gene expression by microarray analysis in purified, characterized, somatic cells from a single human tissue, i.e. placental villi. Importantly, our findings demonstrate that there are cell-phenotype specific, and tissue-specific, sex-biased responses in the human placenta, suggesting fetal sex should be considered as an independent variable in gene expression analysis of human placental villi.


Asunto(s)
Placenta/citología , Adulto , Vellosidades Coriónicas/metabolismo , Femenino , Desarrollo Fetal , Regulación de la Expresión Génica , Humanos , Masculino , Fenotipo , Placenta/metabolismo , Embarazo , Tercer Trimestre del Embarazo , ARN/metabolismo , Factores Sexuales , Transducción de Señal , Transcriptoma , Trofoblastos/citología , Trofoblastos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA