Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 813
Filtrar
Más filtros

Intervalo de año de publicación
1.
Am J Pathol ; 194(9): 1636-1663, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39182948

RESUMEN

Air pollution exposure during pregnancy may affect fetal growth. Fetal growth restriction (FGR) is associated with reduced lung function in children that can persist into adulthood. Using an established model of asymmetrical FGR in Long-Evans rats, this study investigated sex differences in effects of early life ozone exposure on lung development and maturation. Adverse health effects for i) gestational exposure (with impacts on primary alveolarization), ii) peri-adolescent exposure (with impacts on secondary alveolarization), and iii) cumulative exposure across both periods were evaluated. Notably, female offspring were most affected by gestational ozone exposure, likely because of impaired angiogenesis and corresponding decreases in primary alveolarization. Females had diminished lung capacity, fewer mature alveoli, and medial hypertrophy of small and large pulmonary arteries. Males, especially FGR-prone offspring, were more affected by peri-adolescent ozone exposure. Males had increased ductal areas, likely due to disrupted secondary alveolarization. Altered lung development may increase risk of developing diseases, such as pulmonary arterial hypertension or chronic obstructive pulmonary disease. Pulmonary arterial hypertension disproportionately affects women. In the United States, chronic obstructive pulmonary disease prevalence is increasing, especially in women; and prevalence for both men and women is highest in urbanized areas. This investigation underlines the importance of evaluating results separately by sex, and provides biologic plausibility for later consequences of early-life exposure to ozone, a ubiquitous urban air pollutant.


Asunto(s)
Pulmón , Ozono , Efectos Tardíos de la Exposición Prenatal , Ratas Long-Evans , Animales , Ozono/toxicidad , Ozono/efectos adversos , Femenino , Masculino , Embarazo , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/crecimiento & desarrollo , Ratas , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/patología , Caracteres Sexuales , Humanos , Retardo del Crecimiento Fetal/inducido químicamente , Retardo del Crecimiento Fetal/patología , Contaminantes Atmosféricos/toxicidad , Contaminantes Atmosféricos/efectos adversos
2.
FASEB J ; 37(3): e22820, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36801982

RESUMEN

Epidemiological studies suggest that fetal growth restriction (FGR) caused by gestational cholestasis is associated with elevated serum cholic acid (CA). Here, we explore the mechanism by which CA induces FGR. Pregnant mice except controls were orally administered with CA daily from gestational day 13 (GD13) to GD17. Results found that CA exposure decreased fetal weight and crown-rump length, and increased the incidence of FGR in a dose-dependent manner. Furthermore, CA caused placental glucocorticoid (GC) barrier dysfunction via down-regulating the protein but not the mRNA level of placental 11ß-Hydroxysteroid dehydrogenase-2 (11ß-HSD2). Additionally, CA activated placental GCN2/eIF2α pathway. GCN2iB, an inhibitor of GCN2, significantly inhibited CA-induced down-regulation of 11ß-HSD2 protein. We further found that CA caused excessive reactive oxygen species (ROS) production and oxidative stress in mouse placentas and human trophoblasts. NAC significantly rescued CA-induced placental barrier dysfunction by inhibiting activation of GCN2/eIF2α pathway and subsequent down-regulation of 11ß-HSD2 protein in placental trophoblasts. Importantly, NAC rescued CA-induced FGR in mice. Overall, our results suggest that CA exposure during late pregnancy induces placental GC barrier dysfunction and subsequent FGR may be via ROS-mediated placental GCN2/eIF2α activation. This study provides valuable insight for understanding the mechanism of cholestasis-induced placental dysfunction and subsequent FGR.


Asunto(s)
Enfermedades Placentarias , Placenta , Embarazo , Femenino , Ratones , Humanos , Animales , Placenta/metabolismo , Especies Reactivas de Oxígeno/metabolismo , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/genética , Retardo del Crecimiento Fetal/inducido químicamente , Factor 2 Eucariótico de Iniciación/metabolismo , Enfermedades Placentarias/metabolismo
3.
Arterioscler Thromb Vasc Biol ; 43(6): e190-e209, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37051927

RESUMEN

BACKGROUND: Abnormal placental angiogenesis is an important cause of fetal intrauterine growth restriction (IUGR), but its underlying mechanisms and therapies remain unclear. Adenosine and its mediated signaling has been reported to be associated with the development of angiogenesis. However, whether the adenosine-related signaling plays a role in modulating angiogenesis in placenta and the IUGR pregnancy outcomes remains unclear. METHODS: The angiogenesis and adenosine signaling expressions in normal and IUGR placentas were detected in different species. And the role of adenosine in regulating IUGR pregnancy outcomes was evaluated using diet-induced IUGR mouse model. Molecular mechanisms underlying adenosine-induced angiogenesis were investigated by in vitro angiogenesis assays and in vivo Matrigel plug assays. RESULTS: Here, we demonstrated poor angiogenesis and low adenosine concentration and downregulated expression of its receptor A2a (ADORA2A [adenosine A2a receptor]) in IUGR placenta. Additionally, the beneficial effects of adenosine in improving IUGR pregnancy outcomes were revealed in a diet-induced IUGR mouse model. Moreover, adenosine was found to effectively improve adenosine signaling and angiogenesis in IUGR mice placenta. Mechanistically, by using angiogenesis assays in vitro and in vivo, adenosine was shown to activate ADORA2A to promote the phosphorylation of Stat3 (signal transducer and activator of transcription 3) and Akt (protein kinase B), resulting in increased Ang (angiogenin)-dependent angiogenesis. CONCLUSIONS: Collectively, this study uncovers an unexpected mechanism of promoting placental angiogenesis by adenosine-ADORA2A signaling and advances the translation of this signaling as a prognostic indicator and therapeutic target in IUGR treatment.


Asunto(s)
Placenta , Proteínas Proto-Oncogénicas c-akt , Animales , Femenino , Humanos , Ratones , Embarazo , Retardo del Crecimiento Fetal/inducido químicamente , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor de Adenosina A2A/metabolismo , Factor de Transcripción STAT3/metabolismo
4.
Part Fibre Toxicol ; 21(1): 36, 2024 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-39261835

RESUMEN

Microplastics (MPs), a brand-new class of worldwide environmental pollutant, have received a lot of attention. MPs are consumed by both humans and animals through water, food chain and other ways, which may cause potential health risks. However, the effects of MPs on embryonic development, especially placental function, and its related mechanisms still need to be further studied. We investigated the impact on fetal development and placental physiological function of pregnant mice by consecutive gavages of MPs at 0, 25, 50, 100 mg/kg body weight during gestational days (GDs 0-14). The results showed that continuous exposure to high concentrations of MP significantly reduced daily weight gain and impaired reproductive performance of pregnant mice. In addition, MPs could significantly induce oxidative stress and placental dysfunction in pregnant mice. On the other hand, MPs exposure significantly decreased placental barrier function and induced placental inflammation. Specifically, MPs treatment significantly reduced the expression of tight junction proteins in placentas, accompanied by inflammatory cell infiltration and increased mRNA levels of pro-inflammatory cytokines and chemokines in placentas. Finally, we found that MPs induced placental apoptosis and endoplasmic reticulum (ER) stress through the GRP78/IRE1α/JNK axis, leading to placental dysfunction and decreased reproductive performance in pregnant mice. We revealed for the first time that the effects of MPs on placental dysfunction in pregnant animals. Blocking the targets of MPs mediated ER stress will provide potential therapeutic ideas for the toxic effects of MPs on maternal pregnancy.


Asunto(s)
Apoptosis , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico , Microplásticos , Placenta , Animales , Femenino , Embarazo , Chaperón BiP del Retículo Endoplásmico/metabolismo , Apoptosis/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Placenta/efectos de los fármacos , Placenta/metabolismo , Microplásticos/toxicidad , Ratones , Estrés Oxidativo/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas de Choque Térmico/metabolismo , Proteínas de Choque Térmico/genética , Retardo del Crecimiento Fetal/inducido químicamente , Ratones Endogámicos ICR
5.
Arch Toxicol ; 98(6): 1891-1908, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38522057

RESUMEN

Dexamethasone is widely used in pregnant women at risk of preterm birth to reduce the occurrence of neonatal respiratory distress syndrome and subsequently reduce neonatal mortality. Studies have suggested that dexamethasone has developmental toxicity, but there is a notable absence of systematic investigations about its characteristics. In this study, we examined the effects of prenatal dexamethasone exposure (PDE) on mother/fetal mice at different doses (0.2, 0.4, or 0.8 mg/kg b.i.d), stages (gestational day 14-15 or 16-17) and courses (single- or double-course) based on the clinical practice. Results showed that PDE increased intrauterine growth retardation rate, and disordered the serum glucose, lipid and cholesterol metabolic phenotypes, and sex hormone level of mother/fetal mice. PDE was further discovered to interfere with the development of fetal lung, hippocampus and bone, inhibits steroid synthesis in adrenal and testis, and promotes steroid synthesis in the ovary and lipid synthesis in the liver, with significant effects observed at high dose, early stage and double course. The order of severity might be: ovary > lung > hippocampus/bone > others. Correlation analysis revealed that the decreased serum corticosterone and insulin-like growth factor 1 (IGF1) levels were closely related to PDE-induced low birth weight and abnormal multi-organ development in offspring. In conclusion, this study systematically confirmed PDE-induced multi-organ developmental toxicity, elucidated its characteristics, and proposed the potential "glucocorticoid (GC)-IGF1" axis programming mechanism. This research provided an experimental foundation for a comprehensive understanding of the effect and characteristics of dexamethasone on fetal multi-organ development, thereby guiding the application of "precision medicine" during pregnancy.


Asunto(s)
Dexametasona , Relación Dosis-Respuesta a Droga , Desarrollo Fetal , Animales , Femenino , Embarazo , Dexametasona/toxicidad , Dexametasona/administración & dosificación , Masculino , Desarrollo Fetal/efectos de los fármacos , Ratones , Retardo del Crecimiento Fetal/inducido químicamente , Factor I del Crecimiento Similar a la Insulina/metabolismo , Glucocorticoides/toxicidad , Glucocorticoides/administración & dosificación , Exposición Materna/efectos adversos , Efectos Tardíos de la Exposición Prenatal/inducido químicamente
6.
Ecotoxicol Environ Saf ; 283: 116840, 2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-39126814

RESUMEN

BACKGROUND: Nationwide evidence linking maternal ozone exposure with fetal growth restriction (FGR) was extensively scarce, especially in the Middle East with dry climate and distinct religious culture. METHODS: We carried out a national retrospective birth cohort study using registry-based records from 749 hospitals across 31 provinces in Iran from 2013 to 2018. Monthly concentrations of maximum daily average 8-hour (MDA8) ozone at 0.125° × 0.125° resolution were extracted from well-validated spatiotemporal grid dataset. Linear and logistic regression models were employed to evaluate associations of maternal MDA8 ozone exposure with birthweight outcomes. Assuming causality, the comparative risk assessment framework was utilized to estimate the burden of low birthweight (LBW), small for gestational age (SGA), and birthweight loss per livebirth (BLL) attributable to ambient ozone pollution. RESULTS: Of 4030383 livebirths included in the study, 264304 (6.6%) were LBW and 484405 (12.0%) were SGA. Each 10-ppb increase in MDA8 ozone exposure was associated with an odds ratio of 1.123 (95% confidence interval [CI]: 1.104 to 1.142) for LBW and 1.210 (95% CI: 1.197 to 1.223) for SGA, and a 30.5-g (95% CI: 29.0 to 32.0) reduction in birthweight. We observed approximately linear exposure-response relationships of maternal MDA8 ozone exposure with LBW (Pnonlinear= 0.786), SGA (Pnonlinear= 0.156), and birthweight reduction (Pnonlinear= 0.104). Under the premise of causal association, we estimated 6.6% (95% CI: 5.7 to 7.5) of LBW, 10.1% (95% CI: 9.6 to 10.6) of SGA, and 18.8 g (95% CI: 17.9 to 19.7) of BLL could be attributable to maternal ozone exposure in Iran. Considerably greater risk and burden of ozone-related FGR were observed among younger, less-educated, and rural-dwelling mothers. CONCLUSIONS: Our study provided compelling evidence that maternal ozone exposure was associated with heightened FGR risk and burden, particularly among socioeconomically disadvantaged mothers. These findings underscored the urgent need for government to incorporate socioeconomic factors into future ozone-related health policies, not only to mitigate pollution, but also minimize inequality.


Asunto(s)
Contaminantes Atmosféricos , Peso al Nacer , Recién Nacido de Bajo Peso , Exposición Materna , Ozono , Humanos , Ozono/análisis , Ozono/efectos adversos , Femenino , Irán/epidemiología , Recién Nacido , Exposición Materna/estadística & datos numéricos , Exposición Materna/efectos adversos , Embarazo , Estudios Retrospectivos , Peso al Nacer/efectos de los fármacos , Adulto , Contaminantes Atmosféricos/análisis , Recién Nacido Pequeño para la Edad Gestacional , Estudios de Cohortes , Retardo del Crecimiento Fetal/epidemiología , Retardo del Crecimiento Fetal/inducido químicamente , Masculino , Adulto Joven
7.
Ecotoxicol Environ Saf ; 269: 115797, 2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-38070418

RESUMEN

Prenatal caffeine exposure (PCE) is a significant contributor to intrauterine growth retardation (IUGR) in offspring, which has been linked to an increased susceptibility to autism spectrum disorder (ASD) later in life. Additionally, a high-fat diet (HFD) has been shown to exacerbate ASD-like behaviors, but the underlying mechanisms remain unclear. In this study, we first noted in the rat model of IUGR induced by PCE that male PCE offspring exhibited typical ASD-like behaviors post-birth, in contrast to their female counterparts. The female PCE offspring demonstrated only reduced abilities in free exploration and spatial memory. Importantly, both male and female PCE offspring displayed ASD-like behaviors when exposed to HFD. We further observed that PCE + HFD offspring exhibited damaged intestinal mucus barriers and disturbed gut microbiota, resulting in an increased abundance of Escherichia coli (E. coli). The induced differentiation of colonic Th17 cells by E. coli led to an increased secretion of IL-17A, which entered the hippocampus through peripheral circulation and caused synaptic damage in hippocampal neurons, ultimately resulting in ASD development. Our strain transplantation experiment suggested that E. coli-mediated increase of IL-17A may be the core mechanism of ASD with a fetal origin. In conclusion, PCE and HFD are potential risk factors for ASD, and E. coli-mediated IL-17A may play a crucial role in fetal-originated ASD through the gut-brain axis.


Asunto(s)
Trastorno del Espectro Autista , Trastorno Autístico , Cafeína , Microbioma Gastrointestinal , Efectos Tardíos de la Exposición Prenatal , Animales , Femenino , Humanos , Masculino , Embarazo , Ratas , Trastorno del Espectro Autista/inducido químicamente , Trastorno del Espectro Autista/microbiología , Trastorno Autístico/inducido químicamente , Trastorno Autístico/microbiología , Encéfalo , Eje Cerebro-Intestino , Cafeína/efectos adversos , Cafeína/toxicidad , Dieta Alta en Grasa/efectos adversos , Escherichia coli , Retardo del Crecimiento Fetal/inducido químicamente , Microbioma Gastrointestinal/efectos de los fármacos , Interleucina-17/genética , Efectos Tardíos de la Exposición Prenatal/inducido químicamente
8.
Ecotoxicol Environ Saf ; 279: 116492, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38795415

RESUMEN

Pregnant women are a special group that is sensitive to adverse external stimuli, causing metabolic abnormalities and adverse pregnancy outcomes. Microplastics (MPs), an environmental pollutant widely used in various fields, can induce a variety of toxic responses in mammals. Recent studies verified an association between MPs and metabolic disorders. Our research built a gestational mouse model in which polystyrene microplastics (PS-MPs) of 1 µm size were consumed at concentrations of 0.1, 1, and 10 mg/L during pregnancy. Results indicated that PS-MPs induced placental malfunction and fetal growth retardation. Significant glucose disorders, decreased liver function, hepatic inflammation, and oxidative stress were also observed after PS-MPs exposure. The hepatic SIRT1/IRS1/PI3K pathway was inhibited in the 10 mg/L PS-MPs exposure group. Our study found that PS-MPs activated inflammatory response and oxidative stress by increasing hepatic lipopolysaccharide (LPS) that inhibited the hepatic SIRT1/IRS1/PI3K pathway, ultimately leading to insulin resistance, glucose metabolism disorders, and adverse pregnancy outcomes. This study provides a basis for preventing environment-related gestational diabetes and concomitant adverse pregnancy outcomes.


Asunto(s)
Microplásticos , Estrés Oxidativo , Poliestirenos , Resultado del Embarazo , Sirtuina 1 , Femenino , Embarazo , Poliestirenos/toxicidad , Animales , Microplásticos/toxicidad , Ratones , Sirtuina 1/metabolismo , Estrés Oxidativo/efectos de los fármacos , Homeostasis/efectos de los fármacos , Glucosa/metabolismo , Placenta/efectos de los fármacos , Contaminantes Ambientales/toxicidad , Proteínas Sustrato del Receptor de Insulina/metabolismo , Retardo del Crecimiento Fetal/inducido químicamente , Fosfatidilinositol 3-Quinasas/metabolismo , Hígado/efectos de los fármacos
9.
Mol Med ; 29(1): 114, 2023 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-37718409

RESUMEN

BACKGROUND: Sexual dimorphism in placental physiology affects the functionality of placental adaptation during adverse pregnancy. Defects of placental function compromise fetal programming, affecting the offspring's adult life. However, studies focusing on the relationship between sex-specific placental adaptation and consequent fetal maldevelopment under sub-optimal uterus milieu are still elusive. METHODS: Here, we investigated the effects of maternal lipopolysaccharide (LPS) exposure between placental sex. Pregnant ICR mice received intraperitoneal injection of phosphate-buffered saline or 100, 200, and 400 µg/kg LPS on the gestational day (GD) 15.5. To determine whether prenatal maternal LPS exposure resulted in complicated pregnancy outcomes, survival rate of embryos was calculated and the growth of embryos and placentas was examined. To elucidate global transcriptomic changes occurring in the placenta, total RNA-sequencing (RNA-seq) was performed in female and male placentas. RESULTS: LPS administration induced placental inflammation in both sexes at GD 17.5. Prenatal infection resulted in growth retardation in both sexes of embryos, and especially more prevalently in male. Impaired placental development was observed in a sex-specific manner. LPS 400 µg/kg reduced the percentage area of the labyrinth in females and junctional zone in males, respectively. RNA-sequencing revealed widespread sexually dimorphic transcriptional changes in placenta. In particular, representative changes were involved in biological processes such as trophoblast differentiation, nutrient/ion transporter, pregnancy, and immune system. CONCLUSIONS: Our results present the sexually dimorphic responses of placental physiology in intrauterine growth restriction model and provide tentative relationship further to be elucidated between sex-biased placental functional change and long-term effects on the offspring's later life.


Asunto(s)
Retardo del Crecimiento Fetal , Lipopolisacáridos , Femenino , Masculino , Embarazo , Ratones , Animales , Humanos , Ratones Endogámicos ICR , Retardo del Crecimiento Fetal/inducido químicamente , Placenta , ARN
10.
Environ Health ; 22(1): 71, 2023 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-37858139

RESUMEN

BACKGROUND: Few studies have assessed air pollution exposure association with birthweight during both preconception and gestational periods. METHODS: Leveraging a preconception cohort consisting of 14220 pregnant women and newborn children in Shanghai, China during 2016-2018, we aim to assess associations of NO2 and PM2.5 exposure, derived from high-resolution spatial-temporal models, during preconception and gestational periods with outcomes including term birthweight, birthweight Z-score, small-for-gestational age (SGA) and large-for-gestational age (LGA). Linear and logistic regressions were used to estimate 3-month preconception and trimester-averaged air pollution exposure associations; and distributed lag models (DLM) were used to identify critical exposure windows at the weekly resolution from preconception to delivery. Two-pollutant models and children's sex-specific associations were explored. RESULTS: After controlling for covariates, one standard deviation (SD) (11.5 µg/m3, equivalent to 6.1 ppb) increase in NO2 exposure during the second and the third trimester was associated with 13% (95% confidence interval: 2 - 26%) and 14% (95% CI: 1 - 29%) increase in SGA, respectively; and one SD (9.6 µg/m3) increase in PM2.5 exposure during the third trimester was associated with 15% (95% CI: 1 - 31%) increase in SGA. No association have been found for outcomes of birthweight, birthweight Z-score and LGA. DLM found that gestational weeks 22-32 were a critical window, when NO2 exposure had strongest associations with SGA. The associations of air pollution exposure tended to be stronger in female newborns than in male newborns. However, no significant associations of air pollution exposure during preconception period on birthweight outcomes were found. CONCLUSION: Consistent with previous studies, we found that air pollution exposure during mid-to-late pregnancy was associated with adverse birthweight outcomes.


Asunto(s)
Contaminantes Atmosféricos , Contaminación del Aire , Femenino , Recién Nacido , Embarazo , Masculino , Humanos , Peso al Nacer , Contaminantes Atmosféricos/efectos adversos , Contaminantes Atmosféricos/análisis , Estudios Prospectivos , Dióxido de Nitrógeno/análisis , Exposición Materna/efectos adversos , China/epidemiología , Contaminación del Aire/análisis , Retardo del Crecimiento Fetal/inducido químicamente , Material Particulado/análisis
11.
Arch Toxicol ; 97(11): 2929-2941, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37603095

RESUMEN

Maternal lipopolysaccharide (LPS) exposure during pregnancy has been related to IUGR. Here, we explored whether paternal LPS exposure before mating impaired fetal development. All male mice except controls were intraperitoneally injected with LPS every other day for a total of five injections. The next day after the last LPS, male mice were mated with untreated female mice. Interestingly, fetal weight and crown-rump length were reduced, while the incidence of IUGR was increased in paternal LPS exposure group. Additionally, paternal LPS exposure leaded to poor placental development through causing cell proliferation inhibition and apoptosis. Additional experiment demonstrated that the inactivation of placental PI3K/AKT pathway might be involved in paternal LPS-induced cell proliferation inhibition and apoptosis of trophoblast cells. Furthermore, the mRNA and protein levels of mesoderm specific transcript (MEST), a maternally imprinted gene with paternal expression, were significantly decreased in mouse placentas from paternal LPS exposure. Further analysis showed that paternal LPS exposure caused the inactivation of placental PI3K/AKT pathway and then cell proliferation inhibition and apoptosis might be via down-regulating placental MEST. Overall, our results provide evidence that paternal LPS exposure causes poor placental development and subsequently IUGR may be via down-regulating MEST/PI3K/AKT pathway, and then inducing cell proliferation inhibition and apoptosis in placentas.


Asunto(s)
Retardo del Crecimiento Fetal , Lipopolisacáridos , Femenino , Masculino , Embarazo , Animales , Ratones , Humanos , Retardo del Crecimiento Fetal/inducido químicamente , Lipopolisacáridos/toxicidad , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Placenta , Placentación
12.
Development ; 146(11)2019 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-31182432

RESUMEN

The development of pathologies during pregnancy, including pre-eclampsia, hypertension and fetal growth restriction (FGR), often originates from poor functioning of the placenta. In vivo models of maternal stressors, such as nutrient deficiency, and placental insufficiency often focus on inadequate growth of the fetus and placenta in late gestation. These studies rarely investigate the origins of poor placental formation in early gestation, including those affecting the pre-implantation embryo and/or the uterine environment. The current study characterises the impact on blastocyst, uterine and placental outcomes in a rat model of periconceptional alcohol exposure, in which 12.5% ethanol is administered in a liquid diet from 4 days before until 4 days after conception. We show female-specific effects on trophoblast differentiation, embryo-uterine communication, and formation of the placental vasculature, resulting in markedly reduced placental volume at embryonic day 15. Both sexes exhibited reduced trophectoderm pluripotency and global hypermethylation, suggestive of inappropriate epigenetic reprogramming. Furthermore, evidence of reduced placental nutrient exchange and reduced pre-implantation maternal plasma choline levels offers significant mechanistic insight into the origins of FGR in this model.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Etanol/efectos adversos , Fertilización/efectos de los fármacos , Placentación/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal , Trofoblastos/efectos de los fármacos , Consumo de Bebidas Alcohólicas/fisiopatología , Animales , Embrión de Mamíferos , Etanol/administración & dosificación , Femenino , Retardo del Crecimiento Fetal/inducido químicamente , Retardo del Crecimiento Fetal/patología , Retardo del Crecimiento Fetal/fisiopatología , Masculino , Exposición Materna/efectos adversos , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/patología , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Ratas , Ratas Sprague-Dawley , Caracteres Sexuales , Trofoblastos/fisiología
13.
Expert Rev Mol Med ; 24: e26, 2022 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-35687009

RESUMEN

Pregnancy is a complex biological process. The establishment and maintenance of foetal-maternal interface are pivotal events. Decidual immune cells and inflammatory cytokines play indispensable roles in the foetal-maternal interface. The disfunction of decidual immune cells leads to adverse pregnancy outcome. Tumour necrosis factor (TNF)-α, a common inflammatory cytokine, has critical roles in different stages of normal pregnancy process. However, the relationship between the disorder of TNF-α and adverse pregnancy outcomes, including preeclampsia (PE), intrauterine growth restriction (IUGR), spontaneous abortion (SA), preterm birth and so on, is still indefinite. In this review, we thoroughly reviewed the effect of TNF-α disorder on pathological conditions. Moreover, we summarized the reports about the adverse pregnancy outcomes (PE, IUGR, SA and preterm birth) of using anti-TNF-α drugs (infliximab, etanercept and adalimumab, certolizumab and golimumab) currently in the clinical studies. Overall, IUGR, SA and preterm birth are the most common adverse pregnancy outcomes of anti-TNF-α drugs. Our review may provide insight for the immunological treatment of pregnancy-related complication, and help practitioners make informed decisions based on the current evidences.


Asunto(s)
Aborto Espontáneo , Preeclampsia , Complicaciones del Embarazo , Nacimiento Prematuro , Aborto Espontáneo/inducido químicamente , Adalimumab/efectos adversos , Citocinas , Femenino , Retardo del Crecimiento Fetal/inducido químicamente , Retardo del Crecimiento Fetal/etiología , Humanos , Recién Nacido , Preeclampsia/inducido químicamente , Preeclampsia/etiología , Embarazo , Complicaciones del Embarazo/inducido químicamente , Complicaciones del Embarazo/tratamiento farmacológico , Resultado del Embarazo , Nacimiento Prematuro/inducido químicamente , Inhibidores del Factor de Necrosis Tumoral , Factor de Necrosis Tumoral alfa
14.
FASEB J ; 35(12): e22035, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34748230

RESUMEN

Epigenetic mechanisms of paternal inheritance are an emerging area of interest in our efforts to understand fetal alcohol spectrum disorders. In rodent models examining maternal alcohol exposures, different maternal genetic backgrounds protect or sensitize offspring to alcohol-induced teratogenesis. However, whether maternal background can mitigate sperm-inherited alterations in developmental programming and modify the penetrance of growth defects induced by preconception paternal alcohol exposures remains unaddressed. In our previous studies examining pure C57Bl/6J crosses, the offspring of alcohol-exposed sires exhibited fetal growth restriction, enlarged placentas, and decreased placental efficiency. Here, we find that in contrast to our previous studies, the F1 offspring of alcohol-exposed C57Bl/6J sires and CD-1 dams do not exhibit fetal growth restriction, with male fetuses developing smaller placentas and increased placental efficiencies. However, in these hybrid offspring, preconception paternal alcohol exposure induces sex-specific changes in placental morphology. Specifically, the female offspring of alcohol-exposed sires displayed structural changes in the junctional and labyrinth zones, along with increased placental glycogen content. These changes in placental organization are accompanied by female-specific alterations in the expression of imprinted genes Cdkn1c and H19. Although male placentae do not display overt changes in placental histology, using RNA-sequencing, we identified programmed alterations in genes regulating oxidative phosphorylation, mitochondrial function, and Sirtuin signaling. Collectively, our data reveal that preconception paternal alcohol exposure transmits a stressor to developing offspring, that males and females exhibit distinct patterns of placental adaptation, and that maternal genetic background can modulate the effects of paternal alcohol exposure.


Asunto(s)
Adaptación Fisiológica , Etanol/toxicidad , Trastornos del Espectro Alcohólico Fetal/patología , Retardo del Crecimiento Fetal/patología , Herencia Paterna , Penetrancia , Placenta/fisiopatología , Animales , Epigénesis Genética , Femenino , Trastornos del Espectro Alcohólico Fetal/etiología , Retardo del Crecimiento Fetal/inducido químicamente , Masculino , Ratones , Ratones Endogámicos C57BL , Fenotipo , Embarazo , Factores Sexuales , Transcriptoma
15.
Reprod Biomed Online ; 44(4): 595-607, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35232674

RESUMEN

RESEARCH QUESTION: How does progesterone improve fetal outcome and change the expression of placental glucose transporters (GLUT) in dexamethasone-induced intrauterine growth restriction (IUGR)? DESIGN: A total of 64 rats were divided randomly into four different treatment groups based on daily i.p. injections of either saline or dexamethasone in the presence or absence of progesterone. Injections started on the 15th day of gestation (15dg) and lasted until the day of sacrifice at 19dg or 21dg. Maternal plasma progesterone concentrations were measured by enzyme-linked immunosorbent assay. The gene and protein expression of placental GLUT1 and GLUT3 were evaluated in the placental labyrinth and basal zones by real-time polymerase chain reaction and Western blotting, respectively. The localization of GLUT1 and GLUT3 was evaluated by immunohistochemistry. RESULTS: Dexamethasone induced significant decreases in maternal serum progesterone concentrations (P = 0.029) and placental (P < 0.001) and fetal body (P = 0.009) weights. Dexamethasone also reduced the expression of GLUT1 in the labyrinth zone (P = 0.028) and GLUT3 in both the labyrinth (P = 0.002) and basal zones (P = 0.026). Coadministration of dexamethasone and progesterone prevented the reduction in fetal body weight, placental weight and placental GLUT expression compared with that seen in dexamethasone-treated groups. CONCLUSION: These results suggest that progesterone prevents the significant reduction in fetal and placental weights in dexamethasone-induced IUGR, possibly through improving the expression of placental GLUT.


Asunto(s)
Retardo del Crecimiento Fetal , Placenta , Animales , Femenino , Embarazo , Ratas , Dexametasona/efectos adversos , Dexametasona/metabolismo , Retardo del Crecimiento Fetal/inducido químicamente , Glucosa/metabolismo , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Transportador de Glucosa de Tipo 1/genética , Transportador de Glucosa de Tipo 1/metabolismo , Transportador de Glucosa de Tipo 3/genética , Transportador de Glucosa de Tipo 3/metabolismo , Placenta/metabolismo , Progesterona/metabolismo
16.
Alcohol Clin Exp Res ; 46(6): 1036-1049, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35474222

RESUMEN

BACKGROUND: Prenatal alcohol exposure (PAE) has been shown to alter fetal blood flow in utero and is also associated with placental insufficiency and intrauterine growth restriction (IUGR), suggesting an underlying connection between perturbed circulation and pregnancy outcomes. METHODS: Timed-pregnant C57/BL6NHsd mice, bred in-house, were exposed by gavage on gestational day 10 (GD10) to ethanol (3 g/kg) or purified water, as a control. Pulse-wave Doppler ultrasound measurements for umbilical arteries and ascending aorta were obtained post-gavage (GD12, GD14, GD18) on 2 fetuses/litter. RNA from the non-decidual (labyrinthine and junctional zone) portion of placentas was isolated and processed for RNA-seq and subsequent bioinformatic analyses, and the association between transcriptomic changes and fetal phenotypes assessed. RESULTS: Exposure to ethanol in pregnant mice on GD10 attenuates umbilical cord blood flow transiently during gestation, and is associated with indices of IUGR, specifically decreased fetal weight and morphometric indices of cranial growth. Moreover, RNA-seq of the fetal portion of the placenta demonstrated that this single exposure has lasting transcriptomic changes, including upregulation of Tet3, which is associated with spontaneous abortion. Weighted gene co-expression network analysis (WGCNA) identified erythrocyte differentiation and homeostasis as important pathways associated with improved umbilical cord blood flow as gestation progresses. WGCNA also identified sensory perception of chemical stimulus/odorant and receptor activity as important pathways associated with cranial growth. CONCLUSION: Our data suggest that PAE perturbs the expression of placental genes relevant for placental hematopoiesis and environmental sensing, resulting in transient impairment of umbilical cord blood flow and, subsequently, IUGR.


Asunto(s)
Placenta , Efectos Tardíos de la Exposición Prenatal , Animales , Etanol/efectos adversos , Etanol/metabolismo , Femenino , Sangre Fetal/metabolismo , Retardo del Crecimiento Fetal/inducido químicamente , Humanos , Ratones , Placenta/metabolismo , Embarazo , Resultado del Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Transcriptoma
17.
BJOG ; 129(11): 1817-1831, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35352868

RESUMEN

BACKGROUND: The efficacy and safety profile of phosphodiesterase-5 inhibitors (PDE-5i) in pregnancy are unclear from the few relatively small diverse studies that have used them. OBJECTIVE: To assess the safety profile and clinical outcomes of PDE-5i use in pregnancy. SEARCH STRATEGY: We searched Embase, PubMed, CENTRAL, Prospero and Google Scholar to identify randomised controlled trials (RCTs) reporting the use of any PDE-5i in pregnancy up to September 2021. SELECTION CRITERIA: RCTs reporting obstetric or perinatal outcomes or maternal adverse outcomes in women taking PDE5i in pregnancy. DATA COLLECTION AND ANALYSIS: Risk ratios (RR), 95% confidence intervals (95% CI) and 95% prediction intervals were calculated and pooled for analysis. RESULTS: We identified 1324 citations, of which 10 studies including 1090 participants met the inclusion criteria. Only tadalafil and sildenafil were reported as used in pregnancy. Two studies using tadalafil and eight sildenafil. Nine of ten studies were assessed at having of low risk of bias. PDE-5i use was associated with an increased risk of headaches (RR 1.41, 95% CI 0.97-2.05), flushing (RR 2.59, 95% CI 0.69-9.90) and nasal bleeding (RR 10.53, 95% CI 1.36-81.3); an increase in vaginal birth when used for non-fetal growth restriction (FGR) indications (RR 1.24, 95% CI 1.00-1.55) and a reduction in risk of operative birth for intrapartum fetal compromise (RR 0.58, 95% CI 0.38-0.88). There was no evidence of any increase in risk of perinatal death (RR 0.89, 95% CI 0.56-1.43). However, use for the treatment of FGR increased the risk of persistent pulmonary hypertension of the newborn (PPHN) (RR 2.52, 95% CI 1.00-6.32). CONCLUSIONS: This meta-analysis suggests PDE-5i use in pregnancy is associated with mild maternal side effects and lower risk of operative birth for intrapartum fetal distress. Prolonged use for the treatment of FGR may increase the risk of PPHN. TWEETABLE ABSTRACT: PDE-5i use in pregnancy is associated with mild maternal side effects, lower operative birth for intrapartum fetal distress and a possible increase in persistent pulmonary hypertension of the newborn when used for the treatment of fetal growth restriction.


Asunto(s)
Sufrimiento Fetal , Hipertensión Pulmonar , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5 , Femenino , Retardo del Crecimiento Fetal/inducido químicamente , Humanos , Recién Nacido , Inhibidores de Fosfodiesterasa 5/efectos adversos , Embarazo , Citrato de Sildenafil/efectos adversos , Tadalafilo
18.
J Biochem Mol Toxicol ; 36(7): e23056, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35384129

RESUMEN

Cadmium (Cd) is an environmental pollutant and pregnant women are especially susceptible to the effects of exposure to Cd. Our previous study found Cd can be accumulated in the placenta and causes fetal growth restriction (FGR) through damage the placental glucocorticoid barrier. Selenium (Se), as an essential micronutrient, can allivate Cd-induced toxicity. In this study, we aim to explore the protective mechanism of Se against Cd-induced the placental glucocorticoid barrier damage and FGR. Pregnant Sprague Dawley (SD) rats were exposed to CdCl2 (1 mg/kg/day) and Na2 SeO3 (0.1-0.2-0.3 mg/kg/day) by gavage from gestational day (GD) 0 to GD 19. The results showed that reduced fetal weight, increased corticosterone concentrations in the maternal and fetal serum, and impaired placental labyrinth layer blood vessel development, appeared in pregnant rats after Cd exposure and improved after treated with Se. In cell experiments, we confirmed that Se reduces Cd-induced apoptosis. Moreover, Se can abolish Cd-induced 11ß-HSD2 and specificity protein 1 (Sp1) decreasing in vivo and vitro. In human JEG-3 cells, the knockdown of Sp1 expression by small interfering RNA can suppressed the protective effect of Se on Cd-induced 11ß-HSD2 decreasing. In general, our results demonstrated that Se is resistant to Cd-induced FGR through upregulating the placenta barrier via activation of the transcription factor Sp1.


Asunto(s)
Intoxicación por Cadmio , Selenio , Factor de Transcripción Sp1 , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/genética , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/metabolismo , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/farmacología , Animales , Cadmio/toxicidad , Intoxicación por Cadmio/metabolismo , Línea Celular Tumoral , Femenino , Retardo del Crecimiento Fetal/inducido químicamente , Retardo del Crecimiento Fetal/metabolismo , Glucocorticoides/farmacología , Humanos , Placenta/metabolismo , Embarazo , Ratas , Ratas Sprague-Dawley , Selenio/efectos adversos , Factor de Transcripción Sp1/biosíntesis
19.
Ecotoxicol Environ Saf ; 237: 113512, 2022 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-35429798

RESUMEN

BACKGROUND: Maternal exposure to ambient fine particulate matters (PM2.5) is associated with low birth weight (LBW) in offspring, but the underlying biological mechanisms are not yet fully understood. As the bridge that connects mother and fetus, the placenta plays a crucial role in fetal development by providing the fetus with nutrients and oxygen. However, whether PM2.5 exposure would impact the placental development and the related mechanisms are unclear. RESULTS: In the present study, female C57Bl/6j mice were exposed to filtered air (FA) or concentrated ambient PM2.5 (CAP) during pregestational and gestational periods, and the fetal development and placental structure were investigated. Our results showed that maternal exposure to CAP induced fetal growth restriction (FGR) and LBW. The placenta from CAP-exposed mice exhibited abnormal development including significant decrease of surface area, smaller junctional zone and impaired spiral artery remodeling. Meanwhile, CAP exposure altered trophoblast lineage differentiation and disrupted the balance between angiogenic and angiostatic factors in placenta. In addition, the inflammatory cytokines levels in lung, placenta and serum were significantly increased after ambient PM2.5 exposure. CONCLUSION: Our findings indicate that maternal exposure to PM2.5 disrupts normal structure and spiral artery remodeling of placenta and further induces FGR and LBW. This effect may be caused by the placental inflammation response subsequent to the pulmonary and systemic inflammation induced by ambient PM2.5 exposure.


Asunto(s)
Retardo del Crecimiento Fetal , Exposición Materna , Animales , Arterias , Femenino , Retardo del Crecimiento Fetal/inducido químicamente , Humanos , Inflamación , Exposición Materna/efectos adversos , Ratones , Ratones Endogámicos C57BL , Material Particulado/toxicidad , Placenta , Embarazo
20.
Ecotoxicol Environ Saf ; 229: 113103, 2022 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-34929501

RESUMEN

BACKGROUND: Cooking oil fume (COF) is an important source of indoor air pollution which severely affects human health, and sufficient vitamin D3 (VitD3) is necessary for maternal and child health. However, the effects of cooking oil fume-derived PM2.5 (COF-PM2.5) on birth outcomes and whether VitD3 could protect from adverse effects caused by COFs-PM2.5 are still unclear. METHODS: Twenty-four pregnant rats were divided into 4 groups and treated with various treatments: normal feeding, COFs-PM2.5 intratracheal instillation, VitD3 intragastric administration, and COFs-PM2.5 and VitD3 co-treatment, respectively. The fetal rats were obtained in pregnant 21 days and the development of them was recorded. Morphological changes in umbilical cord were measured with HE staining, and the oxidative stress and inflammatory levels were also investigated. Western blotting and RT-PCR was used to detect the expression of angiogenesis related factors. RESULTS: We successfully established an intrauterine growth restriction model in rats induced by COFs-PM2.5 where fetus weight significantly decreased after COFs-PM2.5 exposure. As for the umbilical cord vasculature, the wall thickened and the lumen narrowed down, and the contractility of the umbilical cord vasculature enhanced after COFs-PM2.5 exposure. COFs-PM2.5 exposure also increased the oxidative stress and inflammation level and activated the HIF-1α/eNOS/NO and VEGF/VEGFR2/eNOS signaling pathway. Interestingly, VitD3 intervention significantly increased the fetus weight and attenuated the injury of umbilical cord vascular, and partly or completely reversed the changes in the ROS/eNOS/ET-1 axis caused by COF-PM2.5. CONCLUSIONS: The findings of this study suggested that COF-PM2.5 exposure could contribute to intrauterine growth restriction through disturbing the ROS/eNOS/ET-1 axis, while VitD3 supplementation could be an effective prophylactic measurement.


Asunto(s)
Contaminación del Aire Interior , Material Particulado , Animales , Colecalciferol , Culinaria , Femenino , Retardo del Crecimiento Fetal/inducido químicamente , Retardo del Crecimiento Fetal/prevención & control , Material Particulado/toxicidad , Embarazo , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA