Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.297
Filtrar
1.
Tissue Cell ; 73: 101662, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34628212

RESUMEN

Bisphenol A (BPA) is an endocrine disruptor extensively used in the production of polycarbonate plastics and epoxy resins and a component of liquid and food containers. It is a hazard in the prenatal period because of its presence in the placenta, fetal membranes, amniotic fluid, maternal and fetal blood and its ability to cross the placenta and reach the fetus. Estimation of the risk of BPA exposure during in utero life is extremely important in order to prevent complications of pregnancy and fetal growth. This review describes in vitro models of the human materno-fetal interface. It also outlines the effects of BPA at doses indicated as "physiological", namely at the concentrations found in the general population, and at "supraphysiological" and "subphysiological" doses, i.e. above and below the physiological range. This work will help clarify the discrepancies observed in studies on the effects of BPA on human reproduction and pregnancy, and it will be useful for the choice of appropriate in vitro models for future studies aimed at identifying the potential impact of BPA on specific functional processes.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Intercambio Materno-Fetal/efectos de los fármacos , Especificidad de Órganos , Fenoles/toxicidad , Femenino , Humanos , Modelos Biológicos , Especificidad de Órganos/efectos de los fármacos , Embarazo , Factores de Riesgo
2.
Endocrinology ; 162(12)2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34647996

RESUMEN

Iron deficiency, which occurs when iron demands chronically exceed intake, is prevalent in pregnant women. Iron deficiency during pregnancy poses major risks for the baby, including fetal growth restriction and long-term health complications. The placenta serves as the interface between a pregnant mother and her baby, and it ensures adequate nutrient provisions for the fetus. Thus, maternal iron deficiency may impact fetal growth and development by altering placental function. We used a rat model of diet-induced iron deficiency to investigate changes in placental growth and development. Pregnant Sprague-Dawley rats were fed either a low-iron or iron-replete diet starting 2 weeks before mating. Compared with controls, both maternal and fetal hemoglobin were reduced in dams fed low-iron diets. Iron deficiency decreased fetal liver and body weight, but not brain, heart, or kidney weight. Placental weight was increased in iron deficiency, due primarily to expansion of the placental junctional zone. The stimulatory effect of iron deficiency on junctional zone development was recapitulated in vitro, as exposure of rat trophoblast stem cells to the iron chelator deferoxamine increased differentiation toward junctional zone trophoblast subtypes. Gene expression analysis revealed 464 transcripts changed at least 1.5-fold (P < 0.05) in placentas from iron-deficient dams, including altered expression of genes associated with oxygen transport and lipoprotein metabolism. Expression of genes associated with iron homeostasis was unchanged despite differences in levels of their encoded proteins. Our findings reveal robust changes in placentation during maternal iron deficiency, which could contribute to the increased risk of fetal distress in these pregnancies.


Asunto(s)
Deficiencias de Hierro/fisiopatología , Placentación/fisiología , Complicaciones del Embarazo/fisiopatología , Trofoblastos/fisiología , Animales , Diferenciación Celular/efectos de los fármacos , Dieta , Suplementos Dietéticos , Femenino , Hierro/farmacología , Hierro/uso terapéutico , Deficiencias de Hierro/complicaciones , Deficiencias de Hierro/dietoterapia , Intercambio Materno-Fetal/efectos de los fármacos , Placentación/efectos de los fármacos , Embarazo , Complicaciones del Embarazo/dietoterapia , Ratas , Ratas Sprague-Dawley , Trofoblastos/efectos de los fármacos
3.
Artículo en Inglés | MEDLINE | ID: mdl-34580181

RESUMEN

BACKGROUND AND OBJECTIVES: To determine in a mouse model whether neonatal Fc receptor (FcRn) blockade prevents the placental transfer of class G immunoglobulin (IgG) derived from patients with anti-NMDA receptor (NMDAR) encephalitis and their pathogenic effects on the fetuses and offspring. METHODS: Pregnant C57BL/6J mice were administered via tail vein FcRn antibody (FcRn-ab) or saline solution 6 hours before administration of patients' or controls' IgG on days 14, 15, and 16 of gestation. Three experimental groups were established: mice receiving controls' IgG, patients' IgG, or patients' IgG along with pretreatment with FcRn-ab. Immunohistochemical staining, confocal microscopy, hippocampal long-term potentiation, and standardized developmental and behavioral tasks were used to assess the efficacy of treatment with FcRn-ab. RESULTS: In pregnant mice that received patients' IgG, treatment with FcRn-ab prevented the IgG from reaching the fetal brain, abrogating the decrease of NMDAR clusters and the reduction of cortical plate thickness that were observed in fetuses from untreated pregnant mice. Moreover, among the offspring of mothers that received patients' IgG, those whose mothers were treated with FcRn-ab did not develop the alterations that occurred in offspring of untreated mothers, including impairment in hippocampal plasticity, delay in innate reflexes, and visuospatial memory deficits. DISCUSSION: FcRn blockade prevents placental transfer of IgG from patients with anti-NMDAR encephalitis and abrogates the synaptic and neurodevelopmental alterations caused by patients' antibodies. This model has potential therapeutic implications for other antibody-mediated diseases of the CNS during pregnancy.


Asunto(s)
Encefalitis Antirreceptor N-Metil-D-Aspartato/inmunología , Anticuerpos Bloqueadores/administración & dosificación , Autoanticuerpos/administración & dosificación , Antígenos de Histocompatibilidad Clase I/inmunología , Inmunoglobulina G/administración & dosificación , Intercambio Materno-Fetal/efectos de los fármacos , Circulación Placentaria/efectos de los fármacos , Receptores Fc/inmunología , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Embarazo
4.
Placenta ; 115: 70-77, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34562829

RESUMEN

Species differences are among the main reasons for the high failure rate of preclinical studies. A better awareness and understanding of these differences might help to improve the outcome of preclinical research. In reproduction, the placenta is the central organ regulating fetal exposure to a substance circulating in the maternal organism. Exact information about placental transfer can help to better estimate the toxic potential of a substance. From an evolutionary point of view, the chorioallantoic placenta is the organ with the highest anatomical diversity among species. Moreover, frequently used animal models in reproduction belong to rodents and lagomorphs, two groups that are characterized by the generation of an additional type of placenta, which is crucial for fetal development, but absent from humans: the inverted yolk sac placenta. Taken together, the translatability of placental transfer studies from laboratory animals to humans is challenging, which is supported by the fact that numerous species-dependent toxic effects are described in literature. Thus, reliable human-relevant data are frequently lacking and the toxic potential of chemicals and pharmaceuticals for humans can hardly be estimated, often resulting in recommendations that medical treatments or exposure to chemicals should be avoided for safety reasons. Although species differences of placental anatomy have been described frequently and the need for human-relevant research models has been emphasized, analyses of substances with species-dependent placental transfer have been performed only sporadically. Here, we present examples for species-specific placental transfer, including that of nanoparticles and pharmaceuticals, and discuss potential underlying mechanisms. With respect to the COVID 19-pandemic it might be of interest that some antiviral drugs are reported to feature species-specific placental transfer. Further, differences in placental structure and antibody transfer may affect placental transfer of ZIKA virus.


Asunto(s)
Intercambio Materno-Fetal/fisiología , Placenta/metabolismo , Animales , Antivirales/farmacocinética , Transporte Biológico/fisiología , COVID-19/transmisión , COVID-19/virología , Femenino , Humanos , Transmisión Vertical de Enfermedad Infecciosa , Intercambio Materno-Fetal/efectos de los fármacos , Placenta/efectos de los fármacos , Embarazo , Complicaciones Infecciosas del Embarazo/tratamiento farmacológico , Complicaciones Infecciosas del Embarazo/metabolismo , Complicaciones Infecciosas del Embarazo/virología , SARS-CoV-2/metabolismo , Especificidad de la Especie , Saco Vitelino/metabolismo , Saco Vitelino/fisiología , Virus Zika/metabolismo , Infección por el Virus Zika/tratamiento farmacológico , Infección por el Virus Zika/transmisión , Tratamiento Farmacológico de COVID-19
5.
Front Immunol ; 12: 649135, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33841433

RESUMEN

The perfect synchronization of maternal immune-endocrine mechanisms and those of the fetus is necessary for a successful pregnancy. In this report, decidual immune cells at the maternal-fetal interface were detected that expressed TIGIT (T cell immunoreceptor with Ig and ITIM domains), which is a co-inhibitory receptor that triggers immunological tolerance. We generated recombinant TIGIT-Fc fusion proteins by linking the extracellular domain of TIGIT and silent Fc fragments. The treatment with TIGIT-Fc of human decidual antigen presenting cells (APCs), the decidual dendritic cells (dDCs), and decidual macrophages (dMϕs) increased the production of interleukin 10 and induced the decidua APCs to powerfully polarize the decidual CD4+ T cells toward a classic TH2 phenotype. We further proposed that Notch signaling shows a pivotal effect on the transcriptional regulation in decidual immune cell subsets. Moreover, the administration of TIGIT-Fc to CBA/J pregnant mice at preimplantation induced CD4+ forkhead box P3+ (Foxp3+) regulatory T cells and tolerogenic dendritic cells and increased pregnancy rates in an abortion-prone animal model stress. The results suggested the therapeutic potential of the TIGIT-Fc fusion protein in reinstating immune tolerance in failing pregnancies.


Asunto(s)
Decidua/inmunología , Tolerancia Inmunológica/inmunología , Fragmentos Fc de Inmunoglobulinas/inmunología , Intercambio Materno-Fetal/inmunología , Receptores Inmunológicos/inmunología , Animales , Células Presentadoras de Antígenos/efectos de los fármacos , Células Presentadoras de Antígenos/inmunología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Línea Celular Tumoral , Células Cultivadas , Decidua/citología , Decidua/efectos de los fármacos , Decidua/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Femenino , Humanos , Tolerancia Inmunológica/efectos de los fármacos , Fragmentos Fc de Inmunoglobulinas/química , Fragmentos Fc de Inmunoglobulinas/uso terapéutico , Interleucina-10/inmunología , Interleucina-10/metabolismo , Activación de Linfocitos/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Intercambio Materno-Fetal/efectos de los fármacos , Ratones Endogámicos CBA , Ratones Endogámicos DBA , Embarazo , Receptores Inmunológicos/química , Receptores Inmunológicos/uso terapéutico
6.
Pharm Res ; 38(4): 647-655, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33825113

RESUMEN

PURPOSE: Fexofenadine is a well-identified in vivo probe substrate of P-glycoprotein (P-gp) and/or organic anion transporting polypeptide (OATP). This work aimed to investigate the transplacental pharmacokinetics of fexofenadine enantiomers with and without the selective P-gp inhibitor fluoxetine. METHODS: The chiral transplacental pharmacokinetics of fexofenadine-fluoxetine interaction was determined using the ex vivo human placenta perfusion model (n = 4). In the Control period, racemic fexofenadine (75 ng of each enantiomer/ml) was added in the maternal circuit. In the Interaction period, racemic fluoxetine (50 ng of each enantiomer/mL) and racemic fexofenadine (75 ng of each enantiomer/mL) were added to the maternal circulation. In both periods, maternal and fetal perfusate samples were taken over 90 min. RESULTS: The (S)-(-)- and (R)-(+)-fexofenadine fetal-to-maternal ratio values in Control and Interaction periods were similar (~0.18). The placental transfer rates were similar between (S)-(-)- and (R)-(+)-fexofenadine in both Control (0.0024 vs 0.0019 min-1) and Interaction (0.0019 vs 0.0021 min-1) periods. In both Control and Interaction periods, the enantiomeric fexofenadine ratios [R-(+)/S-(-)] were approximately 1. CONCLUSIONS: Our study showed a low extent, slow rate of non-enantioselective placental transfer of fexofenadine enantiomers, indicating a limited fetal fexofenadine exposure mediated by placental P-gp and/or OATP2B1. The fluoxetine interaction did not affect the non-enantioselective transplacental transfer of fexofenadine. The ex vivo placental perfusion model accurately predicts in vivo placental transfer of fexofenadine enantiomers with remarkably similar values (~0.17), and thus estimates the limited fetal exposure.


Asunto(s)
Antagonistas de los Receptores Histamínicos H1 no Sedantes/farmacocinética , Intercambio Materno-Fetal/efectos de los fármacos , Placenta/metabolismo , Terfenadina/análogos & derivados , Subfamilia B de Transportador de Casetes de Unión a ATP/antagonistas & inhibidores , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Área Bajo la Curva , Interacciones Farmacológicas , Femenino , Fluoxetina/administración & dosificación , Fluoxetina/farmacocinética , Antagonistas de los Receptores Histamínicos H1 no Sedantes/administración & dosificación , Humanos , Perfusión/instrumentación , Perfusión/métodos , Embarazo , Complicaciones del Embarazo/tratamiento farmacológico , Complicaciones del Embarazo/inmunología , Estereoisomerismo , Terfenadina/administración & dosificación , Terfenadina/farmacocinética
7.
Drug Metab Dispos ; 49(4): 337-343, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33531413

RESUMEN

Cannabidiol (CBD), a major component of cannabis, has various effects, such as antiemetic and anxiolytic activities, and has recently been marketed as a supplement. The number of people using CBD during pregnancy is increasing, and there are concerns about its effects on the fetus. In addition, the scientific evidence supporting the fetal safety of CBD use during pregnancy is insufficient. To investigate CBD transfer from the mother to the fetus, a single intravenous dose of CBD was administered to pregnant mice in this study, and fetal pharmacokinetics (distribution and elimination) was analyzed. The transfer of CBD from the maternal blood to the fetus was rapid, and the compound accumulated in the fetal brain, liver, and gastrointestinal tract. Conversely, little CBD was transferred from the mother to the amniotic fluid. We analyzed the pharmacokinetics of CBD using a two-compartment model and found that the maternal and fetal half-lives of CBD were approximately 5 and 2 hours, respectively. Furthermore, we performed a moment analysis of the pharmacokinetics of CBD, observing a mean residence time of less than 2 hours in both the mother and fetus. These results suggest that once-daily CBD intake during pregnancy is unlikely to result in CBD accumulation in the mother or fetus. SIGNIFICANCE STATEMENT: CBD is currently marketed as a supplement, and despite its increasing use during pregnancy, little information concerning its fetal effects has been reported. In the present study, CBD was administered to pregnant mice, and the pharmacokinetics in the fetus was investigated using a two-compartment model and moment analysis. The results of these analyses provide important information for estimating the risk to the fetus if CBD is mistakenly consumed during pregnancy.


Asunto(s)
Cannabidiol/farmacocinética , Feto/efectos de los fármacos , Feto/metabolismo , Intercambio Materno-Fetal/efectos de los fármacos , Embarazo/sangre , Embarazo/efectos de los fármacos , Animales , Anticonvulsivantes/farmacocinética , Femenino , Intercambio Materno-Fetal/fisiología , Ratones , Ratones Endogámicos ICR
8.
J Appl Toxicol ; 41(4): 572-586, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32969501

RESUMEN

Persistent organic pollutants such as organophosphate flame retardants (OPFRs) can accumulate in the body and interact with nuclear receptors that control energy homeostasis. One sensitive window of exposure is during development, either in utero or neonatal. Therefore, we investigated if maternal exposure to a mixture of OPFRs alters metabolism on a low-fat diet (LFD) or a high-fat diet (HFD) in both male and female offspring. Wild-type C57Bl/6J dams were orally dosed with vehicle (sesame oil) or an OPFR mixture (1 mg/kg each of tris(1,3-dichloro-2-propyl)phosphate, triphenyl phosphate, and tricresyl phosphate) from gestation day 7 to postnatal day 14. After weaning, pups were fed LFD or HFD. To assess metabolism, we measured body weight and food intake weekly and determined body composition, metabolism, activity, and glucose homeostasis at 6 months of age. Although maternal OPFR exposure did not alter body weight or adiposity, OPFR exposure altered substrate utilization and energy expenditure depending on diet in both sexes. Systolic and diastolic blood pressure was increased by OPFR in male offspring. OPFR exposure interacted with HFD to increase fasting glucose in females and alter glucose and insulin tolerance in male offspring. Plasma leptin was reduced in male and female offspring when fed HFD, whereas liver expression of Pepck was increased in females and Esr1 (estrogen receptor α) was increased in both sex. The physiological implications indicate maternal exposure to OPFRs programs peripheral organs including the liver and adipose tissue, in a sex-dependent manner, thus changing the response to an obesogenic diet and altering adult offspring energy homeostasis.


Asunto(s)
Metabolismo Energético/efectos de los fármacos , Retardadores de Llama/toxicidad , Homeostasis/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Exposición Materna/efectos adversos , Intercambio Materno-Fetal/efectos de los fármacos , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Metabolismo Energético/genética , Femenino , Homeostasis/genética , Humanos , Metabolismo de los Lípidos/genética , Intercambio Materno-Fetal/genética , Ratones , Ratones Endogámicos C57BL , Embarazo
9.
Ecotoxicol Environ Saf ; 207: 111281, 2021 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-32919195

RESUMEN

Epidemiological studies of human and animal experiments indicated that gestational exposure to atmospheric pollutants could be followed by the abnormal placental development. However, the effects of this exposure on the placental transportation for nutrients have not been systematically investigated. In this study, fine particulate matters (PM2.5) samples were collected in Taiyuan and pregnant rodent models were administered with 3 mg/kg b.w. PM2.5 by oropharyngeal aspiration every other day starting on embryonic day 0.5 (E0.5). Then the pregnant mice were sacrificed and their placentas were collected at different time points. The results showed that maternal PM2.5 exposure (MPE) disrupted the expression of proliferating cell nuclear antigen (PCNA) at all time points and inhibited the cell proliferation in placenta. Following that, the capacity for placental nutrient transport was impaired. The changes at E18.5 were observed most significantly, showing the altered mRNA expression of amino acid, long-chain polyunsaturated fatty acid (LCPUFA), glucose and folate transporters. In addition, the glycogen content was elevated at E18.5, and the triglyceride content was increased at E13.5 and E15.5 and decreased at E18.5 in the placenta after MPE. In a word, the adverse effect induced by MPE revealed that MPE led tothe disruption on the nutrient supply to the developing fetus via modulating the abundance of placental nutrient transporters (PNT).


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Exposición Materna/efectos adversos , Nutrientes/metabolismo , Material Particulado/toxicidad , Placenta/efectos de los fármacos , Contaminantes Atmosféricos/metabolismo , Aminoácidos/metabolismo , Animales , Transporte Biológico , Proliferación Celular/efectos de los fármacos , Ácidos Grasos/metabolismo , Femenino , Glucosa/metabolismo , Glucógeno/metabolismo , Humanos , Intercambio Materno-Fetal/efectos de los fármacos , Ratones , Material Particulado/metabolismo , Placenta/metabolismo , Placenta/patología , Embarazo
10.
J Endocrinol Invest ; 44(3): 443-452, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32514901

RESUMEN

PURPOSE: The use and contribution of prenatal multivitamins (PMV) as iodine source for pregnant women in China, especially in mildly iodine-deficient region, have not been well studied. This study aimed to explore the association between PMV intake during pregnancy and thyroid function in mothers and newborns. METHODS: We performed a study involving women with a history of taking PMV during pregnancy between January 2013 and October 2015, in Shanghai, a mildly iodine-deficient region. Maternal thyroid function in early and late pregnancy, and neonatal TSH on postnatal d 3 were obtained from medical records. We compared the outcomes in pregnant women who took exclusively iodine-containing PMV (I + PMV) with those who took exclusively non-contained PMV (I- PMV). Propensity score matching (PSM) was used to identify women with similar baseline characteristics. RESULTS: After PSM, 1280 women in I + PMV and 2560 in I- PMV had similar propensity scores and were included in the analyses. Introduction of I + PMV to women was associated with slightly higher maternal thyroid hormone production (higher maternal FT4, p = 0.01, non-significantly lower TSH, p = 0.79) and lower neonatal TSH levels (p < 0.0001). The frequency of adverse pregnancy outcomes or thyroid dysfunctions did not differ between groups in late pregnancy. Mothers received I + PMV (0.2 SD) had a stronger association of maternal TSH with neonatal TSH than those who received I- PMV (0.1 SD). These effects were only shown in TPOAb-negative mothers, not in TPOAb-positive mothers. CONCLUSION: TPOAb-positive women display an impaired iodine transport in thyroid and placenta, and this may explain the lack of changes in maternal and neonatal thyroid parameters with I + PMV supplementation in these women. This phenomenon might suggest that these women require different iodine doses or treatment approach in comparison with TPOAb-negative women.


Asunto(s)
Suplementos Dietéticos , Yodo/administración & dosificación , Yodo/deficiencia , Intercambio Materno-Fetal/efectos de los fármacos , Enfermedades de la Tiroides/tratamiento farmacológico , Glándula Tiroides/efectos de los fármacos , Vitaminas/administración & dosificación , Adulto , Femenino , Humanos , Recién Nacido , Edad Materna , Embarazo , Resultado del Embarazo , Enfermedades de la Tiroides/metabolismo , Enfermedades de la Tiroides/patología , Glándula Tiroides/metabolismo , Hormonas Tiroideas/metabolismo
11.
J Cell Mol Med ; 24(21): 12681-12693, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33001560

RESUMEN

The placenta supplies the foetus with critical nutrients such as essential amino acids (AA, eg leucine) for development and growth. It also represents a cellular barrier which is formed by a polarized, differentiated syncytiotrophoblast (STB) monolayer. Active Na+ -independent leucine transport across the placenta is mainly attributed to the System L transporters LAT1/SLC7A5 and LAT2/SLC7A8. This study explored the influence of trophoblast differentiation on the activity of LAT1/LAT2 and the relevance of LAT1/LAT2 in leucine uptake and transfer in trophoblasts by applying specific small molecule inhibitors (JPH203/JG336/JX009). L-leucine uptake (total dose = 167 µmol/L) was sensitive to LAT1-specific inhibition by JPH203 (EC50  = 2.55 µmol/L). The inhibition efficiency of JPH203 was increased by an additional methoxy group in the JPH203-derivate JG336 (EC50  = 1.99 µmol/L). Interestingly, JX009 showed efficient System L inhibition (EC50  = 2.35 µmol/L) and was the most potent inhibitor of leucine uptake in trophoblasts. The application of JPH203 and JX009 in Transwell® -based leucine transfer revealed LAT1 as the major accumulative transporter at the apical membrane, but other System L transporters such as LAT2 as rate-limiting for leucine efflux across the basal membrane. Therefore, differential specificity of the applied inhibitors allowed for estimation of the contribution of LAT1 and LAT2 in materno-foetal AA transfer and their potential impact in pregnancy diseases associated with impaired foetal growth.


Asunto(s)
Sistema de Transporte de Aminoácidos y+/metabolismo , Cadenas Ligeras de la Proteína-1 Reguladora de Fusión/metabolismo , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Leucina/metabolismo , Intercambio Materno-Fetal , Adulto , Transporte Biológico/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular , Femenino , Cadena Pesada de la Proteína-1 Reguladora de Fusión/metabolismo , Humanos , Recién Nacido , Intercambio Materno-Fetal/efectos de los fármacos , Placenta/metabolismo , Embarazo , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Sodio/metabolismo , Trofoblastos/citología , Trofoblastos/efectos de los fármacos , Trofoblastos/metabolismo , Regulación hacia Arriba/efectos de los fármacos
12.
Drug Metab Dispos ; 48(12): 1293-1302, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33051249

RESUMEN

Two types of systems are used in ex vivo human placental perfusion studies to predict fetal drug exposures, that is, closed systems with recirculation of the maternal and fetal buffer and open systems using a single-pass mode without recirculation. The in vivo fetal/maternal (F:M) ratio of metformin, a cationic drug that crosses the placenta, is consistent with that reported in an open system ex vivo but not with that in a closed system. In the present study, we aimed to develop a pharmacokinetic (PK) model of transplacental transfer of metformin to predict in vivo fetal exposure to metformin and to resolve the apparent inconsistency between open and closed ex vivo systems. The developed model shows that the difference between open and closed systems is due to the difference in the time required to achieve the steady state. The model-predicted F:M ratio (approx. 0.88) is consistent with reported in vivo values [mean (95% confidence interval): 1.10 (0.69-1.51)]. The model incorporates bidirectional transport via organic cation transporter 3 (OCT3) at the basal plasma membrane, and simulations indicate that the use of trimethoprim (an OCT3 inhibitor) to prevent microbial growth in the placenta ex vivo has a negligible effect on the overall maternal-to-fetal and fetal-to-maternal clearances. The model could successfully predict in vivo fetal exposure using ex vivo human placental perfusion data from both closed and open systems. This transplacental PK modeling approach is expected to be useful for evaluating human fetal exposures to other poorly permeable compounds, besides metformin. SIGNIFICANCE STATEMENT: We developed a pharmacokinetic model of transplacental transfer of metformin, used to treat gestational diabetes mellitus, in order to predict in vivo fetal exposure and resolve the discrepancy between reported findings in open and closed ex vivo perfusion systems. The discrepancy is due to a difference in the time required to reach the steady state. The model can predict in vivo fetal exposure using data from both closed and open systems.


Asunto(s)
Feto/metabolismo , Intercambio Materno-Fetal/fisiología , Metformina/farmacocinética , Modelos Biológicos , Placenta/metabolismo , Membrana Celular/metabolismo , Simulación por Computador , Femenino , Feto/irrigación sanguínea , Humanos , Intercambio Materno-Fetal/efectos de los fármacos , Proteínas de Transporte de Catión Orgánico/antagonistas & inhibidores , Proteínas de Transporte de Catión Orgánico/metabolismo , Perfusión , Placenta/irrigación sanguínea , Placenta/citología , Embarazo , Trimetoprim/farmacología
13.
J Toxicol Sci ; 45(10): 639-650, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33012732

RESUMEN

Bisphenol A (BPA), an endocrine disruptor with estrogenic effects, is widely used as a raw material for manufacturing polycarbonate plastic and epoxy resins. Prenatal and postnatal exposure to BPA affects brain morphogenesis. However, the effects of prenatal and postnatal BPA exposure on postnatal neurogenesis in mice are poorly understood. In this study, we developed a mouse model of prenatal and postnatal BPA exposure and analyzed its effects on hippocampal neurogenesis. The hippocampal dentate gyrus is vulnerable to chemical exposure, as neurogenesis continues in this region even after birth. Our results showed that in mice, prenatal and postnatal BPA exposure decreased the number of type-1, 2a, 2b, and 3 neural progenitor cells, as well as in granule cells, in the hippocampal dentate gyrus on postnatal days 16 and 70. The effect of prenatal and postnatal BPA exposure on neural progenitors were affected at all differentiation stages. In addition, prenatal and postnatal BPA exposure affects the maintenance of long-term memory on postnatal day 70. Our results suggest that neurodevelopmental toxicity due to prenatal and postnatal BPA exposure might affect postnatal morphogenesis and functional development of the hippocampal dentate gyrus.


Asunto(s)
Animales Recién Nacidos , Compuestos de Bencidrilo/toxicidad , Giro Dentado/efectos de los fármacos , Disruptores Endocrinos/toxicidad , Hipocampo/efectos de los fármacos , Exposición Materna/efectos adversos , Intercambio Materno-Fetal/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Fenoles/toxicidad , Animales , Diferenciación Celular/efectos de los fármacos , Femenino , Masculino , Ratones , Modelos Animales , Embarazo
14.
Aliment Pharmacol Ther ; 52(10): 1551-1562, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32981127

RESUMEN

BACKGROUND: The impact of pregnancy on levels of biologic agents in patients with IBD is undefined and time to elimination in vedolizumab-exposed infants is unknown. AIMS: To determine the effect of pregnancy on infliximab, adalimumab and vedolizumab levels and to study infant vedolizumab clearance METHODS: In a prospective observational study, maternal drug levels were measured pre-conception, in each trimester, at delivery and postpartum. The association between drug levels and gestation in weeks was assessed using generalised estimating equation modelling. Infant vedolizumab levels were performed at birth (cord blood), 6 weeks and 3 months or until undetectable. RESULTS: We included 50 IBD patients (23 on infliximab, 15 on adalimumab and 12 on vedolizumab) with at least two intrapartum observations, plus 5 patients on vedolizumab with only mother and baby samples at delivery. Modelling showed no change in adalimumab levels, an increase in infliximab levels of 0.16 (95% CI 0.08-0.24) µg/L/week (P < 0.001) and a decrease of 0.18 (95% CI: -0.33 to -0.02) µg/L/week (P = 0.03) for vedolizumab. In 17 mother-baby pairs, median infant vedolizumab levels at birth were lower than maternal levels (P < 0.05) with an infant:maternal ratio of 0.7 (IQR 0.5-0.9). Vedolizumab was undetectable between 15 and 16 weeks of age in all 12 infants completing follow-up testing. CONCLUSIONS: During pregnancy, adalimumab levels remain stable, while infliximab levels increase and vedolizumab levels decrease. However, the increments were small suggesting that intrapartum therapeutic drug monitoring and dose adjustment are not indicated. Unlike infliximab and adalimumab, infant vedolizumab levels are lower in cord blood than in mothers and appear to clear rapidly.


Asunto(s)
Adalimumab/sangre , Anticuerpos Monoclonales Humanizados/sangre , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Infliximab/sangre , Complicaciones del Embarazo/tratamiento farmacológico , Efectos Tardíos de la Exposición Prenatal/sangre , Adalimumab/administración & dosificación , Adalimumab/farmacocinética , Adulto , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/farmacocinética , Análisis Químico de la Sangre/estadística & datos numéricos , Estudios de Cohortes , Relación Dosis-Respuesta a Droga , Monitoreo de Drogas , Femenino , Sangre Fetal/química , Sangre Fetal/metabolismo , Edad Gestacional , Humanos , Inactivación Metabólica/fisiología , Lactante , Recién Nacido , Enfermedades Inflamatorias del Intestino/sangre , Infliximab/administración & dosificación , Infliximab/farmacocinética , Masculino , Pruebas de Detección del Suero Materno , Intercambio Materno-Fetal/efectos de los fármacos , Madres , Embarazo , Complicaciones del Embarazo/sangre , Complicaciones del Embarazo/metabolismo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/diagnóstico , Efectos Tardíos de la Exposición Prenatal/metabolismo , Estudios Prospectivos
15.
Nutrients ; 12(6)2020 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-32545151

RESUMEN

Endocrine-disrupting chemicals (EDCs) are exogenous substances able to mimic or to interfere with the endocrine system, thus altering key biological processes such as organ development, reproduction, immunity, metabolism and behavior. High concentrations of EDCs are found in several everyday products including plastic bottles and food containers and they could be easily absorbed by dietary intake. In recent years, considerable interest has been raised regarding the biological effects of EDCs, particularly Bisphenol A (BPA) and phthalates, on human pregnancy and fetal development. Several evidence obtained on in vitro and animal models as well as by epidemiologic and population studies strongly indicated that endocrine disruptors could negatively impact fetal and placental health by interfering with the embryonic developing epigenome, thus establishing disease paths into adulthood. Moreover, EDCs could cause and/or contribute to the onset of severe gestational conditions as Preeclampsia (PE), Fetal Growth Restriction (FGR) and gestational diabetes in pregnancy, as well as obesity, diabetes and cardiovascular complications in reproductive age. Therefore, despite contrasting data being present in the literature, endocrine disruptors must be considered as a therapeutic target. Future actions aimed at reducing or eliminating EDC exposure during the perinatal period are mandatory to guarantee pregnancy success and preserve fetal and adult health.


Asunto(s)
Compuestos de Bencidrilo/efectos adversos , Disruptores Endocrinos/efectos adversos , Desarrollo Fetal/efectos de los fármacos , Exposición Materna/efectos adversos , Intercambio Materno-Fetal/efectos de los fármacos , Fenoles/efectos adversos , Ácidos Ftálicos/efectos adversos , Placenta/diagnóstico por imagen , Adulto , Animales , Enfermedades Cardiovasculares/inducido químicamente , Diabetes Gestacional/inducido químicamente , Femenino , Desarrollo Fetal/genética , Retardo del Crecimiento Fetal/inducido químicamente , Humanos , Obesidad Materna/inducido químicamente , Preeclampsia/inducido químicamente , Embarazo
16.
J Pineal Res ; 69(2): e12670, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32421880

RESUMEN

Melatonin has been shown to improve in vitro fertilization and offspring survival after bacterial infection, but its role in regulating maternal-fetal communication during early pregnancy has not been investigated. Results of this study demonstrated expression of abundant melatonin receptors in conceptus and endometrium during early pregnancy. In gilts, expression of melatonin receptor 1A (MTNR1A or MT1) and melatonin receptor 1B (MTNR1B or MT2) increased in trophectoderm (Tr) and uterine luminal epithelium (LE) with advancing days during early pregnancy in a different manner. Melatonin increased proliferation and migration of porcine trophectoderm (pTr) cell, the percent pTr cells in the G2 phase of the cell cycle, and the expression of implantation-related genes by pTr cells and endometrial luminal epithelium (pLE). Melatonin also attenuated the production of LPS-induced pro-inflammatory cytokines and tunicamycin-induced endoplasmic reticulum (ER) stress-sensing proteins. The expression of sirtuin 1 (SIRT1) as a potential target of melatonin increased between Days 9 and 14 of gestation. Co-treatment with SIRT1 inhibitor EX527 and melatonin restored cell-cell interactions through PI3K and MAPK signaling. Knockdown of SIRT1 decreased the expression of implantation-related genes, as well as migration of pTr and pLE cells. The expression of microRNAs regulated by SIRT1 was suppressed in response to melatonin. Furthermore, melatonin significantly increased lipopolysaccharide (LPS)-reduced fertilization and embryogenesis in zebrafish model. These results suggest that melatonin may improve the uterine-conceptus interactions via the regulation of SIRT1 during early pregnancy.


Asunto(s)
Embrión de Mamíferos/embriología , Intercambio Materno-Fetal/efectos de los fármacos , Melatonina/farmacología , Sirtuina 1/metabolismo , Útero/metabolismo , Animales , Femenino , Embarazo , Porcinos
17.
Nutrients ; 12(5)2020 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-32403449

RESUMEN

This work aims to clarify the effect of dietary supplementation with Bisphenol A (BPA), a chemical widely present in beverage and food containers, on placental glucose transfer and pregnancy outcome. The study was performed on female Sprague Dawley rats fed with a diet containing BPA (2.5, 25 or 250 µg/Kg/day) for a period of a month (virgin state) plus 20 days during pregnancy. Western blot analysis and immunohistochemistry were performed in placental tissues for glucose type 1 transporter (GLUT1). Furthermore, human trophoblast, HTR8-SV/neo cells, were used to evaluate the effect of BPA on glucose transport and uptake. Studies in rats showed that food supplementation with BPA, produces a higher fetal weight (FW) to placenta weight (PW) ratio at the lowest BPA concentration. Such low concentrations also reduced maternal weight gain in late pregnancy and up-regulated placental expression of GLUT1. Treatment of HTR8-SV/neo with the non-toxic dose of 1 nM BPA confirmed up-regulation of GLUT1 expression and revealed higher activity of the transporter with an increase in glucose uptake and GLUT1 membrane translocation. Overall, these results indicate that prenatal exposure to BPA affects pregnancy and fetal growth producing changes in the placental nutrients-glucose transfer.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Glucosa/metabolismo , Intercambio Materno-Fetal/efectos de los fármacos , Fenoles/toxicidad , Placenta/metabolismo , Trofoblastos/efectos de los fármacos , Animales , Compuestos de Bencidrilo/administración & dosificación , Peso Corporal/efectos de los fármacos , Línea Celular , Femenino , Peso Fetal/efectos de los fármacos , Transportador de Glucosa de Tipo 1/análisis , Transportador de Glucosa de Tipo 1/metabolismo , Humanos , Tamaño de los Órganos/efectos de los fármacos , Fenoles/administración & dosificación , Placenta/anatomía & histología , Placenta/química , Embarazo , Ratas , Ratas Sprague-Dawley , Trofoblastos/metabolismo
18.
PLoS One ; 15(4): e0232140, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32353010

RESUMEN

BACKGROUND: Congenital cytomegalovirus infection can lead to severe sequelae. When fetal infection is confirmed, we hypothesize that fetal treatment could improve the outcome. Maternal oral administration of an effective drug crossing the placenta could allow fetal treatment. Letermovir (LMV) and Maribavir (MBV) are new CMV antivirals, and potential candidates for fetal treatment. METHODS: The objective was to investigate the placental transfer of LMV and MBV in the ex vivo method of the human perfused cotyledon. Term placentas were perfused, in an open-circuit model, with LMV or MBV at concentrations in the range of clinical peak plasma concentrations. Concentrations were measured using ultraperformance liquid chromatography coupled with tandem mass spectrometry. Mean fetal transfer rate (FTR) (fetal (FC) /maternal concentration), clearance index (CLI), accumulation index (AI) (retention of each drug in the cotyledon tissue) were measured. Mean FC were compared with half maximal effective concentrations of the drugs (EC50(LMV) and EC50(MBV)). RESULTS: For LMV, the mean FC was (± standard deviation) 1.1 ± 0.2 mg/L, 1,000-fold above the EC50(LMV). Mean FTR, CLI and AI were 9 ± 1%, 35 ± 6% and 4 ± 2% respectively. For MBV, the mean FC was 1.4 ± 0.2 mg/L, 28-fold above the EC50(MBV). Mean FTR, CLI and AI were 10 ± 1%, 50 ± 7% and 2 ± 1% respectively. CONCLUSIONS: Drugs' concentrations in the fetal side should be in the range for in utero treatment of fetuses infected with CMV as the mean FC was superior to the EC50 for both molecules.


Asunto(s)
Infecciones por Citomegalovirus/tratamiento farmacológico , Intercambio Materno-Fetal/efectos de los fármacos , Placenta/efectos de los fármacos , Acetatos/farmacología , Adulto , Antivirales/farmacología , Bencimidazoles/farmacología , Cromatografía Liquida/métodos , Femenino , Humanos , Cinética , Modelos Biológicos , Perfusión , Embarazo , Quinazolinas/farmacología , Ribonucleósidos/farmacología , Espectrometría de Masas en Tándem/métodos
19.
Nutrients ; 12(5)2020 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-32365792

RESUMEN

Gestational diabetes mellitus (GDM) results in reduced docosahexaenoic acid (DHA) transfer to the fetus, likely due to placental dysfunction. Sirtuin-1 (SIRT1) is a nutrient sensor and regulator of lipid metabolism. This study investigated whether the high glucose and insulin condition of GDM regulates DHA transfer and expression of fatty acid transporters and if this effect is related to SIRT1 expression and function. Syncytialized primary human trophoblasts were treated with and without glucose (25 mmol/L) and insulin (10-7 mol/L) for 72 h to mimic the insulin-resistance conditions of GDM pregnancies. In control conditions, DHA transfer across trophoblasts increased in a time- and dose-dependent manner. Exposure to GDM conditions significantly decreased DHA transfer, but increased triglyceride accumulation and fatty acid transporter expression (CD36, FABP3, and FABP4). GDM conditions significantly suppressed SIRT1 mRNA and protein expression. The SIRT1 inhibitor decreased DHA transfer across control trophoblasts, and recombinant SIRT1 and SIRT1 activators restored the decreased DHA transport induced by GDM conditions. The results demonstrate a novel role of SIRT1 in the regulation of DHA transfer across trophoblasts. The suppressed SIRT1 expression and the resultant decrease in placental DHA transfer caused by high glucose and insulin levels suggest new insights of molecular mechanisms linking GDM to fetal DHA deficiency.


Asunto(s)
Ácidos Docosahexaenoicos/metabolismo , Expresión Génica/efectos de los fármacos , Glucosa/efectos adversos , Insulina/efectos adversos , Intercambio Materno-Fetal/efectos de los fármacos , Sirtuina 1/metabolismo , Sirtuina 1/fisiología , Trofoblastos/metabolismo , Antígenos CD36/metabolismo , Células Cultivadas , Diabetes Gestacional/genética , Diabetes Gestacional/metabolismo , Relación Dosis-Respuesta a Droga , Proteína 3 de Unión a Ácidos Grasos/metabolismo , Proteínas de Transporte de Ácidos Grasos/genética , Proteínas de Transporte de Ácidos Grasos/metabolismo , Femenino , Humanos , Placenta/metabolismo , Embarazo , ARN Mensajero/metabolismo , Sirtuina 1/genética
20.
Prenat Diagn ; 40(9): 1156-1167, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32335932

RESUMEN

Drug entry into the adult brain is controlled by efflux mechanisms situated in various brain barrier interfaces. The effectiveness of these protective mechanisms in the embryo, fetus and newborn brain is less clear. The longstanding belief that "the" blood-brain barrier is absent or immature in the fetus and newborn has led to many misleading statements with potential clinical implications. Here we review the properties of brain barrier mechanisms in the context of drug entry into the developing brain and discuss the limited number of studies published on the subject. We noticed that most of available literature suffers from some experimental limitations, notably that drug levels in fetal blood and cerebrospinal fluid have not been measured. This means that the relative contribution to the overall brain protection provided by individual barriers such as the placenta (which contains similar efflux mechanisms) and the brain barriers cannot be separately ascertained. Finally, we propose that systematic studies in appropriate animal models of drug entry into the brain at different stages of development would provide a rational basis for use of medications in pregnancy and in newborns, especially prematurely born, where protection usually provided by the placenta is no longer present.


Asunto(s)
Intercambio Materno-Fetal/efectos de los fármacos , Preparaciones Farmacéuticas , Complicaciones del Embarazo/tratamiento farmacológico , Animales , Toma de Decisiones , Femenino , Feto/efectos de los fármacos , Humanos , Recién Nacido , Madres , Preparaciones Farmacéuticas/clasificación , Embarazo , Complicaciones del Embarazo/epidemiología , Mujeres Embarazadas , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/epidemiología , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...