Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34.826
Filtrar
1.
Lancet Planet Health ; 8(5): e297-e308, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38723642

RESUMEN

BACKGROUND: Pregnancy air pollution exposure (PAPE) has been linked to a wide range of adverse birth and childhood outcomes, but there is a paucity of data on its influence on the placental epigenome, which can regulate the programming of physiological functions and affect child development. This study aimed to investigate the association between prenatal air pollutant exposure concentrations and changes in placental DNA methylation patterns, and to explore the potential windows of susceptibility and sex-specific alterations. METHODS: This multi-site study used three prospective population-based mother-child cohorts: EDEN, PELAGIE, and SEPAGES, originating from four French geographical regions (Nancy, Poitiers, Brittany, and Grenoble). Pregnant women were included between 2003 and 2006 for EDEN and PELAGIE, and between 2014 and 2017 for SEPAGES. The main eligibility criteria were: being older than 18 years, having a singleton pregnancy, and living and planning to deliver in one of the maternity clinics in one of the study areas. A total of 1539 mother-child pairs were analysed, measuring placental DNA methylation using Illumina BeadChips. We used validated spatiotemporally resolved models to estimate PM2·5, PM10, and NO2 exposure over each trimester of pregnancy at the maternal residential address. We conducted a pooled adjusted epigenome-wide association study to identify differentially methylated 5'-C-phosphate-G-3' (CpG) sites and regions (assessed using the Infinium HumanMethylationEPIC BeadChip array, n=871), including sex-specific and sex-linked alterations, and independently validated our results (assessed using the Infinium HumanMethylation450 BeadChip array, n=668). FINDINGS: We identified four CpGs and 28 regions associated with PAPE in the total population, 469 CpGs and 87 regions in male infants, and 150 CpGs and 66 regions in female infants. We validated 35% of the CpGs available. More than 30% of the identified CpGs were related to one (or more) birth outcome and most significant alterations were enriched for neural development, immunity, and metabolism related genes. The 28 regions identified for both sexes overlapped with imprinted genes (four genes), and were associated with neurodevelopment (nine genes), immune system (seven genes), and metabolism (five genes). Most associations were observed for the third trimester for female infants (134 of 150 CpGs), and throughout pregnancy (281 of 469 CpGs) and the first trimester (237 of 469 CpGs) for male infants. INTERPRETATION: These findings highlight the molecular pathways through which PAPE might affect child health in a widespread and sex-specific manner, identifying the genes involved in the major physiological functions of a developing child. Further studies are needed to elucidate whether these epigenetic changes persist and affect health later in life. FUNDING: French Agency for National Research, Fondation pour la Recherche Médicale, Fondation de France, and the Plan Cancer.


Asunto(s)
Contaminantes Atmosféricos , Contaminación del Aire , Metilación de ADN , Exposición Materna , Placenta , Humanos , Femenino , Embarazo , Placenta/efectos de los fármacos , Placenta/metabolismo , Estudios Prospectivos , Exposición Materna/efectos adversos , Adulto , Contaminación del Aire/efectos adversos , Masculino , Contaminantes Atmosféricos/efectos adversos , Contaminantes Atmosféricos/análisis , Francia , Efectos Tardíos de la Exposición Prenatal/genética , Resultado del Embarazo , Recién Nacido , Adulto Joven
2.
Environ Health Perspect ; 132(5): 57002, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38728218

RESUMEN

BACKGROUND: Endocrine-disrupting chemicals may play a role in adiposity development during childhood. Until now literature in this scope suffers from methodologic limitations in exposure assessment using one or few urine samples and missing assessment during the infancy period. OBJECTIVES: We investigated the associations between early-life exposure to quickly metabolized chemicals and post-natal growth, relying on repeated within-subject urine collections over pregnancy and infancy. METHODS: We studied the associations of four phenols, four parabens, seven phthalates, and one nonphthalate plasticizer from weekly pooled urine samples collected from the mother during second and third trimesters (median 18 and 34 gestational weeks, respectively) and infant at 2 and 12 months of age, and child growth until 36 months. We relied on repeated measures of height, weight and head circumference from study visits and the child health booklet to predict growth outcomes at 3 and 36 months using the Jenss-Bayley nonlinear mixed model. We assessed associations with individual chemicals using adjusted linear regression and mixtures of chemicals using a Bayesian kernel machine regression model. RESULTS: The unipollutant analysis revealed few associations. Bisphenol S (BPS) at second trimester was positively associated with all infant growth parameters at 3 and 36 months, with similar patterns between exposure at third trimester and all infant growth parameters at 3 months. Mono-n-butyl phthalate (MnBP) at 12 months was positively associated with body mass index (BMI), weight, and head circumference at 36 months. Mixture analysis revealed positive associations between exposure at 12 months and BMI and weight at 36 months, with MnBP showing the highest effect size within the mixture. CONCLUSIONS: This study suggests that exposure in early infancy may be associated with increased weight and BMI in early childhood, which are risk factors of obesity in later life. Furthermore, this study highlighted the impact of BPS, a compound replacing bisphenol A, which has never been studied in this context. https://doi.org/10.1289/EHP13644.


Asunto(s)
Disruptores Endocrinos , Parabenos , Fenoles , Ácidos Ftálicos , Efectos Tardíos de la Exposición Prenatal , Humanos , Ácidos Ftálicos/orina , Fenoles/orina , Fenoles/toxicidad , Femenino , Lactante , Embarazo , Disruptores Endocrinos/orina , Disruptores Endocrinos/toxicidad , Contaminantes Ambientales/orina , Masculino , Exposición Materna/estadística & datos numéricos , Exposición Materna/efectos adversos , Estudios Longitudinales , Preescolar , Antropometría
3.
Sci Rep ; 14(1): 10773, 2024 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-38730262

RESUMEN

The developing brain is vulnerable to maternal bacterial and viral infections which induce strong inflammatory responses in the mother that are mimicked in the offspring brain, resulting in irreversible neurodevelopmental defects, and associated cognitive and behavioural impairments. In contrast, infection during pregnancy and lactation with the immunoregulatory murine intestinal nematode, Heligmosomoides bakeri, upregulates expression of genes associated with long-term potentiation (LTP) of synaptic networks in the brain of neonatal uninfected offspring, and enhances spatial memory in uninfected juvenile offspring. As the hippocampus is involved in spatial navigation and sensitive to immune events during development, here we assessed hippocampal gene expression, LTP, and neuroimmunity in 3-week-old uninfected offspring born to H. bakeri infected mothers. Further, as maternal immunity shapes the developing immune system, we assessed the impact of maternal H. bakeri infection on the ability of offspring to resist direct infection. In response to maternal infection, we found an enhanced propensity to induce LTP at Schaffer collateral synapses, consistent with RNA-seq data indicating accelerated development of glutamatergic synapses in uninfected offspring, relative to those from uninfected mothers. Hippocampal RNA-seq analysis of offspring of infected mothers revealed increased expression of genes associated with neurogenesis, gliogenesis, and myelination. Furthermore, maternal infection improved resistance to direct infection of H. bakeri in offspring, correlated with transfer of parasite-specific IgG1 to their serum. Hippocampal immunohistochemistry and gene expression suggest Th2/Treg biased neuroimmunity in offspring, recapitulating peripheral immunoregulation of H. bakeri infected mothers. These findings indicate maternal H. bakeri infection during pregnancy and lactation alters peripheral and neural immunity in uninfected offspring, in a manner that accelerates neural maturation to promote hippocampal LTP, and upregulates the expression of genes associated with neurogenesis, gliogenesis, and myelination.


Asunto(s)
Hipocampo , Plasticidad Neuronal , Animales , Femenino , Hipocampo/metabolismo , Hipocampo/parasitología , Embarazo , Ratones , Infecciones por Nematodos/inmunología , Infecciones por Nematodos/parasitología , Potenciación a Largo Plazo , Efectos Tardíos de la Exposición Prenatal/inmunología , Infecciones por Strongylida/inmunología , Infecciones por Strongylida/parasitología , Masculino , Neuroinmunomodulación
4.
Int J Mol Sci ; 25(9)2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38731818

RESUMEN

Early life exposure lays the groundwork for the risk of developing cardiovascular-kidney-metabolic (CKM) syndrome in adulthood. Various environmental chemicals to which pregnant mothers are commonly exposed can disrupt fetal programming, leading to a wide range of CKM phenotypes. The aryl hydrocarbon receptor (AHR) has a key role as a ligand-activated transcription factor in sensing these environmental chemicals. Activating AHR through exposure to environmental chemicals has been documented for its adverse impacts on cardiovascular diseases, hypertension, diabetes, obesity, kidney disease, and non-alcoholic fatty liver disease, as evidenced by both epidemiological and animal studies. In this review, we compile current human evidence and findings from animal models that support the connection between antenatal chemical exposures and CKM programming, focusing particularly on AHR signaling. Additionally, we explore potential AHR modulators aimed at preventing CKM syndrome. As the pioneering review to present evidence advocating for the avoidance of toxic chemical exposure during pregnancy and deepening our understanding of AHR signaling, this has the potential to mitigate the global burden of CKM syndrome in the future.


Asunto(s)
Enfermedades Cardiovasculares , Efectos Tardíos de la Exposición Prenatal , Receptores de Hidrocarburo de Aril , Receptores de Hidrocarburo de Aril/metabolismo , Receptores de Hidrocarburo de Aril/genética , Humanos , Embarazo , Animales , Femenino , Efectos Tardíos de la Exposición Prenatal/metabolismo , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/inducido químicamente , Enfermedades Renales/inducido químicamente , Enfermedades Renales/metabolismo , Enfermedades Renales/etiología , Exposición Materna/efectos adversos , Transducción de Señal/efectos de los fármacos , Riñón/metabolismo , Riñón/efectos de los fármacos , Riñón/patología , Desarrollo Fetal/efectos de los fármacos , Contaminantes Ambientales/toxicidad , Contaminantes Ambientales/efectos adversos , Reprogramación Metabólica
5.
BMC Public Health ; 24(1): 1238, 2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38711042

RESUMEN

BACKGROUND: We conducted this meta-analysis to investigate the potential association between maternal smoking, alcohol and caffeinated beverages consumption during pregnancy and the risk of childhood brain tumors (CBTs). METHODS: A thorough search was carried out on PubMed, Embase, Web of Science, Cochrane Library, and China National Knowledge Internet to identify pertinent articles. Fixed or random effects model was applied to meta-analyze the data. RESULTS: The results suggested a borderline statistically significant increased risk of CBTs associated with maternal smoking during pregnancy (OR 1.04, 95% CI 0.99-1.09). We found that passive smoking (OR 1.12, 95% CI 1.03-1.20), rather than active smoking (OR 1.00, 95% CI 0.93-1.07), led to an increased risk of CBTs. The results suggested a higher risk in 0-1 year old children (OR 1.21, 95% CI 0.94-1.56), followed by 0-4 years old children (OR 1.12, 95% CI 0.97-1.28) and 5-9 years old children (OR 1.11, 95% CI 0.95-1.29). This meta-analysis found no significant association between maternal alcohol consumption during pregnancy and CBTs risk (OR 1.00, 95% CI 0.80-1.24). An increased risk of CBTs was found to be associated with maternal consumption of caffeinated beverages (OR 1.16, 95% CI 1.07-1.26) during pregnancy, especially coffee (OR 1.18, 95% CI 1.00-1.38). CONCLUSIONS: Maternal passive smoking, consumption of caffeinated beverages during pregnancy should be considered as risk factors for CBTs, especially glioma. More prospective cohort studies are warranted to provide a higher level of evidence.


Asunto(s)
Consumo de Bebidas Alcohólicas , Neoplasias Encefálicas , Cafeína , Estudios Observacionales como Asunto , Efectos Tardíos de la Exposición Prenatal , Humanos , Embarazo , Femenino , Consumo de Bebidas Alcohólicas/efectos adversos , Consumo de Bebidas Alcohólicas/epidemiología , Efectos Tardíos de la Exposición Prenatal/epidemiología , Neoplasias Encefálicas/epidemiología , Neoplasias Encefálicas/inducido químicamente , Neoplasias Encefálicas/etiología , Niño , Preescolar , Cafeína/efectos adversos , Lactante , Recién Nacido , Fumar/epidemiología , Fumar/efectos adversos , Factores de Riesgo , Bebidas/efectos adversos
6.
An Acad Bras Cienc ; 96(2): e20240014, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38747842

RESUMEN

Despite the prevalence of substance use during pregnancy, studies focusing exclusively on Neonatal Intensive Care Units (NICU) admissions remain limited. This study investigates the impact of maternal use of tobacco, alcohol, and/or crack, on neonatal outcomes among infants admitted to three Brazilian NICUs. Additionally, the investigation explores the impact of substance use on DNA damage in newborns. Over a one-year period, data from 254 newborns were collected through medical records, accompanied by blood samples. Findings revealed that 16.1% of newborns had mothers reporting substance use during pregnancy. Significant associations were found between maternal substance use and adverse neonatal outcomes, including low birth weight, preterm birth, and sexually transmitted infections. Maternal variables linked to substance use encompassed non-white skin color, low education, non-masonry housing, lower income, diseases in other children, and fewer prenatal consultations. Notably, neonatal DNA damage showed no significant association with substance use. Our results underscore the substantial impact of maternal substance use on NICU-admitted infants, emphasizing the necessity for targeted interventions that address both neonatal health and maternal well-being, thereby underscoring the crucial role of comprehensive care in NICU settings.


Asunto(s)
Consumo de Bebidas Alcohólicas , Unidades de Cuidado Intensivo Neonatal , Humanos , Embarazo , Femenino , Recién Nacido , Brasil/epidemiología , Adulto , Consumo de Bebidas Alcohólicas/efectos adversos , Complicaciones del Embarazo , Masculino , Adulto Joven , Resultado del Embarazo , Recién Nacido de Bajo Peso , Cocaína Crack/efectos adversos , Trastornos Relacionados con Cocaína/epidemiología , Factores de Riesgo , Factores Socioeconómicos , Daño del ADN , Efectos Tardíos de la Exposición Prenatal
7.
J Agric Food Chem ; 72(19): 10679-10691, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38695770

RESUMEN

There has been a dramatic surge in the prevalence of food allergy (FA) that cannot be explained solely by genetics, identifying mechanisms of sensitization that are driven by environmental factors has become increasingly important. Diet, gut microbiota, and their metabolites have been shown to play an important role in the development of FA. In this review, we discuss the latest epidemiological evidence on the impact of two major dietary patterns and key nutrients in early life on the risk of offspring developing FA. The Western diet typically includes high sugar and high fat, which may affect the immune system of offspring and increase susceptibility to FA. In contrast, the Mediterranean diet is rich in fiber, which may reduce the risk of FA in offspring. Furthermore, we explore the potential mechanisms by which maternal dietary nutrients during a window of opportunity (pregnancy, birth, and lactation) influences the susceptibility of offspring to FA through multi-interface crosstalk. Finally, we discuss the limitations and gaps in the available evidence regarding the relationship between maternal dietary nutrients and the risk of FA in offspring. This review provides novel perspective on the regulation of offspring FA by maternal diet and nutrients.


Asunto(s)
Hipersensibilidad a los Alimentos , Microbioma Gastrointestinal , Fenómenos Fisiologicos Nutricionales Maternos , Nutrientes , Humanos , Femenino , Hipersensibilidad a los Alimentos/inmunología , Hipersensibilidad a los Alimentos/prevención & control , Hipersensibilidad a los Alimentos/etiología , Embarazo , Nutrientes/metabolismo , Animales , Dieta , Efectos Tardíos de la Exposición Prenatal
8.
Front Public Health ; 12: 1377456, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38706545

RESUMEN

Regression discontinuity design (RDD) is a quasi-experimental approach to study the causal effect of an exposure on later outcomes by exploiting the discontinuity in the exposure probability at an assignment variable cut-off. With the intent of facilitating the use of RDD in the Developmental Origins of Health and Disease (DOHaD) research, we describe the main aspects of the study design and review the studies, assignment variables and exposures that have been investigated to identify short- and long-term health effects of early life exposures. We also provide a brief overview of some of the methodological considerations for the RDD identification using an example of a DOHaD study. An increasing number of studies investigating the effects of early life environmental stressors on health outcomes use RDD, mostly in the context of education, social and welfare policies, healthcare organization and insurance, and clinical management. Age and calendar time are the mostly used assignment variables to study the effects of various early life policies and programs, shock events and guidelines. Maternal and newborn characteristics, such as age, birth weight and gestational age are frequently used assignment variables to study the effects of the type of neonatal care, health insurance, and newborn benefits, while socioeconomic measures have been used to study the effects of social and welfare programs. RDD has advantages, including intuitive interpretation, and transparent and simple graphical representation. It provides valid causal estimates if the assumptions, relatively weak compared to other non-experimental study designs, are met. Its use to study health effects of exposures acting early in life has been limited to studies based on registries and administrative databases, while birth cohort data has not been exploited so far using this design. Local causal effect around the cut-off, difficulty in reaching high statistical power compared to other study designs, and the rarity of settings outside of policy and program evaluations hamper the widespread use of RDD in the DOHaD research. Still, the assignment variables' cut-offs for exposures applied in previous studies can be used, if appropriate, in other settings and with additional outcomes to address different research questions.


Asunto(s)
Proyectos de Investigación , Humanos , Femenino , Recién Nacido , Embarazo , Exposición a Riesgos Ambientales/efectos adversos , Efectos Tardíos de la Exposición Prenatal , Análisis de Regresión
9.
Clin Epigenetics ; 16(1): 65, 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38741114

RESUMEN

OBJECTIVE: Youth-onset type 2 diabetes (T2D) is physiologically distinct from adult-onset, but it is not clear how the two diseases differ at a molecular level. In utero exposure to maternal type 2 diabetes (T2D) is known to be a specific risk factor for youth-onset T2D. DNA methylation (DNAm) changes associated with T2D but which differ between youth- and adult-onset might delineate the impacts of T2D development at different ages and could also determine the contribution of exposure to in utero diabetes. METHODS: We performed an epigenome-wide analysis of DNAm on whole blood from 218 youth with T2D and 77 normoglycemic controls from the iCARE (improving renal Complications in Adolescents with type 2 diabetes through REsearch) cohort. Associations were tested using multiple linear regression models while adjusting for maternal diabetes, sex, age, BMI, smoking status, second-hand smoking exposure, cell-type proportions and genetic ancestry. RESULTS: We identified 3830 differentially methylated sites associated with youth T2D onset, of which 3794 were moderately (adjusted p-value < 0.05 and effect size estimate > 0.01) associated and 36 were strongly (adjusted p-value < 0.05 and effect size estimate > 0.05) associated. A total of 3725 of these sites were not previously reported in the EWAS Atlas as associated with T2D, adult obesity or youth obesity. Moreover, three CpGs associated with youth-onset T2D in the PFKFB3 gene were also associated with maternal T2D exposure (FDR < 0.05 and effect size > 0.01). This is the first study to link PFKFB3 and T2D in youth. CONCLUSION: Our findings support that T2D in youth has different impacts on DNAm than adult-onset, and suggests that changes in DNAm could provide an important link between in utero exposure to maternal diabetes and the onset of T2D.


Asunto(s)
Metilación de ADN , Diabetes Mellitus Tipo 2 , Efectos Tardíos de la Exposición Prenatal , Humanos , Diabetes Mellitus Tipo 2/genética , Femenino , Metilación de ADN/genética , Embarazo , Adolescente , Masculino , Efectos Tardíos de la Exposición Prenatal/genética , Epigénesis Genética/genética , Edad de Inicio , Niño , Estudios de Casos y Controles , Diabetes Gestacional/genética , Adulto , Epigenoma/genética
10.
Clin Epigenetics ; 16(1): 61, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38715048

RESUMEN

BACKGROUND: Diabetes in pregnancy is associated with increased risk of long-term metabolic disease in the offspring, potentially mediated by in utero epigenetic variation. Previously, we identified multiple differentially methylated single CpG sites in offspring of women with gestational diabetes mellitus (GDM), but whether stretches of differentially methylated regions (DMRs) can also be identified in adolescent GDM offspring is unknown. Here, we investigate which DNA regions in adolescent offspring are differentially methylated in blood by exposure to diabetes in pregnancy. The secondary aim was to characterize the RNA expression of the identified DMR, which contained the nc886 non-coding RNA. METHODS: To identify DMRs, we employed the bump hunter method in samples from young (9-16 yr, n = 92) offspring of women with GDM (O-GDM) and control offspring (n = 94). Validation by pyrosequencing was performed in an adult offspring cohort (age 28-33 years) consisting of O-GDM (n = 82), offspring exposed to maternal type 1 diabetes (O-T1D, n = 67) and control offspring (O-BP, n = 57). RNA-expression was measured using RT-qPCR in subcutaneous adipose tissue and skeletal muscle. RESULTS: One significant DMR represented by 10 CpGs with a bimodal methylation pattern was identified, located in the nc886/VTRNA2-1 non-coding RNA gene. Low methylation status across all CpGs of the nc886 in the young offspring was associated with maternal GDM. While low methylation degree in adult offspring in blood, adipose tissue, and skeletal muscle was not associated with maternal GDM, adipose tissue nc886 expression was increased in O-GDM compared to O-BP, but not in O-T1D. In addition, adipose tissue nc886 expression levels were positively associated with maternal pre-pregnancy BMI (p = 0.006), but not with the offspring's own adiposity. CONCLUSIONS: Our results highlight that nc886 is a metastable epiallele, whose methylation in young offspring is negatively correlated with maternal obesity and GDM status. The physiological effect of nc886 may be more important in adipose tissue than in skeletal muscle. Further research should aim to investigate how nc886 regulation in adipose tissue by exposure to GDM may contribute to development of metabolic disease.


Asunto(s)
Tejido Adiposo , Metilación de ADN , Diabetes Gestacional , Epigénesis Genética , Músculo Esquelético , Efectos Tardíos de la Exposición Prenatal , Humanos , Embarazo , Femenino , Diabetes Gestacional/genética , Epigénesis Genética/genética , Adulto , Metilación de ADN/genética , Músculo Esquelético/metabolismo , Adolescente , Tejido Adiposo/metabolismo , Masculino , Efectos Tardíos de la Exposición Prenatal/genética , Niño , Diabetes Mellitus Tipo 1/genética , ARN no Traducido/genética , ARN no Traducido/sangre , ARN Largo no Codificante/genética , Islas de CpG/genética
11.
J Neuroinflammation ; 21(1): 118, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38715090

RESUMEN

Maternal inflammation during gestation is associated with a later diagnosis of neurodevelopmental disorders including autism spectrum disorder (ASD). However, the specific impact of maternal immune activation (MIA) on placental and fetal brain development remains insufficiently understood. This study aimed to investigate the effects of MIA by analyzing placental and brain tissues obtained from the offspring of pregnant C57BL/6 dams exposed to polyinosinic: polycytidylic acid (poly I: C) on embryonic day 12.5. Cytokine and mRNA content in the placenta and brain tissues were assessed using multiplex cytokine assays and bulk-RNA sequencing on embryonic day 17.5. In the placenta, male MIA offspring exhibited higher levels of GM-CSF, IL-6, TNFα, and LT-α, but there were no differences in female MIA offspring. Furthermore, differentially expressed genes (DEG) in the placental tissues of MIA offspring were found to be enriched in processes related to synaptic vesicles and neuronal development. Placental mRNA from male and female MIA offspring were both enriched in synaptic and neuronal development terms, whereas females were also enriched for terms related to excitatory and inhibitory signaling. In the fetal brain of MIA offspring, increased levels of IL-28B and IL-25 were observed with male MIA offspring and increased levels of LT-α were observed in the female offspring. Notably, we identified few stable MIA fetal brain DEG, with no male specific difference whereas females had DEG related to immune cytokine signaling. Overall, these findings support the hypothesis that MIA contributes to the sex- specific abnormalities observed in ASD, possibly through altered neuron developed from exposure to inflammatory cytokines. Future research should aim to investigate how interactions between the placenta and fetal brain contribute to altered neuronal development in the context of MIA.


Asunto(s)
Encéfalo , Citocinas , Ratones Endogámicos C57BL , Trastornos del Neurodesarrollo , Placenta , Efectos Tardíos de la Exposición Prenatal , Caracteres Sexuales , Femenino , Animales , Embarazo , Masculino , Citocinas/metabolismo , Citocinas/genética , Ratones , Encéfalo/metabolismo , Encéfalo/inmunología , Encéfalo/embriología , Placenta/metabolismo , Placenta/inmunología , Efectos Tardíos de la Exposición Prenatal/inmunología , Efectos Tardíos de la Exposición Prenatal/metabolismo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/inmunología , Trastornos del Neurodesarrollo/metabolismo , Poli I-C/toxicidad , Transcriptoma , Modelos Animales de Enfermedad , Feto/metabolismo
12.
Epigenetics Chromatin ; 17(1): 14, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38715099

RESUMEN

BACKGROUND: Prenatal nicotine exposure (PNE) has been documented to cause numerous deleterious effects on fetal development. However, the epigenetic changes promoted by nicotine exposure on germ cells are still not well understood. OBJECTIVES: In this study, we focused on elucidating the impact of prenatal nicotine exposure on regulatory epigenetic mechanisms important for germ cell development. METHODS: Sprague-Dawley rats were exposed to nicotine during pregnancy and male progeny was analyzed at 11 weeks of age. Testis morphology was analyzed using frozen testis sections and expression of germ cell markers was examined by RT-qPCR; histone modifications were assessed by Western Blot (WB). DNA methylation analysis was performed by methylation-specific PCR of bisulfite converted DNA. Genome-wide DNA methylation was analyzed using Methylated DNA immunoprecipitation (MeDIP)-seq. We also carried out transcriptomics analysis of pituitary glands by RNA-seq. RESULTS: We show that gestational exposure to nicotine reduces germ cell numbers, perturbs meiosis, affects the expression of germ line reprogramming responsive genes, and impacts the DNA methylation of nervous system genes in the testis. PNE also causes perturbation of gene expression in the pituitary gland of the brain. CONCLUSIONS: Our data demonstrate that PNE leads to perturbation of male spermatogenesis, and the observed effects are associated with changes of peripheral nervous system signaling pathways. Alterations in the expression of genes associated with diverse biological activities such as cell migration, cell adhesion and GABA signaling in the pituitary gland underscore the complexity of the effects of nicotine exposure during pregnancy.


Asunto(s)
Metilación de ADN , Epigénesis Genética , Nicotina , Efectos Tardíos de la Exposición Prenatal , Ratas Sprague-Dawley , Testículo , Animales , Masculino , Femenino , Embarazo , Ratas , Testículo/efectos de los fármacos , Testículo/metabolismo , Epigénesis Genética/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/metabolismo , Transducción de Señal/efectos de los fármacos , Espermatogénesis/efectos de los fármacos , Espermatogénesis/genética , Sistema Nervioso Periférico/efectos de los fármacos , Sistema Nervioso Periférico/metabolismo
13.
Sci Adv ; 10(19): eadg9674, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38718116

RESUMEN

Prenatal opioid exposure is an established public health problem, in particular among Medicaid-covered births. Yet, existing prevalence rates are plausibly underestimated. We leverage extensive linked longitudinal administrative data for all Medicaid-covered live births in Wisconsin from 2010 to 2019 to estimate a range of prevalence rates using an innovative strategy that jointly accounts for both likelihood of exposure and potential risk to prenatal development. We find that 20.8% of infants may have been prenatally exposed to opioids, with 1.7% diagnosed with neonatal abstinence syndrome and an additional 1.2% having a high combined likelihood of exposure and potential risk to prenatal development, 2.6% a moderate combined likelihood and risk, and 15.3% a low or uncertain combined likelihood and risk. We assess improvements in prevalence estimates based on our nuanced classification relative to those of prior studies. Our strategy could be broadly used to quantify the scope of the opioid crisis for pregnant populations, target interventions, and promote child health and development.


Asunto(s)
Analgésicos Opioides , Medicaid , Efectos Tardíos de la Exposición Prenatal , Humanos , Wisconsin/epidemiología , Embarazo , Femenino , Estados Unidos/epidemiología , Efectos Tardíos de la Exposición Prenatal/epidemiología , Analgésicos Opioides/efectos adversos , Recién Nacido , Síndrome de Abstinencia Neonatal/epidemiología , Trastornos Relacionados con Opioides/epidemiología , Prevalencia , Adulto , Factores de Riesgo
14.
JAMA Netw Open ; 7(5): e2410151, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38713462

RESUMEN

Importance: The prevalence of cannabis use in pregnancy is rising and is associated with adverse perinatal outcomes. In parallel, combined prenatal use of cannabis and nicotine is also increasing, but little is known about the combined impact of both substances on pregnancy and offspring outcomes compared with each substance alone. Objective: To assess the perinatal outcomes associated with combined cannabis and nicotine exposure compared with each substance alone during pregnancy. Design, Setting, and Participants: This retrospective population-based cohort study included linked hospital discharge data (obtained from the California Department of Health Care Access and Information) and vital statistics (obtained from the California Department of Public Health) from January 1, 2012, through December 31, 2019. Pregnant individuals with singleton gestations and gestational ages of 23 to 42 weeks were included. Data were analyzed from October 14, 2023, to March 4, 2024. Exposures: Cannabis-related diagnosis and prenatal nicotine product use were captured using codes from International Classification of Diseases, Ninth Revision, Clinical Modification, and International Statistical Classification of Diseases, Tenth Revision, Clinical Modification. Main Outcome and Measures: The main outcomes were infant and neonatal death, infants small for gestational age, and preterm delivery. Results were analyzed by multivariable Poisson regression models. Results: A total of 3 129 259 pregnant individuals were included (mean [SD] maternal age 29.3 [6.0] years), of whom 23 007 (0.7%) had a cannabis-related diagnosis, 56 811 (1.8%) had a nicotine-use diagnosis, and 10 312 (0.3%) had both in pregnancy. Compared with nonusers, those with cannabis or nicotine use diagnoses alone had increased rates of infant (0.7% for both) and neonatal (0.3% for both) death, small for gestational age (14.3% and 13.7%, respectively), and preterm delivery (<37 weeks) (12.2% and 12.0%, respectively). Moreover, risks in those with both cannabis and nicotine use were higher for infant death (1.2%; adjusted risk ratio [ARR], 2.18 [95% CI, 1.82-2.62]), neonatal death (0.6%; ARR, 1.76 [95% CI, 1.36-2.28]), small for gestational age (18.0%; ARR, 1.94 [95% CI, 1.86-2.02]), and preterm delivery (17.5%; ARR, 1.83 [95% CI, 1.75-1.91]). Conclusions and Relevance: These findings suggest that co-occurring maternal use of cannabis and nicotine products in pregnancy is associated with an increased risk of infant and neonatal death and maternal and neonatal morbidity compared with use of either substance alone. Given the increasing prevalence of combined cannabis and nicotine use in pregnancy, these findings can help guide health care practitioners with preconception and prenatal counseling, especially regarding the benefits of cessation.


Asunto(s)
Nicotina , Efectos Tardíos de la Exposición Prenatal , Humanos , Femenino , Embarazo , Recién Nacido , Adulto , Estudios Retrospectivos , Nicotina/efectos adversos , California/epidemiología , Efectos Tardíos de la Exposición Prenatal/epidemiología , Nacimiento Prematuro/epidemiología , Recién Nacido Pequeño para la Edad Gestacional , Resultado del Embarazo/epidemiología , Lactante , Cannabis/efectos adversos , Adulto Joven
15.
Sci Adv ; 10(18): eadl3747, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38701212

RESUMEN

Early-life tobacco exposure serves as a non-negligible risk factor for aging-related diseases. To understand the underlying mechanisms, we explored the associations of early-life tobacco exposure with accelerated biological aging and further assessed the joint effects of tobacco exposure and genetic susceptibility. Compared with those without in utero exposure, participants with in utero tobacco exposure had an increase in Klemera-Doubal biological age (KDM-BA) and PhenoAge acceleration of 0.26 and 0.49 years, respectively, but a decrease in telomere length of 5.34% among 276,259 participants. We also found significant dose-response associations between the age of smoking initiation and accelerated biological aging. Furthermore, the joint effects revealed that high-polygenic risk score participants with in utero exposure and smoking initiation in childhood had the highest accelerated biological aging. There were interactions between early-life tobacco exposure and age, sex, deprivation, and diet on KDM-BA and PhenoAge acceleration. These findings highlight the importance of reducing early-life tobacco exposure to improve healthy aging.


Asunto(s)
Envejecimiento , Predisposición Genética a la Enfermedad , Efectos Tardíos de la Exposición Prenatal , Humanos , Femenino , Masculino , Efectos Tardíos de la Exposición Prenatal/genética , Envejecimiento/genética , Adulto , Embarazo , Nicotiana/efectos adversos , Nicotiana/genética , Fumar/efectos adversos , Factores de Riesgo , Persona de Mediana Edad
16.
Brain Behav ; 14(5): e3515, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38702895

RESUMEN

INTRODUCTION: Maternal sleep deprivation (MSD), which induces inflammation and synaptic dysfunction in the hippocampus, has been associated with learning and memory impairment in offspring. Melatonin (Mel) has been shown to have anti-inflammatory, antioxidant, and neuroprotective function. However, the beneficial effect of Mel on MSD-induced cognitive impairment and its mechanisms are unknown. METHODS: In the present study, adult offspring suffered from MSD were injected with Mel (20 mg/kg) once a day during postnatal days 61-88. The cognitive function was evaluated by the Morris water maze test. Levels of proinflammatory cytokines were examined by enzyme-linked immunosorbent assay. The mRNA and protein levels of synaptic plasticity associated proteins were examined using reverse transcription-polymerase chain reaction and western blotting. RESULTS: The results showed that MSD impaired learning and memory in the offspring mice. MSD increased the levels of interleukin (IL)-1creIL-6, and tumor necrosis factor-α and decreased the expression levels of brain-derived neurotrophic factor, tyrosine kinase receptor B, postsynaptic density protein-95, and synaptophysin in the hippocampus. Furthermore, Mel attenuated cognitive impairment and restored markers of inflammation and synaptic plasticity to control levels. CONCLUSIONS: These findings indicated that Mel could ameliorate learning and memory impairment induced by MSD, and these beneficial effects were related to improvement in inflammation and synaptic dysfunction.


Asunto(s)
Hipocampo , Melatonina , Trastornos de la Memoria , Plasticidad Neuronal , Privación de Sueño , Animales , Melatonina/farmacología , Melatonina/administración & dosificación , Privación de Sueño/complicaciones , Privación de Sueño/tratamiento farmacológico , Privación de Sueño/fisiopatología , Ratones , Masculino , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos , Femenino , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/etiología , Trastornos de la Memoria/fisiopatología , Plasticidad Neuronal/efectos de los fármacos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Embarazo , Privación Materna , Disfunción Cognitiva/etiología , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/fisiopatología , Efectos Tardíos de la Exposición Prenatal/metabolismo , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Enfermedades Neuroinflamatorias/tratamiento farmacológico
17.
BMC Pregnancy Childbirth ; 24(1): 344, 2024 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-38704541

RESUMEN

BACKGROUND: Climate change, particularly global warming, is amongst the greatest threats to human health. While short-term effects of heat exposure in pregnancy, such as preterm birth, are well documented, long-term effects have received less attention. This review aims to systematically assess evidence on the long-term impacts on the foetus of heat exposure in utero. METHODS: A search was conducted in August 2019 and updated in April 2023 in MEDLINE(PubMed). We included studies on the relationship of environmental heat exposure during pregnancy and any long-term outcomes. Risk of bias was assessed using tools developed by the Joanna-Briggs Institute, and the evidence was appraised using the GRADE approach. Synthesis without Meta-Analysis (SWiM) guidelines were used. RESULTS: Eighteen thousand six hundred twenty one records were screened, with 29 studies included across six outcome groups. Studies were mostly conducted in high-income countries (n = 16/25), in cooler climates. All studies were observational, with 17 cohort, 5 case-control and 8 cross-sectional studies. The timeline of the data is from 1913 to 2019, and individuals ranged in age from neonates to adults, and the elderly. Increasing heat exposure during pregnancy was associated with decreased earnings and lower educational attainment (n = 4/6), as well as worsened cardiovascular (n = 3/6), respiratory (n = 3/3), psychiatric (n = 7/12) and anthropometric (n = 2/2) outcomes, possibly culminating in increased overall mortality (n = 2/3). The effect on female infants was greater than on males in 8 of 9 studies differentiating by sex. The quality of evidence was low in respiratory and longevity outcome groups to very low in all others. CONCLUSIONS: Increasing heat exposure was associated with a multitude of detrimental outcomes across diverse body systems. The biological pathways involved are yet to be elucidated, but could include epigenetic and developmental perturbations, through interactions with the placenta and inflammation. This highlights the need for further research into the long-term effects of heat exposure, biological pathways, and possible adaptation strategies in studies, particularly in neglected regions. Heat exposure in-utero has the potential to compound existing health and social inequalities. Poor study design of the included studies constrains the conclusions of this review, with heterogenous exposure measures and outcomes rendering comparisons across contexts/studies difficult. TRIAL REGISTRATION: PROSPERO CRD 42019140136.


Asunto(s)
Calor , Efectos Tardíos de la Exposición Prenatal , Humanos , Femenino , Embarazo , Calor/efectos adversos , Cambio Climático , Recién Nacido , Adulto
18.
Reprod Toxicol ; 1232024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38706688

RESUMEN

Exposure to gestational diabetes mellitus (GDM) during pregnancy has significant consequences for the unborn baby and newborn infant. However, whether and how GDM exposure induces the development of neonatal brain hypoxia/ischemia-sensitive phenotype and the underlying molecular mechanisms remain unclear. In this study, we used a late GDM rat model induced by administration of streptozotocin (STZ) on gestational day 12 and investigated its effects of GDM on neonatal brain development. The pregnant rats exhibited increased blood glucose levels in a dose-dependent manner after STZ administration. STZ-induced maternal hyperglycemia led to reduced blood glucose levels in neonatal offspring, resulting in growth restriction and an increased brain to body weight ratio. Importantly, GDM exposure increased susceptibility to hypoxia/ischemia (HI)-induced brain infarct sizes compared to the controls in both male and female neonatal offspring. Further molecular analysis revealed alterations in the PTEN/AKT/mTOR/autophagy signaling pathway in neonatal male offspring brains, along with increased ROS production and autophagy-related proteins (Atg5 and LC3-II). Treatment with the PTEN inhibitor bisperoxovanadate (BPV) eliminated the differences in HI-induced brain infarct sizes between the GDM-exposed and the control groups. These findings provide novel evidence of the development of a brain hypoxia/ischemia-sensitive phenotype in response to GDM exposure and highlight the role of the PTEN/AKT/mTOR/autophagy signaling pathway in this process.


Asunto(s)
Animales Recién Nacidos , Autofagia , Encéfalo , Diabetes Gestacional , Hipoxia-Isquemia Encefálica , Fosfohidrolasa PTEN , Proteínas Proto-Oncogénicas c-akt , Ratas Sprague-Dawley , Transducción de Señal , Estreptozocina , Serina-Treonina Quinasas TOR , Animales , Femenino , Embarazo , Hipoxia-Isquemia Encefálica/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Autofagia/efectos de los fármacos , Diabetes Gestacional/inducido químicamente , Diabetes Gestacional/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Masculino , Fosfohidrolasa PTEN/metabolismo , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/patología , Efectos Tardíos de la Exposición Prenatal , Glucemia , Ratas
19.
Cereb Cortex ; 34(13): 146-160, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38696608

RESUMEN

Autism spectrum disorder is a neurodevelopmental disability that includes sensory disturbances. Hearing is frequently affected and ranges from deafness to hypersensitivity. In utero exposure to the antiepileptic valproic acid is associated with increased risk of autism spectrum disorder in humans and timed valproic acid exposure is a biologically relevant and validated animal model of autism spectrum disorder. Valproic acid-exposed rats have fewer neurons in their auditory brainstem and thalamus, fewer calbindin-positive neurons, reduced ascending projections to the midbrain and thalamus, elevated thresholds, and delayed auditory brainstem responses. Additionally, in the auditory cortex, valproic acid exposure results in abnormal responses, decreased phase-locking, elevated thresholds, and abnormal tonotopic maps. We therefore hypothesized that in utero, valproic acid exposure would result in fewer neurons in auditory cortex, neuronal dysmorphology, fewer calbindin-positive neurons, and reduced connectivity. We approached this hypothesis using morphometric analyses, immunohistochemistry, and retrograde tract tracing. We found thinner cortical layers but no changes in the density of neurons, smaller pyramidal and non-pyramidal neurons in several regions, fewer neurons immunoreactive for calbindin-positive, and fewer cortical neurons projecting to the inferior colliculus. These results support the widespread impact of the auditory system in autism spectrum disorder and valproic acid-exposed animals and emphasize the utility of simple, noninvasive auditory screening for autism spectrum disorder.


Asunto(s)
Corteza Auditiva , Trastorno del Espectro Autista , Calbindinas , Modelos Animales de Enfermedad , Ácido Valproico , Animales , Trastorno del Espectro Autista/patología , Trastorno del Espectro Autista/metabolismo , Trastorno del Espectro Autista/inducido químicamente , Ácido Valproico/toxicidad , Femenino , Calbindinas/metabolismo , Corteza Auditiva/patología , Corteza Auditiva/efectos de los fármacos , Corteza Auditiva/metabolismo , Embarazo , Neuronas/patología , Neuronas/metabolismo , Ratas , Masculino , Vías Auditivas/patología , Vías Auditivas/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal/patología , Ratas Sprague-Dawley , Anticonvulsivantes
20.
Reprod Fertil Dev ; 362024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38739740

RESUMEN

Context A maternal high-fat diet is thought to pose a risk to spermatogenesis in the progeny. Aims We tested whether a maternal high-fat diet would affect Sertoli cell expression of transcription factors (insulin-like growth factor I (IGF-I); glial-cell line-derived neurotrophic factor (GDNF); Ets variant 5 (ETV5)) and cell proliferation and apoptotic proteins, in the testis of adult offspring. Methods Pregnant rats were fed ad libitum with a standard diet (Control) or a high-fat diet (HFat) throughout pregnancy and lactation. After weaning, male pups were fed the standard diet until postnatal day 160. Males were monitored daily from postnatal day 34 to determine onset of puberty. On postnatal day 160, their testes were processed for morphometry and immunohistochemistry. Key results The HFat diet increased seminiferous-tubule diameter (P P P P P P P P Conclusions A maternal high-fat diet alters the balance between spermatogonia proliferation and spermatid apoptosis. Implications A maternal high-fat diet seems to 'program' adult male fertility.


Asunto(s)
Apoptosis , Proliferación Celular , Dieta Alta en Grasa , Lactancia , Fenómenos Fisiologicos Nutricionales Maternos , Efectos Tardíos de la Exposición Prenatal , Testículo , Animales , Femenino , Masculino , Embarazo , Apoptosis/fisiología , Lactancia/fisiología , Testículo/metabolismo , Testículo/patología , Ratas , Efectos Tardíos de la Exposición Prenatal/patología , Efectos Tardíos de la Exposición Prenatal/metabolismo , Fenómenos Fisiologicos Nutricionales Maternos/fisiología , Espermatogénesis/fisiología , Células de Sertoli/metabolismo , Células de Sertoli/patología , Factor I del Crecimiento Similar a la Insulina/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA