Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Stem Cells Dev ; 32(9-10): 225-236, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36803114

RESUMO

SARS-CoV-2 infection during pregnancy has been associated with poor maternal and neonatal outcomes and placental defects. The placenta, which acts as a physical and immunological barrier at the maternal-fetal interface, is not established until the end of the first trimester. Therefore, localized viral infection of the trophoblast compartment early in gestation could trigger an inflammatory response resulting in altered placental function and consequent suboptimal conditions for fetal growth and development. In this study, we investigated the effect of SARS-CoV-2 infection in early gestation placentae using placenta-derived human trophoblast stem cells (TSCs), a novel in vitro model, and their extravillous trophoblast (EVT) and syncytiotrophoblast (STB) derivatives. SARS-CoV-2 was able to productively replicate in TSC-derived STB and EVT, but not undifferentiated TSCs, which is consistent with the expression of SARS-CoV-2 entry host factors, ACE2 (angiotensin-converting enzyme 2) and TMPRSS2 (transmembrane cellular serine protease) in these cells. In addition, both TSC-derived EVT and STB infected with SARS-CoV-2 elicited an interferon-mediated innate immune response. Combined, these results suggest that placenta-derived TSCs are a robust in vitro model to investigate the effect of SARS-CoV-2 infection in the trophoblast compartment of the early placenta and that SARS-CoV-2 infection in early gestation activates the innate immune response and inflammation pathways. Therefore, placental development could be adversely affected by early SARS-CoV-2 infection by directly infecting the developing differentiated trophoblast compartment, posing a higher risk for poor pregnancy outcomes.


Assuntos
COVID-19 , SARS-CoV-2 , Recém-Nascido , Gravidez , Feminino , Humanos , COVID-19/metabolismo , Trofoblastos/metabolismo , Interferons , Placenta
3.
Cells ; 11(12)2022 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-35741027

RESUMO

Preeclampsia (PE) is a pregnancy-specific disorder that affects 3 to 5% of pregnancies worldwide and is one of the leading causes of maternal and fetal morbidity and mortality. Nevertheless, how these events occur remains unclear. We hypothesized that the induction of hypoxic conditions in vitro in primary human trophoblast cells would mimic several characteristics of PE found in vivo. We applied and characterized a model of primary cytotrophoblasts isolated from healthy pregnancies that were placed under different oxygen concentrations: ambient O2 (5% pCO2, 21%pO2, 24 h, termed "normoxia"), low O2 concentration (5% pCO2, 1.5% pO2, 24 h, termed "hypoxia"), or "hypoxia/reoxygenation" (H/R: 6 h intervals of normoxia and hypoxia for 24 h). Various established preeclamptic markers were assessed in this cell model and compared to placental tissues obtained from PE pregnancies. Seventeen PE markers were analyzed by qPCR, and the protein secretion of soluble fms-like tyrosine kinase 1 (sFlT-1) and the placenta growth factor (PlGF) was determined by ELISA. Thirteen of seventeen genes associated with angiogenesis, the renin-angiotensin system, oxidative stress, endoplasmic reticulum stress, and the inflammasome complex were susceptible to H/R and hypoxia, mimicking the expression pattern of PE tissue. In cell culture supernatants, the secretion of sFlT-1 was increased in hypoxia, while PlGF release was significantly reduced in H/R and hypoxia. In the supernatants of our cell models, the sFlT-1/PlGF ratio in hypoxia and H/R was higher than 38, which is a strong indicator for PE in clinical practice. These results suggest that our cellular models reflect important pathological processes occurring in PE and are therefore suitable as PE in vitro models.


Assuntos
Pré-Eclâmpsia , Biomarcadores/metabolismo , Feminino , Humanos , Hipóxia/metabolismo , Fenótipo , Placenta/metabolismo , Pré-Eclâmpsia/metabolismo , Gravidez , Trofoblastos/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo
4.
Placenta ; 123: 32-40, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35537250

RESUMO

INTRODUCTION: In pregnancy, aldosterone is linked to maternal plasma volume expansion, improved fetal and placental growth/angiogenesis and reduced maternal blood pressure. Aldosterone levels are low in women with pre-eclampsia. Given the placental growth properties of aldosterone in pregnancy, we hypothesised that increased aldosterone improves placental function ex vivo. We applied aldosterone in the dual human placenta perfusion model and analysed specific regulatory markers. METHODS: A single cotyledon was perfused using a trimodal perfusion setup consisting of a control phase (CP; basic perfusion medium (BPM) alone) and two consecutive experimental phases (EP1/EP2; BPM supplemented with 1.5 x 10-9M and 1.5 x 10-7M aldosterone, respectively). CP and EP1/EP2 were conducted in closed circuits lasting 2 h each. Quality/time control perfusions using BPM alone were performed for 360 min to distinguish time-dependent effects from aldosterone-related effects. Perfusates were assessed for control parameters (pH/pO2/pCO2/glucose/lactate/creatinine/antipyrine). Maternal perfusates were analysed for placental growth factor (PlGF), soluble fms-like tyrosine kinase-1 (sFlt-1), interleukin-10 (IL-10) and tumour necrosis factor-alpha (TNF-α) using ELISAs. mRNA expression of abovementioned factors was measured by qPCR in post-perfusion tissue. RESULTS: Data from quality/time control perfusions indicated that TNF-α and IL-10 release continuously increased over time. Contrary, in the trimodal perfusion setup the application of aldosterone decreased TNF-α secretion (P < 0.05, EP1/EP2 vs CP, 120 min) and increased PlGF release (P < 0.05, EP1 vs CP, 90/120 min) into the maternal perfusates. mRNA expression followed similar trends, but did not reach significance. DISCUSSION: Our ex vivo placental perfusion data suggest that increasing aldosterone promotes anti-inflammatory and pro-angiogenic factors, which could positively contribute to healthy pregnancy outcomes.


Assuntos
Placenta , Pré-Eclâmpsia , Aldosterona/metabolismo , Feminino , Humanos , Interleucina-10/metabolismo , Perfusão , Placenta/metabolismo , Fator de Crescimento Placentário , Pré-Eclâmpsia/metabolismo , Gravidez , Resultado da Gravidez , RNA Mensageiro/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo
5.
Stem Cell Reports ; 17(6): 1303-1317, 2022 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-35594858

RESUMO

Trophoblast stem cells (TSCs) have recently been derived from human embryos and early-first-trimester placenta; however, aside from ethical challenges, the unknown disease potential of these cells limits their scientific utility. We have previously established a bone morphogetic protein 4 (BMP4)-based two-step protocol for differentiation of primed human pluripotent stem cells (hPSCs) into functional trophoblasts; however, those trophoblasts could not be maintained in a self-renewing TSC-like state. Here, we use the first step from this protocol, followed by a switch to newly developed TSC medium, to derive bona fide TSCs. We show that these cells resemble placenta- and naive hPSC-derived TSCs, based on their transcriptome as well as their in vitro and in vivo differentiation potential. We conclude that primed hPSCs can be used to generate functional TSCs through a simple protocol, which can be applied to a widely available set of existing hPSCs, including induced pluripotent stem cells, derived from patients with known birth outcomes.


Assuntos
Células-Tronco Pluripotentes Induzidas , Células-Tronco Pluripotentes , Diferenciação Celular , Feminino , Humanos , Placenta , Gravidez , Trofoblastos
6.
Front Cell Dev Biol ; 10: 820286, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35273963

RESUMO

Cytotrophoblasts are progenitor cells that proliferate and fuse to form the multinucleated syncytiotrophoblast layer, implicated in placental endocrine and transport functions. While membrane transporters play a critical role in the distribution of nutrients, hormones, and xenobiotics at the maternal-fetal interface, their selectivity to the syncytiotrophoblast layer is poorly characterized. We aimed to evaluate the regulation of placental transporters in response to trophoblast differentiation in vitro. Experiments were carried out in isolated primary human trophoblast cells before and after syncytialization. Gene expression of six molecular markers and thirty membrane transporters was investigated by qPCR analysis. Subsequently, functional expression was evaluated for proteins involved in the transplacental transfer of essential nutrients i.e., cholesterol (ABCA1, ABCG1), glucose (SLC2A1), leucine (SLC3A2, SLC7A5), and iron (transferrin receptor, TfR1). We identified that human chorionic gonadotropin, placental lactogen, endoglin, and cadherin-11 serve as optimal gene markers for the syncytialization process. We showed that trophoblast differentiation was associated with differential gene expression (mostly up-regulation) of several nutrient and drug transporters. Further, we revealed enhanced protein expression and activity of ABCG1, SLC3A2, SLC7A5, and TfR1 in syncytialized cells, with ABCA1 and GLUT1 displaying no change. Taken together, these results indicate that the syncytiotrophoblast has a dominant role in transporting essential nutrients cholesterol, leucine, and iron. Nonetheless, we present evidence that the cytotrophoblast cells may also be linked to transport functions that could be critical for the cell fusion processes. Our findings collectively yield new insights into the cellular functions associated with or altered by the trophoblast fusion. Importantly, defective syncytialization could lead to nutrient transfer imbalance, ultimately compromising fetal development and programming.

7.
Biol Reprod ; 106(3): 540-550, 2022 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-34791028

RESUMO

The Bone Morphogenetic Protein (BMP) pathway is involved in numerous developmental processes, including cell growth, apoptosis, and differentiation. In mouse embryogenesis, BMP signaling is a well-known morphogen for both mesoderm induction and germ cell development. Recent evidence points to a potential role in development of the extraembryonic compartment, including trophectoderm-derived tissues. In this study, we investigated the effect of BMP signaling in both mouse and human trophoblast stem cells (TSC) in vitro, evaluating the expression and activation of the BMP signaling response machinery, and the effect of BMP signaling manipulation during TSC maintenance and differentiation. Both mouse trophoblast stem cells (mTSC) and human trophoblast stem cells (hTSC) expressed various BMP ligands and the receptors BMPR1A and BMPR2, necessary for BMP response, and displayed maximal active BMP signaling when undifferentiated. We also observed a conserved modulatory role of BMP signaling during trophoblast differentiation, whereby maintenance of active BMP signaling blunted differentiation of TSC in both species. Conversely, the effect of BMP signaling on the undifferentiated state of TSC appeared to be species-specific, with SMAD-independent signaling important in maintenance of mTSC, and a more subtle role for both SMAD-dependent and -independent BMP signaling in hTSC. Altogether, these data establish an autocrine role for the BMP pathway in the trophoblast compartment. As specification and correct differentiation of the extraembryonic compartment are fundamental for implantation and early placental development, insights on the role of the BMP signaling in early development might prove useful in the setting of in vitro fertilization as well as targeting trophoblast-associated placental dysfunction.


Assuntos
Placenta , Trofoblastos , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Diferenciação Celular/fisiologia , Feminino , Humanos , Camundongos , Placenta/metabolismo , Gravidez , Células-Tronco/metabolismo , Trofoblastos/metabolismo
8.
Int J Mol Sci ; 22(19)2021 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-34638773

RESUMO

Intrahepatic cholestasis of pregnancy (ICP) is a pregnancy-related condition characterized by increased maternal circulating bile acids (BAs) having adverse fetal effects. We investigated whether the human placenta expresses specific regulation patterns to prevent fetal exposition to harmful amounts of BAs during ICP. Using real-time quantitative PCR, we screened placentae from healthy pregnancies (n = 12) and corresponding trophoblast cells (n = 3) for the expression of 21 solute carriers and ATP-binding cassette transporter proteins, all acknowledged as BA- and/or cholestasis-related genes. The placental gene expression pattern was compared between healthy women and ICP patients (n = 12 each). Placental SLCO3A1 (OATP3A1) gene expression was significantly altered in ICP compared with controls. The other 20 genes, including SLC10A2 (ASBT) and EPHX1 (EPOX, mEH) reported for the first time in trophoblasts, were comparably abundant in healthy and ICP placentae. ABCG5 was undetectable in all placentae. Placental SLC10A2 (ASBT), SLCO4A1 (OATP4A1), and ABCC2 mRNA levels were positively correlated with BA concentrations in ICP. Placental SLC10A2 (ASBT) mRNA was also correlated with maternal body mass index. We conclude that at the transcriptional level only a limited response of BA transport systems is found under ICP conditions. However, the extent of the transcriptional response may also depend on the severity of the ICP condition and the magnitude by which the maternal BA levels are increased.


Assuntos
Proteínas de Transporte/biossíntese , Colestase Intra-Hepática/metabolismo , Regulação da Expressão Gênica , Glicoproteínas de Membrana/biossíntese , Placenta/metabolismo , Complicações na Gravidez/metabolismo , Colestase Intra-Hepática/patologia , Feminino , Humanos , Proteína 2 Associada à Farmacorresistência Múltipla , Placenta/patologia , Gravidez , Complicações na Gravidez/patologia
9.
Front Cell Dev Biol ; 9: 702046, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34540826

RESUMO

During pregnancy, conceptus-derived extravillous trophoblast (EVT) invades the endomyometrium, anchors the placenta to the maternal uterus, and remodels the spiral arteries in order to establish maternal blood supply to the fetoplacental unit. Recent reports have described early gestation EVT as polyploid and senescent. Here, we extend these reports by performing comprehensive profiling of both the genomic organization and transcriptome of first trimester and term EVT. We define pathways and gene regulatory networks involved in both initial differentiation and maturation of this important trophoblast lineage at the maternal-fetal interface. Our results suggest that like first trimester EVT, term EVT undergoes senescence and endoreduplication, is primarily tetraploid, and lacks high rates of copy number variations. Additionally, we have highlighted senescence and polyploidy-related genes, pathways, networks, and transcription factors that appeared to be important in normal EVT differentiation and maturation and validated a key role for the unfolded protein response in this context.

10.
Int J Mol Sci ; 22(4)2021 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-33567726

RESUMO

Steroid hormones play a crucial role in supporting a successful pregnancy and ensuring proper fetal development. The placenta is one of the principal tissues in steroid production and metabolism, expressing a vast range of steroidogenic enzymes. Nevertheless, a comprehensive characterization of steroidogenic pathways in the human placenta and potential developmental changes occurring during gestation are poorly understood. Furthermore, the specific contribution of trophoblast cells in steroid release is largely unknown. Thus, this study aimed to (i) identify gestational age-dependent changes in the gene expression of key steroidogenic enzymes and (ii) explore the role of trophoblast cells in steroid biosynthesis and metabolism. Quantitative and Droplet Digital PCR analysis of 12 selected enzymes was carried out in the first trimester (n = 13) and term (n = 20) human placentas. Primary trophoblast cells (n = 5) isolated from human term placentas and choriocarcinoma-derived cell lines (BeWo, BeWo b30 clone, and JEG-3) were further screened for gene expression of enzymes involved in placental synthesis/metabolism of steroids. Finally, de novo steroid synthesis by primary human trophoblasts was evaluated, highlighting the functional activity of steroidogenic enzymes in these cells. Collectively, we provide insights into the expression patterns of steroidogenic enzymes as a function of gestational age and delineate the cellular origin of steroidogenesis in the human placenta.


Assuntos
Coriocarcinoma/metabolismo , Regulação da Expressão Gênica , Placenta/metabolismo , Primeiro Trimestre da Gravidez/metabolismo , Esteroide Hidroxilases/metabolismo , Esteroides/metabolismo , Trofoblastos/metabolismo , Adulto , Células Cultivadas , Coriocarcinoma/patologia , Feminino , Idade Gestacional , Humanos , Recém-Nascido , Placenta/citologia , Gravidez , Esteroide Hidroxilases/genética , Trofoblastos/citologia
11.
Placenta ; 105: 50-60, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33548684

RESUMO

INTRODUCTION: The uptake of low- and high-density lipoproteins (LDL and HDL) through the LDL receptor (LDLR) and the scavenger receptor class B type I (SR-BI) mediates maternal to fetal cholesterol transfer in syncytiotrophoblast (STB) cells. STB cells deliver cholesterol via cholesterol efflux through the ATP-binding cassette transporters A1 (ABCA1, to ApoA-I), G1 (ABCG1, to HDL), and SR-BI (to HDL). In the human placenta, these proteins are localized in the apical (LDLR, SR-BI, ABCA1) and basal (SR-BI, ABCA1, ABCG1) membrane of STB cells. However, whether these proteins in polarized primary culture models of STB show a similar localization to those in the human placenta is currently unknown. METHODS: Primary human trophoblasts (PHT) were isolated from normal placentas and cultured in Transwells® with Matrigel to obtain a polarized STB monolayer, proteins were determined by immunofluorescence and cholesterol efflux determined to different acceptors. RESULTS: At day 5, LDLR and ABCA1 localized mainly in the apical membrane, ABCG1 in the basal membrane, and SR-BI in both. Cholesterol efflux towards the apical compartment was higher to adult and neonatal HDL compared to ApoA-I. When acceptors were added in the basal compartment, cholesterol was retained in the Matrigel. DISCUSSION: Polarized STB monolayers express LDLR, SR-BI, ABCA1 and ABCG1, and their apical/basal localization resembles the one described in human placental tissue. This study confirms the high physiological value and suitability of this model for use in functional studies. Our findings also suggest that ABCA1 and SR-BI participate in cholesterol efflux to the maternal side of the cells.


Assuntos
Colesterol/metabolismo , Placenta/metabolismo , Receptores de Lipoproteínas/metabolismo , Trofoblastos/metabolismo , Adulto , Transporte Biológico , Feminino , Humanos , Lipoproteínas/metabolismo , Gravidez
12.
Biochem Soc Trans ; 48(3): 775-786, 2020 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-32369555

RESUMO

Cholesterol is a major nutrient required for fetal growth. It is also a precursor for the synthesis of steroid hormones and essential for the development and maturation of fetal organs. During pregnancy, the placenta controls the transport of cholesterol from the mother to the fetus and vice versa. Cholesterol originating from the maternal circulation has to cross two main membrane barriers to reach the fetal circulation: Firstly, cholesterol is acquired by the apical side of the syncytiotrophoblast (STB) from the maternal circulation as high-density lipoprotein (HDL)-, low-density lipoprotein (LDL)- or very-low-density lipoprotein (VLDL)-cholesterol and secreted at the basal side facing the villous stroma. Secondly, from the villous stroma cholesterol is taken up by the endothelium of the fetal vasculature and transported to the fetal vessels. The proteins involved in the uptake of HDL-, LDL-, VLDL- or unesterified-cholesterol are scavenger receptor type B class 1 (SR-B1), cubulin, megalin, LDL receptor (LDLR) or Niemann-Pick-C1 (NPC1) which are localized at the apical and/or basal side of the STB or at the fetal endothelium. Through interaction with apolipoproteins (e.g. apoA1) cholesterol is effluxed either to the maternal or fetal circulation via the ATP-binding-cassette (ABC)-transporter A1 and ABCG1 localized at the apical/basal side of the STB or the endothelium. In this mini-review, we summarize the transport mechanisms of cholesterol across the human placenta, the expression and localization of proteins involved in the uptake and efflux of cholesterol, and the expression pattern of cholesterol transport proteins in pregnancy pathologies such as pre-eclampsia, gestational diabetes mellitus and intrauterine growth retardation.


Assuntos
Transporte Biológico , Colesterol/metabolismo , Troca Materno-Fetal , Placenta/metabolismo , Aborto Espontâneo , Animais , Apolipoproteínas/metabolismo , Proteínas de Transporte/metabolismo , Membrana Celular/metabolismo , HDL-Colesterol/metabolismo , VLDL-Colesterol/metabolismo , Técnicas de Cocultura , Feminino , Humanos , Gravidez , Trofoblastos/metabolismo
13.
Sci Rep ; 10(1): 5264, 2020 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-32210256

RESUMO

Maternal physiological (MPH) or supraphysiological hypercholesterolaemia (MSPH) occurs during pregnancy. Cholesterol trafficking from maternal to foetal circulation requires the uptake of maternal LDL and HDL by syncytiotrophoblast and cholesterol efflux from this multinucleated tissue to ApoA-I and HDL. We aimed to determine the effects of MSPH on placental cholesterol trafficking. Placental tissue and primary human trophoblast (PHT) were isolated from pregnant women with total cholesterol <280 md/dL (MPH, n = 27) or ≥280 md/dL (MSPH, n = 28). The lipid profile in umbilical cord blood from MPH and MSPH neonates was similar. The abundance of LDL receptor (LDLR) and HDL receptor (SR-BI) was comparable between MSPH and MPH placentas. However, LDLR was localized mainly in the syncytiotrophoblast surface and was associated with reduced placental levels of its ligand ApoB. In PHT from MSPH, the uptake of LDL and HDL was lower compared to MPH, without changes in LDLR and reduced levels of SR-BI. Regarding cholesterol efflux, in MSPH placentas, the abundance of cholesterol transporter ABCA1 was increased, while ABCG1 and SR-BI were reduced. In PHT from MSPH, the cholesterol efflux to ApoA-I was increased and to HDL was reduced, along with reduced levels of ABCG1, compared to MPH. Inhibition of SR-BI did not change cholesterol efflux in PHT. The TC content in PHT was comparable in MPH and MSPH cells. However, free cholesterol was increased in MSPH cells. We conclude that MSPH alters the trafficking and content of cholesterol in placental trophoblasts, which could be associated with changes in the placenta-mediated maternal-to-foetal cholesterol trafficking.


Assuntos
Colesterol/metabolismo , Hipercolesterolemia/sangue , Recém-Nascido/sangue , Complicações na Gravidez/sangue , Trofoblastos/metabolismo , Transportador 1 de Cassete de Ligação de ATP/metabolismo , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Adulto , Apolipoproteína A-I/metabolismo , Transporte Biológico , Células Cultivadas , Feminino , Sangue Fetal/química , Humanos , Lipoproteínas/sangue , Troca Materno-Fetal , Pessoa de Meia-Idade , Placenta/metabolismo , Gravidez , Receptores de LDL/metabolismo , Triglicerídeos/sangue , Adulto Jovem
14.
Sci Rep ; 9(1): 6225, 2019 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-30996342

RESUMO

High levels of atherogenic lipids in pregnancy are associated with health complications for the mother, the fetus and the newborn. As endocrine secretory tissue, the human placenta releases apolipoproteins (apos), particularly apoA1 and apoE. However, the magnitude and the directionality of the apo secretions remain unknown. We aimed to 1) determine the amount and orientation (apical-maternal versus basal-fetal) of placentally secreted apoA1 and apoE using human perfused placenta and primary trophoblast cell (PTC) culture, 2) compare apoA1 and apoE secretions of PTC with that of hepatocytes and 3) associate the obtained results with human blood levels by determining apoA1 and apoE concentrations in maternal and fetal serum samples. In perfused placenta and serum samples, apoA1 and apoE concentrations were significantly higher at the maternal compared to the fetal side. For apoE a similar trend was found in PTC. For apoA1, the secretion to the apical side declined over time while release to the basal side was stable resulting in significantly different apoA1 concentrations between both sides. Unexpectedly, PTC secreted significantly higher amounts of apoA1 and apoE compared to hepatocytes. Our data indicate that the placenta may play an important role in maternal and fetal cholesterol homeostasis via secretion of anti-atherogenic apos.


Assuntos
Apolipoproteína A-I/sangue , Apolipoproteínas E/sangue , Aterosclerose/metabolismo , Colesterol/metabolismo , Feto/metabolismo , Homeostase/fisiologia , Trofoblastos/metabolismo , Adulto , Transporte Biológico/fisiologia , Células Cultivadas , Feminino , Hepatócitos/metabolismo , Humanos , Gravidez
15.
Placenta ; 84: 9-13, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-30773233

RESUMO

Workshops are an important part of the IFPA annual meeting as they allow for discussion of specialized topics. At IFPA meeting 2018 there were nine themed workshops, five of which are summarised in this report. These workshops discussed new perspectives and knowledge in the following areas of research: 1) preeclampsia; 2) abnormally invasive placenta; 3) placental infection; 4) gestational trophoblastic disease; 4) drug delivery to treat placental dysfunction.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Doença Trofoblástica Gestacional , Inflamação , Doenças Placentárias , Pré-Eclâmpsia , Complicações Infecciosas na Gravidez , Animais , Pesquisa Biomédica/organização & administração , Pesquisa Biomédica/tendências , Educação/organização & administração , Educação/normas , Feminino , Doença Trofoblástica Gestacional/tratamento farmacológico , Doença Trofoblástica Gestacional/etiologia , Doença Trofoblástica Gestacional/patologia , Ginecologia/organização & administração , Ginecologia/normas , Ginecologia/tendências , História do Século XXI , Humanos , Inflamação/tratamento farmacológico , Inflamação/etiologia , Inflamação/patologia , Japão , Obstetrícia/organização & administração , Obstetrícia/normas , Obstetrícia/tendências , Placenta/efeitos dos fármacos , Placenta/metabolismo , Doenças Placentárias/tratamento farmacológico , Doenças Placentárias/etiologia , Doenças Placentárias/patologia , Pré-Eclâmpsia/tratamento farmacológico , Pré-Eclâmpsia/etiologia , Pré-Eclâmpsia/patologia , Gravidez , Complicações Infecciosas na Gravidez/tratamento farmacológico , Complicações Infecciosas na Gravidez/etiologia , Complicações Infecciosas na Gravidez/patologia , Sociedades Médicas/organização & administração
16.
Placenta ; 72-73: 48-52, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30501881

RESUMO

Although placental membrane transporters have an important impact on materno-fetal nutrient transfer, placental cell models are poorly characterized regarding transporter expression. We assessed the mRNA expression of 26 physiologically important solute carriers and ABC transporters in BeWo (b30 clone) and primary human trophoblast cells (PHT) before and after syncytialization. 77% of the transporters showed similar mRNA expression changes between BeWo and PHT after syncytialization. Selected transporters, however, were either lacking in BeWo or showed different trends after syncytialization. In conclusion, BeWo cells generally represent an apt model for transporter studies, but their suitability should be confirmed for each transporter by comparison with PHT.


Assuntos
Expressão Gênica , Proteínas de Membrana Transportadoras/genética , Transportadores de Cassetes de Ligação de ATP/genética , Diferenciação Celular , Linhagem Celular , Feminino , Humanos , Modelos Biológicos , Gravidez , RNA Mensageiro/análise , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Carreadoras de Solutos/genética , Trofoblastos/metabolismo
17.
Am J Physiol Lung Cell Mol Physiol ; 315(6): L921-L932, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30211653

RESUMO

Differentiation of primary alveolar type II epithelial cells (AEC II) to AEC type I in culture is a major barrier in the study of the alveolar epithelium in vitro. The establishment of an AEC II cell line derived from induced pluripotent stem cells (iPSC) represents a novel opportunity to study alveolar epithelial cell biology, for instance, in the context of lung injury, fibrosis, and repair. In the present study, we generated long-lasting AEC II from iPSC (LL-iPSC-AEC II). LL-iPSC-AEC II displayed morphological characteristics of AEC II, including growth in a cobblestone monolayer, the presence of lamellar bodies, and microvilli, as shown by electron microscopy. Also, LL-iPSC-AEC II expressed AEC type II proteins, such as cytokeratin, surfactant protein C, and LysoTracker DND 26 (a marker for lamellar bodies). Furthermore, the LL-iPSC-AEC II exhibited functional properties of AEC II by an increase of transepithelial electrical resistance over time, secretion of inflammatory mediators in biologically relevant quantities (IL-6 and IL-8), and efficient in vitro alveolar epithelial wound repair. Consistent with the AEC II phenotype, the cell line showed the ability to uptake and release surfactant protein B, to secrete phospholipids, and to differentiate into AEC type I. In summary, we established a long-lasting, but finite AEC type II cell line derived from iPSC as a novel cellular model to study alveolar epithelial cell biology in lung health and disease.


Assuntos
Células Epiteliais Alveolares/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Diferenciação Celular/fisiologia , Linhagem Celular , Células HEK293 , Humanos , Lesão Pulmonar/patologia , Fenótipo , Alvéolos Pulmonares/citologia , Mucosa Respiratória/citologia
18.
Int J Mol Sci ; 19(8)2018 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-30096856

RESUMO

Cholesterol is indispensable for cellular membrane composition and function. It is also a precursor for the synthesis of steroid hormones, which promote, among others, the maturation of fetal organs. A role of the ATP-binding-cassette-transporter-A1 (ABCA1) in the transport of maternal cholesterol to the fetus was suggested by transferring cholesterol to apolipoprotein-A-1 (apo-A1), but the directionality of the apoA-1/ABCA1-dependent cholesterol transport remains unclear. We isolated primary trophoblasts from term placentae to test the hypotheses that (1) apoA-1/ABCA1 dispatches cholesterol mainly towards the fetus to support fetal developmental maturation at term, and (2) differentiated syncytiotrophoblasts (STB) exert higher cholesterol transport activity than undifferentiated cytotrophoblasts (CTB). As experimental models, we used (1) trophoblast monolayers grown on Transwell® system consisting of apical (maternal-like) and basal (fetal-like) compartments, and (2) trophoblasts grown on conventional culture plates at CTB and STB stages. Surprisingly, apoA-1-mediated cholesterol efflux operated almost exclusively at the apical-maternal side, where ABCA1 was also localized by immunofluorescence. We found greater cholesterol efflux capacity in STB, which was increased by liver-X-receptor agonist treatment and decreased by ABCA1 inhibition. We conclude that at term the apoA-1/ABCA1 pathway is rather involved in cholesterol transport to the mother than in transfer to the fully developed fetus.


Assuntos
Transportador 1 de Cassete de Ligação de ATP/genética , Apolipoproteína A-I/genética , Transporte Biológico/genética , Colesterol/metabolismo , Transportador 1 de Cassete de Ligação de ATP/metabolismo , Apolipoproteína A-I/metabolismo , Membrana Celular/genética , Membrana Celular/metabolismo , Colesterol/genética , Feminino , Feto/metabolismo , Hormônios Esteroides Gonadais/biossíntese , Hormônios Esteroides Gonadais/genética , Humanos , Relações Materno-Fetais , Redes e Vias Metabólicas/genética , Placenta/metabolismo , Gravidez , Trofoblastos/metabolismo
19.
Mol Hum Reprod ; 22(6): 442-56, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26931579

RESUMO

STUDY HYPOTHESIS: Using optimized conditions, primary trophoblast cells isolated from human term placenta can develop a confluent monolayer in vitro, which morphologically and functionally resembles the microvilli structure found in vivo. STUDY FINDING: We report the successful establishment of a confluent human primary trophoblast monolayer using pre-coated polycarbonate inserts, where the integrity and functionality was validated by cell morphology, biophysical features, cellular marker expression and secretion, and asymmetric glucose transport. WHAT IS KNOWN ALREADY: Human trophoblast cells form the initial barrier between maternal and fetal blood to regulate materno-fetal exchange processes. Although the method for isolating pure human cytotrophoblast cells was developed almost 30 years ago, a functional in vitro model with primary trophoblasts forming a confluent monolayer is still lacking. STUDY DESIGN, SAMPLES/MATERIALS, METHODS: Human term cytotrophoblasts were isolated by enzymatic digestion and density gradient separation. The purity of the primary cells was evaluated by flow cytometry using the trophoblast-specific marker cytokeratin 7, and vimentin as an indicator for potentially contaminating cells. We screened different coating matrices for high cell viability to optimize the growth conditions for primary trophoblasts on polycarbonate inserts. During culture, cell confluency and polarity were monitored daily by determining transepithelial electrical resistance (TEER) and permeability properties of florescent dyes. The time course of syncytia-related gene expression and hCG secretion during syncytialization were assessed by quantitative RT-PCR and enzyme-linked immunosorbent assay, respectively. The morphology of cultured trophoblasts after 5 days was determined by light microscopy, scanning electron microscopy (SEM) and transmission electron microscopy (TEM). Membrane makers were visualized using confocal microscopy. Additionally, glucose transport studies were performed on the polarized trophoblasts in the same system. MAIN RESULTS AND THE ROLE OF CHANCE: During 5-day culture, the highly pure trophoblasts were cultured on inserts coated with reconstituted basement membrane matrix . They exhibited a confluent polarized monolayer, with a modest TEER and a size-dependent apparent permeability coefficient (Papp) to fluorescently labeled compounds (MW ∼400-70 000 Da). The syncytialization progress was characterized by gradually increasing mRNA levels of fusogen genes and elevating hCG secretion. SEM analyses confirmed a confluent trophoblast layer with numerous microvilli, and TEM revealed a monolayer with tight junctions. Immunocytochemistry on the confluent trophoblasts showed positivity for the cell-cell adhesion molecule E-cadherin, the tight junction protein 1 (ZO-1) and the membrane proteins ATP-binding cassette transporter A1 (ABCA1) and glucose transporter 1 (GLUT1). Applying this model to study the bidirectional transport of a non-metabolizable glucose derivative indicated a carrier-mediated placental glucose transport mechanism with asymmetric kinetics. LIMITATIONS, REASONS FOR CAUTION: The current study is only focused on primary trophoblast cells isolated from healthy placentas delivered at term. It remains to be evaluated whether this system can be extended to pathological trophoblasts isolated from diverse gestational diseases. WIDER IMPLICATIONS OF THE FINDINGS: These findings confirmed the physiological properties of the newly developed human trophoblast barrier, which can be applied to study the exchange of endobiotics and xenobiotics between the maternal and fetal compartment, as well as intracellular metabolism, paracellular contributions and regulatory mechanisms influencing the vectorial transport of molecules. LARGE-SCALE DATA: Not applicable. STUDY FUNDING AND COMPETING INTERESTS: This study was supported by the Swiss National Center of Competence in Research, NCCR TransCure, University of Bern, Switzerland, and the Swiss National Science Foundation (grant no. 310030_149958, C.A.). All authors declare that their participation in the study did not involve factual or potential conflicts of interests.


Assuntos
Glucose/metabolismo , Placenta/metabolismo , Trofoblastos/metabolismo , Transporte Biológico/genética , Transporte Biológico/fisiologia , Células Cultivadas , Feminino , Humanos , Imuno-Histoquímica , Troca Materno-Fetal/genética , Troca Materno-Fetal/fisiologia , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Placenta/ultraestrutura , Gravidez , Trofoblastos/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...