Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 805
Filtrar
1.
Front Public Health ; 12: 1351786, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38665245

RESUMEN

Recent evidence has revealed associations between endocrine-disrupting chemicals (EDCs) and placental insufficiency due to altered placental growth, syncytialization, and trophoblast invasion. However, no epidemiologic study has reported associations between exposure to EDCs and asymmetric fetal growth restriction (FGR) caused by placenta insufficiency. The aim of this study was to evaluate the association between EDC exposure and asymmetric FGR. This was a prospective cohort study including women admitted for delivery to the Maternal Fetal Center at Seoul St. Mary's Hospital between October 2021 and October 2022. Maternal urine and cord blood samples were collected, and the levels of bisphenol-A (BPA), monoethyl phthalates, and perfluorooctanoic acid in each specimen were analyzed. We investigated linear and non-linear associations between the levels of EDCs and fetal growth parameters, including the head circumference (HC)/abdominal circumference (AC) ratio as an asymmetric parameter. The levels of EDCs were compared between fetuses with and without asymmetric FGR. Of the EDCs, only the fetal levels of BPA showed a linear association with the HC/AC ratio after adjusting for confounding variables (ß = 0.003, p < 0.05). When comparing the normal growth and asymmetric FGR groups, the asymmetric FGR group showed significantly higher maternal and fetal BPA levels compared to the normal growth group (maternal urine BPA, 3.99 µg/g creatinine vs. 1.71 µg/g creatinine [p < 0.05]; cord blood BPA, 1.96 µg/L vs. -0.86 µg/L [p < 0.05]). In conclusion, fetal exposure levels of BPA show linear associations with asymmetric fetal growth patterns. High maternal and fetal exposure to BPA might be associated with asymmetric FGR.


Asunto(s)
Compuestos de Bencidrilo , Disruptores Endocrinos , Sangre Fetal , Retardo del Crecimiento Fetal , Exposición Materna , Fenoles , Humanos , Femenino , Disruptores Endocrinos/efectos adversos , Disruptores Endocrinos/sangre , Disruptores Endocrinos/orina , Estudios Prospectivos , Embarazo , Retardo del Crecimiento Fetal/inducido químicamente , Adulto , Compuestos de Bencidrilo/efectos adversos , Compuestos de Bencidrilo/orina , Compuestos de Bencidrilo/sangre , Fenoles/orina , Fenoles/efectos adversos , Fenoles/sangre , Exposición Materna/efectos adversos , Sangre Fetal/química , Fluorocarburos/sangre , Fluorocarburos/efectos adversos , Ácidos Ftálicos/orina , Ácidos Ftálicos/efectos adversos , Caprilatos/sangre , Caprilatos/efectos adversos , Insuficiencia Placentaria , República de Corea/epidemiología , Seúl/epidemiología
2.
Microbiome ; 12(1): 28, 2024 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-38365714

RESUMEN

BACKGROUND: Bisphenol A (BPA) is an environmental contaminant with endocrine-disrupting properties that induce fetal growth restriction (FGR). Previous studies on pregnant ewes revealed that BPA exposure causes placental apoptosis and oxidative stress (OS) and decreases placental efficiency, consequently leading to FGR. Nonetheless, the response of gut microbiota to BPA exposure and its role in aggravating BPA-mediated apoptosis, autophagy, mitochondrial dysfunction, endoplasmic reticulum stress (ERS), and OS of the maternal placenta and intestine are unclear in an ovine model of gestation. RESULTS: Two pregnant ewe groups (n = 8/group) were given either a subcutaneous (sc) injection of corn oil (CON group) or BPA (5 mg/kg/day) dissolved in corn oil (BPA group) once daily, from day 40 to day 110 of gestation. The maternal colonic digesta and the ileum and placental tissue samples were collected to measure the biomarkers of autophagy, apoptosis, mitochondrial dysfunction, ERS, and OS. To investigate the link between gut microbiota and the BPA-induced FGR in pregnant ewes, gut microbiota transplantation (GMT) was conducted in two pregnant mice groups (n = 10/group) from day 0 to day 18 of gestation after removing their intestinal microbiota by antibiotics. The results indicated that BPA aggravates apoptosis, ERS and autophagy, mitochondrial function injury of the placenta and ileum, and gut microbiota dysbiosis in pregnant ewes. GMT indicated that BPA-induced ERS, autophagy, and apoptosis in the ileum and placenta are attributed to gut microbiota dysbiosis resulting from BPA exposure. CONCLUSIONS: Our findings indicate the underlying role of gut microbiota dysbiosis and gut-placental axis behind the BPA-mediated maternal intestinal and placental apoptosis, OS, and FGR. The findings further provide novel insights into modulating the balance of gut microbiota through medication or probiotics, functioning via the gut-placental axis, to alleviate gut-derived placental impairment or FGR. Video Abstract.


Asunto(s)
Compuestos de Bencidrilo , Microbioma Gastrointestinal , Enfermedades Mitocondriales , Fenoles , Humanos , Embarazo , Ovinos , Femenino , Animales , Ratones , Placenta , Retardo del Crecimiento Fetal/inducido químicamente , Retardo del Crecimiento Fetal/metabolismo , Disbiosis/inducido químicamente , Disbiosis/metabolismo , Aceite de Maíz/metabolismo , Estrés Oxidativo , Enfermedades Mitocondriales/metabolismo
3.
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
4.
Chem Biol Interact ; 387: 110812, 2024 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-37993079

RESUMEN

Fetal growth restriction (FGR) is a prevalent complication in obstetrics, yet its exact aetiology remains unknown. Numerous studies suggest that the degradation of the living environment is a significant risk factor for FGR. 1-Nitropyrene (1-NP) is a widespread environmental pollutant as a representative substance of nitro-polycyclic aromatic hydrocarbons. In this study, we revealed that 1-NP induced FGR in fetal mice by constructing 1-NP exposed pregnant mice models. Intriguingly, we found that placental trophoblasts of 1-NP exposed mice exhibited significant ferroptosis, which was similarly detected in placental trophoblasts from human FGR patients. In this regard, we established a 1-NP exposed cell model in vitro using two human trophoblast cell lines, HTR8/SVneo and JEG-3. We found that 1-NP not only impaired the proliferation, migration, invasion and angiogenesis of trophoblasts, but also induced severe cellular ferroptosis. Meanwhile, the ferroptosis inhibitor ferrostatin-1 (Fer-1) effectively rescued 1-NP-induced trophoblast biological function impairment. Mechanistically, we revealed that 1-NP regulated ferroptosis by activating the ERK signaling pathway. Moreover, we innovatively revealed that CYP1B1 was essential for the activation of ERK signaling pathway induced by 1-NP. Overall, our study innovatively identified ferroptosis as a significant contributor to 1-NP induced trophoblastic functional impairment leading to FGR and clarified the specific mechanism by which 1-NP induced ferroptosis via the CYP1B1/ERK signaling pathway. Our study provided novel insights into the aetiology of FGR and revealed new mechanisms of reproductive toxicity of environmental pollutants.


Asunto(s)
Ferroptosis , Placenta , Pirenos , Animales , Femenino , Humanos , Ratones , Embarazo , Línea Celular Tumoral , Citocromo P-450 CYP1B1/metabolismo , Retardo del Crecimiento Fetal/inducido químicamente , Retardo del Crecimiento Fetal/metabolismo , Placenta/efectos de los fármacos , Placenta/patología , Transducción de Señal , Trofoblastos/metabolismo , Pirenos/toxicidad
5.
Reprod Sci ; 31(4): 997-1005, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37973775

RESUMEN

Tumor necrosis factor-α (TNF-α) antagonists are highly effective in controlling autoimmune diseases. This has led to speculation that they might also be useful in treating inflammatory placental conditions, such as chronic villitis of unknown etiology (VUE). VUE affects 10-15% of term placentas and is associated with recurrent fetal growth restriction (FGR) and pregnancy loss. We aimed to evaluate outcomes in patients with autoimmune diseases with and without anti-TNF-α biologic exposure during gestation. This retrospective cohort study compared pregnant women with autoimmune disease taking anti-TNF-α biologics (n = 89) to pregnant women with autoimmune disease but not taking a biologic (n = 53). We extracted data on all patients meeting our inclusion criteria over a 20-year period. Our primary outcome was the diagnosis of VUE by histology. Our secondary outcomes were maternal and neonatal complications such as preeclampsia, FGR, and neonatal intensive care admission. Kruskal-Wallis and chi-squared tests were performed as appropriate for statistical analysis. Maternal characteristics were comparable between groups, and there was no increase in adverse pregnancy outcomes based on anti-TNF-α treatment. Exposure to anti-TNF-α therapy had no significant effect on the incidence of VUE or other obstetric complications. Within the cohort exposed to anti-TNF-α biologics during pregnancy, the rate of VUE was 9.3%, which is comparable to the reported general population risk. Our data support the safety profile of biologic use in pregnancy.


Asunto(s)
Enfermedades Autoinmunes , Productos Biológicos , Corioamnionitis , Enfermedades Placentarias , Recién Nacido , Humanos , Embarazo , Femenino , Placenta/patología , Inhibidores del Factor de Necrosis Tumoral/efectos adversos , Enfermedades Placentarias/diagnóstico , Vellosidades Coriónicas/patología , Estudios Retrospectivos , Resultado del Embarazo , Retardo del Crecimiento Fetal/inducido químicamente , Retardo del Crecimiento Fetal/patología , Enfermedades Autoinmunes/diagnóstico , Enfermedades Autoinmunes/tratamiento farmacológico , Enfermedades Autoinmunes/complicaciones , Productos Biológicos/efectos adversos
6.
Reprod Sci ; 31(3): 779-790, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37845590

RESUMEN

Fetal growth restriction (FGR) seriously threatens perinatal health. The main cause of FGR is placental malperfusion, but the specific mechanism is still unclear, and there is no effective treatment for FGR. We constructed a FGR mouse model by adding exogenous asymmetric dimethylarginine (ADMA) through in vivo experiments and found that ADMA could cause placental dysplasia and induce the occurrence of FGR. Compared with the control group, reactive oxygen species (ROS) production in the placenta was increased in mice with FGR, and the expression of autophagy-related proteins p-AKT/AKT, p-mTOR/mTOR, and P62 was significantly decreased, while the expression of Beclin-1 and LC3-II was significantly increased in the FGR group. Furthermore, ADMA had a favorable effect in promoting the formation of autophagosomes. Hydroxychloroquine (HCQ) and N-acetylcysteine (NAC) improved ADMA-induced disorders of placental development and alleviated ADMA-induced FGR. This study found that ADMA could cause excessive autophagy of trophoblasts by increasing the level of oxidative stress, ultimately leading to the occurrence of FGR, and HCQ and NAC had therapeutic effects on ADMA-induced FGR.


Asunto(s)
Acetilcisteína , Arginina/análogos & derivados , Placenta , Humanos , Embarazo , Ratones , Femenino , Animales , Placenta/metabolismo , Acetilcisteína/farmacología , Acetilcisteína/uso terapéutico , Acetilcisteína/metabolismo , Retardo del Crecimiento Fetal/inducido químicamente , Retardo del Crecimiento Fetal/tratamiento farmacológico , Retardo del Crecimiento Fetal/metabolismo , Hidroxicloroquina/farmacología , Hidroxicloroquina/uso terapéutico , Hidroxicloroquina/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Estrés Oxidativo , Serina-Treonina Quinasas TOR/metabolismo , Autofagia
7.
Free Radic Biol Med ; 211: 127-144, 2024 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-38103660

RESUMEN

Exposure to bisphenol A (BPA) during gestation leads to fetal growth restriction (FGR), whereby the underlying mechanisms remain unknown. Here, we found that FGR patients showed higher levels of BPA in the urine, serum, and placenta; meanwhile, trophoblast ferroptosis was observed in FGR placentas, as indicated by accumulated intracellular iron, impaired antioxidant molecules, and increased lipid peroxidation products. To investigate the role of ferroptosis in placental and fetal growth, BPA stimulation was performed both in vivo and in vitro. BPA exposure during gestation was associated with FGR in mice; also, it induces ferroptosis in mouse placentas and human placental trophoblast. Pretreatment with ferroptosis inhibitor ferritin-1 (Fer-1) alleviated BPA-induced oxidative damage and cell death. Notably, BPA reduced the trophoblastic expression of Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ), which regulated tissue growth and organ size. YAP or TAZ siRNA enhanced BPA-induced ferroptosis, suggesting that trophoblast ferroptosis is dependent on YAP/TAZ downregulation after BPA stimulation. Consistently, the protein levels of YAP/TAZ were also reduced in FGR placentas. Further results revealed that silencing YAP/TAZ promoted BPA-induced ferroptosis through autophagy. Pretreatment with autophagy inhibitor chloroquine (CQ) attenuated BPA-induced trophoblast ferroptosis. Ferritinophagy, an autophagic degradation of ferritin (FTH1), was observed in FGR placentas. Similarly, BPA reduced the protein level of FTH1 in placental trophoblast. Pretreatment with iron chelator desferrioxamine (DFO) and NCOA4 (an autophagy cargo receptor) siRNA weakened the ferroptosis of trophoblast after exposure to BPA, indicating that autophagy mediates ferroptosis in BPA-stimulated trophoblast by degrading ferritin. In summary, ferroptosis was featured in BPA-associated FGR and trophoblast injury; the regulation of ferroptosis involved the YAP/TAZ-autophagy-ferritin axis.


Asunto(s)
Ferroptosis , Placenta , Humanos , Embarazo , Femenino , Animales , Ratones , Placenta/metabolismo , Retardo del Crecimiento Fetal/inducido químicamente , Retardo del Crecimiento Fetal/metabolismo , Ferritinas/genética , Ferritinas/metabolismo , ARN Interferente Pequeño/metabolismo
8.
Microbiome ; 11(1): 245, 2023 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-37932832

RESUMEN

BACKGROUND: Autism spectrum disorder (ASD) has been associated with intrauterine growth restriction (IUGR), but the underlying mechanisms are unclear. RESULTS: We found that the IUGR rat model induced by prenatal caffeine exposure (PCE) showed ASD-like symptoms, accompanied by altered gut microbiota and reduced production of indole 3-propionic acid (IPA), a microbiota-specific metabolite and a ligand of aryl hydrocarbon receptor (AHR). IUGR children also had a reduced serum IPA level consistent with the animal model. We demonstrated that the dysregulated IPA/AHR/NF-κB signaling caused by disturbed gut microbiota mediated the hippocampal microglia hyperactivation and neuronal synapse over-pruning in the PCE-induced IUGR rats. Moreover, postnatal IPA supplementation restored the ASD-like symptoms and the underlying hippocampal lesions in the IUGR rats. CONCLUSIONS: This study suggests that the microbiota-IPA-brain axis regulates ASD susceptibility in PCE-induced IUGR offspring, and supplementation of microbiota-derived IPA might be a promising interventional strategy for ASD with a fetal origin. Video Abstract.


Asunto(s)
Trastorno del Espectro Autista , Microbioma Gastrointestinal , Animales , Femenino , Embarazo , Ratas , Encéfalo , Cafeína/toxicidad , Retardo del Crecimiento Fetal/inducido químicamente , Microbioma Gastrointestinal/fisiología , Hipocampo , Microglía , Plasticidad Neuronal
9.
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
10.
Environ Sci Pollut Res Int ; 30(51): 110956-110969, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37798517

RESUMEN

Di-(2-ethylhexyl)-phthalate (DEHP) is a ubiquitous environmental pollutant and is widely used in industrial plastics. Intrahepatic cholestasis of pregnancy (ICP), distinguished by maternal pruritus and elevated serum bile acid levels, is linked to unfavorable pregnancy consequences. Few studies have investigated the potential effect of gestational DEHP exposure on the cholestasis in pregnant female mice, and the underlying mechanisms remain unclear. In the present study, a mouse model of cholestasis during pregnancy was established by DEHP exposure. We found that DEHP induces elevated bile acid levels by affecting bile acid synthesis and transporter receptor expression in the maternal liver and placenta of pregnant female mice, ultimately leading to intrauterine growth restriction (IUGR). In addition, DEHP changed the bile acid composition of maternal serum and liver as well as placenta and amniotic fluid in pregnant female mice; Importantly, we found that DEHP down-regulates the expression of farnesoid X receptor (FXR), which is considered to be a bile acid receptor. FXR agonist obeticholic acid (OCA) effectively alleviated the adverse effects of DEHP on pregnant female mice. While, OCA itself had no adverse effects on normal pregnant female mice. In summary, DEHP could induces bile acid disorder and IUGR in pregnant female mice by affect FXR, which was reversed by OCA.


Asunto(s)
Colestasis , Dietilhexil Ftalato , Embarazo , Humanos , Femenino , Animales , Ratones , Ácidos y Sales Biliares/toxicidad , Retardo del Crecimiento Fetal/inducido químicamente , Retardo del Crecimiento Fetal/metabolismo , Dietilhexil Ftalato/toxicidad
11.
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
12.
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
13.
Chemosphere ; 336: 139217, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37336441

RESUMEN

BACKGROUND: Fetal growth restriction (FGR) is a major determinant of perinatal morbidity and mortality, with adverse long-term neurocognitive effects in childhood and adulthood. Prenatal exposure to environmental pollutants has been reported to be associated with FGR. Neonicotinoids (NEOs) are extensively used insecticides worldwide and are suggested to have embryonic and developmental neurotoxicity. However, the effects of NEOs exposure on FGR is unknown. OBJECTIVES: We aimed to quantify the single and combined associations of maternal exposure to NEOs and FGR. METHODS: We conducted a nested case-control study based on the Guangxi Zhuang Birth Cohort, China. A total of 387 with FGR cases and 1096 without- FGR controls were included between 2015 and 2018. Ten NEOs were measured by UPLC-MS from the maternal blood samples were pre-collected in the first trimester. After adjusting for potential confounders, multivariable logistic regressions, weighted quantile sum regression and quantile g-computation were performed for individual and NEOs mixtures. RESULTS: In the individual exposure models, each 1-standard deviation increment of the natural-log in dinotefuran and acetamiprid concentrations were significantly associated with odds ratios of 1.93 (95% CI: 1.69, 2.20) and 1.31 (95% CI: 1.07, 1.59) higher odds of FGR, respectively. However, the FGR risk was negatively associated with thiacloprid, sulfoxaflor, and nitenpyram (OR = 0.23, 95%CI: 0.15, 0.34; OR = 0.48, 95%CI: 0.41, 0.56; OR = 0.86, 95%CI: 0.80, 0.93; respectively). Similar findings were found in the combined exposure analysis. Dinotefuran was the most strongly attributable to increase FGR, while sulfoxaflor and thiacloprid contributed the highest negative weighted on FGR. Furthermore, each quintile increase in all ten NEOs exposures was associated with FGR (OR = 0.21, 95% CI: 0.08, 0.54). CONCLUSION: Our findings suggest that maternal single and combined exposures to NEOs were associated with varying FGR risks. They contribute to the mounting evidence on serum NEOs exposure impact on FGR. However, a replication of these associations in other populations is warranted.


Asunto(s)
Insecticidas , Embarazo , Femenino , Humanos , Insecticidas/toxicidad , Insecticidas/análisis , Retardo del Crecimiento Fetal/inducido químicamente , Retardo del Crecimiento Fetal/epidemiología , Exposición Materna , Cohorte de Nacimiento , Estudios de Casos y Controles , Cromatografía Liquida , China/epidemiología , Espectrometría de Masas en Tándem , Neonicotinoides/análisis
14.
Environ Pollut ; 328: 121602, 2023 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-37031847

RESUMEN

Cadmium (Cd), an environmental contaminant, can result in placental non-selective autophagy activation and fetal growth restriction (FGR). However, the role of placental lipophagy, a selective autophagy, in Cd-induced FGR is unclear. This work uses case-control study, animal experiments and cultures of primary human placental trophoblast cells to explore the role of placental lipophagy in Cd-induced FGR. We found association of placental lipophagy and all-cause FGR. Meanwhile, pregnancy Cd exposure induced FGR and placental lipophgay. Inhibition of placental lipophagy by pharmacological and genetic means (Atg5-/- mice) exacerbated Cd-caused FGR. Inversely, activating of placental lipophagy relieved Cd-stimulated FGR. Subsequently, we found that activation of Atg5-dependent lipophagy degrades lipid droplets to produce free cholesterol, and promotes placental progesterone (P4) synthesis. Gestational P4 supplementation significantly reversed Cd-induced FGR. Altogether, activation of Atg5-dependent placental lipophagy ameliorates Cd-induced FGR.


Asunto(s)
Cadmio , Placenta , Embarazo , Femenino , Humanos , Animales , Ratones , Placenta/metabolismo , Cadmio/toxicidad , Cadmio/metabolismo , Retardo del Crecimiento Fetal/inducido químicamente , Estudios de Casos y Controles , Autofagia , Proteína 5 Relacionada con la Autofagia/genética , Proteína 5 Relacionada con la Autofagia/metabolismo
15.
Environ Sci Pollut Res Int ; 30(21): 60805-60819, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37037937

RESUMEN

Bisphenol-A (BPA) is a common environmental toxicant that is known to be associated with fetal growth restriction (FGR). However, the mechanisms of how BPA induce FGR is poorly characterized. We conducted proteomics to identify the abnormal expression of SRB1 in female placental tissues with high BPA-induced FGR and further verified its decreased expression in human placenta and BeWo cells. Next, the effect of BPA on fetal development was further confirmed in pregnant C57BL/6 mice. The expression of SRB1 was consistently downregulated in human FGR placentas, BPA-exposed trophoblasts and mouse placentas. In addition, we found that SRB1 interacted with PCNA, and BPA exposure indirectly reduced the expression of PCNA and further inhibited placental proliferation. In vitro studies showed that BPA exposure reduced the expression of CDK1, CDK2, cyclin B and phosphorylated Rb in placental trophoblast cells, indicating cell cycle arrest after exposure to BPA. In addition, the expression of γ-H2AX and phosphorylated ATM was upregulated in BPA-exposed trophoblasts, indicating increased DNA damage. Our results indicate that BPA-induced FGR is achieved by reducing the expression of SRB1, inhibiting placental proliferation and increasing DNA damage. Our findings not only explain the mechanism of BPA-associated developmental toxicity but also shed light upon developing novel therapeutic targets.


Asunto(s)
Placenta , Trofoblastos , Animales , Ratones , Embarazo , Femenino , Humanos , Placenta/metabolismo , Trofoblastos/metabolismo , Retardo del Crecimiento Fetal/inducido químicamente , Retardo del Crecimiento Fetal/genética , Retardo del Crecimiento Fetal/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Antígeno Nuclear de Célula en Proliferación/farmacología , Ratones Endogámicos C57BL , Proliferación Celular
16.
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
17.
J Nutr Biochem ; 118: 109359, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37085060

RESUMEN

Lipopolysaccharide (LPS) could induce adverse birth outcomes by evoking inflammation. We investigated the effect and mechanism of docosahexaenoic acid (DHA) on LPS-induced placental inflammation and fetal growth restriction (FGR). In vivo, pregnant CD-1 mice were divided into four groups: Ctrl, DHA, LPS and DHA+LPS group. We found that DHA pretreatment reduced the incidence of FGR induced by LPS and activated the expression of peroxisome proliferators-activated receptor gamma (PPARγ) in placental tissue. Moreover, the LPS-induced increase of mRNA levels of Tnf-α, Il-6, Il-1ß, Mip-2 and Kc in placental tissue was significantly attenuated by DHA pretreatment. A similar effect of DHA was observed in serum of pregnant mice and amniotic fluid. In contrast, the levels of the IL-10 were significantly increased after DHA pretreatment. In vitro, we clarified that DHA antagonized the activation of the NF-κB signaling pathway induced by LPS, which was dependent on PPARγ. Subsequently, CHX (translation inhibitor) was used to indicated that PPARγ significantly increased the degradation rate of p65, an effect that was inhibited by MG132 (proteasome inhibitor) treatment. Finally, it was confirmed that the activation of PPARγ could significantly promote the ubiquitination and degradation of p65. Our results suggested that DHA alleviated LPS-induced inflammatory responses and FGR by activating PPARγ expression, leading to p65 ubiquitination and degradation.


Asunto(s)
FN-kappa B , Placenta , Humanos , Femenino , Embarazo , Animales , Ratones , FN-kappa B/metabolismo , Placenta/metabolismo , Lipopolisacáridos/metabolismo , Trofoblastos/metabolismo , Ácidos Docosahexaenoicos/farmacología , Ácidos Docosahexaenoicos/metabolismo , PPAR gamma/genética , PPAR gamma/metabolismo , Retardo del Crecimiento Fetal/inducido químicamente , Inflamación/metabolismo
18.
Alcohol ; 110: 65-81, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-36898643

RESUMEN

INTRODUCTION: Prenatal alcohol exposure can impair placentation and cause intrauterine growth restriction (IUGR), fetal demise, and fetal alcohol spectrum disorder (FASD). Previous studies showed that ethanol's inhibition of placental insulin and insulin-like growth factor, type 1 (IGF-1) signaling compromises trophoblastic cell motility and maternal vascular transformation at the implantation site. Since soy isolate supports insulin responsiveness, we hypothesized that dietary soy could be used to normalize placentation and fetal growth in an experimental model of FASD. METHODS: Pregnant Long-Evans rat dams were fed with isocaloric liquid diets containing 0% or 8.2% ethanol (v/v) from gestation day (GD) 6. Dietary protein sources were either 100% soy isolate or 100% casein (standard). Gestational sacs were harvested on GD19 to evaluate fetal resorption, fetal growth parameters, and placental morphology. Placental insulin/IGF-1 signaling through Akt pathways was assessed using commercial bead-based multiplex enzyme-linked immunosorbent assays. RESULTS: Dietary soy markedly reduced or prevented the ethanol-associated fetal loss, IUGR, FASD dysmorphic features, and impairments in placentation/maturation. Furthermore, ethanol's inhibitory effects on the placental glycogen cell population at the junctional zone, invasive trophoblast populations at the implantation site, maternal vascular transformation, and signaling through the insulin and IGF1 receptors, Akt and PRAS40 were largely abrogated by co-administration of soy. CONCLUSION: Dietary soy may provide an economically feasible and accessible means of reducing adverse pregnancy outcomes linked to gestational ethanol exposure.


Asunto(s)
Trastornos del Espectro Alcohólico Fetal , Efectos Tardíos de la Exposición Prenatal , Ratas , Animales , Humanos , Embarazo , Femenino , Placentación , Placenta/metabolismo , Insulina/metabolismo , Retardo del Crecimiento Fetal/inducido químicamente , Retardo del Crecimiento Fetal/prevención & control , Factor I del Crecimiento Similar a la Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/farmacología , Trastornos del Espectro Alcohólico Fetal/prevención & control , Trastornos del Espectro Alcohólico Fetal/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas Long-Evans , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Etanol/efectos adversos , Muerte Fetal , Dieta
19.
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
20.
Environ Int ; 172: 107791, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36739855

RESUMEN

BACKGROUND: Epidemiological studies suggest that both ambient ozone (O3) and temperature were associated with increased risks of adverse birth outcomes. However, very few studies explored their interaction effects, especially for small for gestational age (SGA) and large for gestational age (LGA). OBJECTIVES: To estimate the modification effects of ambient temperature on associations of ambient O3 exposure before and during pregnancy with preterm birth (PTB), low birth weight (LBW), SGA and LGA based on multicity birth cohorts. METHODS: A total of 56,905 singleton pregnant women from three birth cohorts conducted in Tianjin, Beijing and Maoming, China, were included in the study. Maximum daily 8-h average O3 concentrations of each pregnant woman from the preconception period to delivery for every day were estimated by matching their home addresses with the Tracking Air Pollution in China (TAP) datasets. We first applied the Cox proportional-hazards regression model to evaluate the city-specific effects of O3 exposure before and during pregnancy on adverse birth outcomes at different temperature levels with adjustment for potential confounders, and then a meta-analysis across three birth cohorts was conducted to calculate the pooled associations. RESULTS: In pooled analysis, significant modification effects of ambient temperature on associations of ambient O3 with PTB, LBW and LGA were observed (Pinteraction < 0.05). For a 10 µg/m3 increase in ambient O3 exposure at high temperature level (> 75th percentile), the risk of LBW increased by 28 % (HR: 1.28, 95% CI: 1.13-1.46) during the second trimester and the risk of LGA increased by 116% (HR: 2.16, 95%CI: 1.16-4.00) during the entire pregnancy, while the null or weaker association was observed at corresponding low (≤ 25th percentile) and medium (> 25th and ≤ 75th percentile) temperature levels. CONCLUSION: This multicity study added new evidence that ambient high temperature may enhance the potential effects of ambient O3 on adverse birth outcomes.


Asunto(s)
Contaminantes Atmosféricos , Contaminación del Aire , Ozono , Complicaciones del Embarazo , Nacimiento Prematuro , Recién Nacido , Embarazo , Humanos , Femenino , Contaminantes Atmosféricos/análisis , Nacimiento Prematuro/epidemiología , Nacimiento Prematuro/inducido químicamente , Temperatura , Contaminación del Aire/efectos adversos , Contaminación del Aire/análisis , Ozono/análisis , Complicaciones del Embarazo/inducido químicamente , China/epidemiología , Retardo del Crecimiento Fetal/inducido químicamente , Exposición Materna/efectos adversos , Material Particulado/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA