Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Birth Defects Res ; 116(6): e2369, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38877673

RESUMEN

BACKGROUND: Prenatal alcohol exposure (PAE) has been linked to congenital heart disease and fetal alcohol syndrome. The heart primarily relies on mitochondria to generate energy, so impaired mitochondrial function due to alcohol exposure can significantly affect cardiac development and function. Our study aimed to investigate the impact of PAE on myocardial and mitochondrial functions in offspring mice. METHODS: We administered 30% alcohol (3 g/kg) to pregnant C57BL/6 mice during the second trimester. We assessed cardiac function by transthoracic echocardiography, observed myocardial structure and fibrosis through staining tests and electron transmission microscopy, and detected cardiomyocyte apoptosis with dUTP nick end labeling assay and real-time quantitative PCR. Additionally, we measured the reactive oxygen species content, ATP level, and mitochondrial DNA copy number in myocardial mitochondria. Mitochondrial damage was evaluated by assessing the level of mitochondrial membrane potential and the opening degree of mitochondrial permeability transition pores. RESULTS: Our findings revealed that PAE caused cardiac systolic dysfunction, ventricular enlargement, thinned ventricular wall, cardiac fibrosis in the myocardium, scattered loss of cardiomyocytes, and disordered arrangement of myocardial myotomes in the offspring. Furthermore, we observed a significant increase in mitochondrial reactive oxygen species content, a decrease in mitochondrial membrane potential, ATP level, and mitochondrial DNA copy number, and sustained opening of mitochondrial permeability transition pores in the heart tissues of the offspring. CONCLUSIONS: These results indicated that PAE had adverse effects on the cardiac structure and function of the newborn mice and could trigger oxidative stress in their myocardia and contribute to mitochondrial dysfunction.


Asunto(s)
Etanol , Ratones Endogámicos C57BL , Miocitos Cardíacos , Efectos Tardíos de la Exposición Prenatal , Especies Reactivas de Oxígeno , Animales , Femenino , Embarazo , Ratones , Efectos Tardíos de la Exposición Prenatal/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Etanol/efectos adversos , Etanol/toxicidad , ADN Mitocondrial/efectos de los fármacos , ADN Mitocondrial/metabolismo , Apoptosis/efectos de los fármacos , Mitocondrias Cardíacas/metabolismo , Mitocondrias Cardíacas/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Miocardio/metabolismo , Miocardio/patología , Estrés Oxidativo/efectos de los fármacos , Trastornos del Espectro Alcohólico Fetal/metabolismo , Trastornos del Espectro Alcohólico Fetal/patología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Ecocardiografía , Fibrosis
2.
Alcohol ; 118: 45-55, 2024 08.
Artículo en Inglés | MEDLINE | ID: mdl-38705312

RESUMEN

Prenatal alcohol exposure can have persistent effects on learning, memory, and synaptic plasticity. Previous work from our group demonstrated deficits in long-term potentiation (LTP) of excitatory synapses on dentate gyrus granule cells in adult offspring of rat dams that consumed moderate levels of alcohol during pregnancy. At present, there are no pharmacotherapeutic agents approved for these deficits. Prior work established that systemic administration of the histaminergic H3R inverse agonist ABT-239 reversed deficits in LTP observed following moderate PAE. The present study examines the effect of a second H3R inverse agonist, SAR-152954, on LTP deficits following moderate PAE. We demonstrate that systemic administration of 1 mg/kg of SAR-152954 reverses deficits in potentiation of field excitatory post-synaptic potentials (fEPSPs) in adult male rats exposed to moderate PAE. Time-frequency analyses of evoked responses revealed PAE-related reductions in power during the fEPSP, and increased power during later components of evoked responses which are associated with feedback circuitry that are typically not assessed with traditional amplitude-based measures. Both effects were reversed by SAR-152954. These findings provide further evidence that H3R inverse agonism is a potential therapeutic strategy to address deficits in synaptic plasticity associated with PAE.


Asunto(s)
Potenciación a Largo Plazo , Efectos Tardíos de la Exposición Prenatal , Receptores Histamínicos H3 , Animales , Potenciación a Largo Plazo/efectos de los fármacos , Femenino , Masculino , Ratas , Embarazo , Receptores Histamínicos H3/metabolismo , Receptores Histamínicos H3/efectos de los fármacos , Agonistas de los Receptores Histamínicos/farmacología , Ratas Sprague-Dawley , Etanol/farmacología , Agonismo Inverso de Drogas , Potenciales Postsinápticos Excitadores/efectos de los fármacos
3.
Biol Res ; 56(1): 61, 2023 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-37978540

RESUMEN

Prenatal ethanol exposure (PEE) (mainly through maternal alcohol consumption) has become widespread. However, studies suggest that it can cause intrauterine growth retardation (IUGR) and multi-organ developmental toxicity in offspring, and susceptibility to various chronic diseases (such as neuropsychiatric diseases, metabolic syndrome, and related diseases) in adults. Through ethanol's direct effects and its indirect effects mediated by maternal-derived glucocorticoids, PEE alters epigenetic modifications and organ developmental programming during fetal development, which damages the offspring health and increases susceptibility to various chronic diseases after birth. Ethanol directly leads to the developmental toxicity of multiple tissues and organs in many ways. Regarding maternal-derived glucocorticoid-mediated IUGR, developmental programming, and susceptibility to multiple conditions after birth, ethanol induces programmed changes in the neuroendocrine axes of offspring, such as the hypothalamus-pituitary-adrenal (HPA) and glucocorticoid-insulin-like growth factor 1 (GC-IGF1) axes. In addition, the differences in ethanol metabolic enzymes, placental glucocorticoid barrier function, and the sensitivity to glucocorticoids in various tissues and organs mediate the severity and sex differences in the developmental toxicity of ethanol exposure during pregnancy. Offspring exposed to ethanol during pregnancy have a "thrifty phenotype" in the fetal period, and show "catch-up growth" in the case of abundant nutrition after birth; when encountering adverse environments, these offspring are more likely to develop diseases. Here, we review the developmental toxicity, functional alterations in multiple organs, and neuroendocrine metabolic programming mechanisms induced by PEE based on our research and that of other investigators. This should provide new perspectives for the effective prevention and treatment of ethanol developmental toxicity and the early prevention of related fetal-originated diseases.


Asunto(s)
Glucocorticoides , Efectos Tardíos de la Exposición Prenatal , Ratas , Animales , Adulto , Femenino , Embarazo , Humanos , Masculino , Glucocorticoides/metabolismo , Glucocorticoides/farmacología , Ratas Wistar , Placenta/metabolismo , Desarrollo Fetal , Etanol/toxicidad , Enfermedad Crónica
4.
Dev Psychobiol ; 65(7): e22426, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37860900

RESUMEN

Prenatal ethanol exposure (PEE) causes several neurobehavioral impairments in the fetus. Postnatal days (PDs) 4-9 in rodents are considered equivalent to the third trimester of gestation in humans. This period is characterized by high rates of synaptogenesis and myelination and the maturation of key structures and transmitter systems. Nutritional supplements, such as folate, have gained attention as putative treatments to mitigate detrimental effects of PEE. Folate is crucial for DNA synthesis and amino acid metabolism and heightens antioxidant defenses. The present study examined neurobehavioral effects of the concurrent administration of folate (20 mg/kg/day) and ethanol (5 g/kg/day) during PDs 4-9 in male and female Wistar rats. During PDs 16-18, the rat pups were tested for anxiety-like and exploratory activity in the light-dark box (LDB), open field (OF), and concentric square field (CSF) tests. After weaning, they were tested for sucrose preference and ethanol intake. Neonatal ethanol exposure reduced body weight in infancy but did not enhance ethanol self-administration or significantly affect performance in the OF or LDB. Neonatal ethanol exposure also reduced sucrose intake in the preference test and increased shelter-seeking in the CSF, and folate significantly inhibited these effects. The present findings suggest that folate, a treatment that is devoid of serious side effects, can ameliorate some neurobehavioral effects of PEE.


Asunto(s)
Etanol , Efectos Tardíos de la Exposición Prenatal , Embarazo , Humanos , Ratas , Animales , Masculino , Femenino , Etanol/farmacología , Ratas Wistar , Ácido Fólico/farmacología , Sacarosa
5.
Cells ; 12(18)2023 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-37759545

RESUMEN

Fetal alcohol spectrum disorders (FASD) are a set of abnormalities caused by prenatal exposure to ethanol and are characterized by developmental defects in the brain that lead to various overt and non-overt physiological abnormalities. Growing evidence suggests that in utero alcohol exposure induces functional and structural abnormalities in gliogenesis and neuron-glia interactions, suggesting a possible role of glial cell pathologies in the development of FASD. However, the molecular mechanisms of neuron-glia interactions that lead to the development of FASD are not clearly understood. In this review, we discuss glial cell pathologies with a particular emphasis on microglia, primary resident immune cells in the brain. Additionally, we examine the involvement of several neuroimmune molecules released by glial cells, their signaling pathways, and epigenetic mechanisms responsible for FASD-related alteration in brain functions. Growing evidence suggests that extracellular vesicles (EVs) play a crucial role in the communication between cells via transporting bioactive cargo from one cell to the other. This review emphasizes the role of EVs in the context of neuron-glia interactions during prenatal alcohol exposure. Finally, some potential applications involving nutritional, pharmacological, cell-based, and exosome-based therapies in the treatment of FASD are discussed.

6.
Pharmacol Res ; 194: 106855, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37460002

RESUMEN

Fetal alcohol spectrum disorders (FASD) are a group of preventable and nongenetic birth defects caused by prenatal alcohol exposure that can result in a range of cognitive, behavioral, emotional, and functioning deficits, as well as craniofacial dysmorphology and other congenital defects. During embryonic development, neural crest cells (NCCs) play a critical role in giving rise to many cell types in the developing embryos, including those in the peripheral nervous system and craniofacial structures. Ethanol exposure during this critical period can have detrimental effects on NCC induction, migration, differentiation, and survival, leading to a broad range of structural and functional abnormalities observed in individuals with FASD. This review article provides an overview of the current knowledge on the detrimental effects of ethanol on NCC induction, migration, differentiation, and survival. The article also examines the molecular mechanisms involved in ethanol-induced NCC dysfunction, such as oxidative stress, altered gene expression, apoptosis, epigenetic modifications, and other signaling pathways. Furthermore, the review highlights potential therapeutic strategies for preventing or mitigating the detrimental effects of ethanol on NCCs and reducing the risk of FASD. Overall, this article offers a comprehensive overview of the current understanding of the impact of ethanol on NCCs and its role in FASD, shedding light on potential avenues for future research and intervention.


Asunto(s)
Trastornos del Espectro Alcohólico Fetal , Efectos Tardíos de la Exposición Prenatal , Humanos , Femenino , Embarazo , Trastornos del Espectro Alcohólico Fetal/prevención & control , Cresta Neural , Efectos Tardíos de la Exposición Prenatal/metabolismo , Transducción de Señal , Etanol/toxicidad
7.
Am J Drug Alcohol Abuse ; 49(1): 63-75, 2023 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-36722686

RESUMEN

Background: Prenatal ethanol exposure (PEE) induces heightened ethanol intake at adolescence in preclinical studies. Ethanol intake alters the absorption of folate, a methyl-group donor critical for numerous cellular functions. The prenatal administration of folate is, therefore, a promising approach to reduce the effects of PEE.Objectives: Experiment 1 determined if prenatal folate modulated the effects of PEE on ethanol intake, anxiety-like response, and exploratory behaviors (Experiment 1) in Wistar rats. Experiment 2 assessed, in rats not given PEE, if postnatal folate reversed effects of ethanol exposure at postnatal days 28-42. Experiment 3 assessed if folate altered blood ethanol levels (BELs).Methods: Experiment 1 involved 242 (125 male) adolescent Wistar rats derived from dams given folate (20 mg/kg, gestational days - GD- 13-20) + ethanol (2.0 g/kg, GD 17-20), ethanol, or vehicle only at pregnancy. Experiment 2 involved 29 male adolescents administered vehicle or ethanol doses co-administered or not with folate. In Experiment 3 twelve adult females were tested for BELs after folate administration. These tests were applied: intake tests, light dark box (LDB), elevated plus maze, open field and concentric square field.Results: PEE heightened ethanol intake (η2 ps = 0.06-07) and induced hyperactivity and a reduced latency to exit the white area of the LDB (η2 ps = 0.12-17). These effects were partially inhibited by folate (p > .05). Rats exposed to ethanol exposure at adolescence exhibited reduced motor activity (η2 p = .17), regardless of folate treatment. Folate did not affect BELs.Conclusion: Folate administration should be considered as a preventive or acute treatment to attenuate the neurobehavioral effects of PEE.


Asunto(s)
Etanol , Ácido Fólico , Embarazo , Femenino , Ratas , Masculino , Animales , Ratas Wistar , Consumo de Bebidas Alcohólicas , Ansiedad
8.
Nutr Neurosci ; 26(1): 60-71, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34957933

RESUMEN

Objectives: Ceramide (Cer), known as apoptotic markers, increases with prenatal ethanol (EtOH) exposure, resulting in neuroapoptosis. Whether maternal nutrition can impact Cer concentrations in brain, via altering plasma and brain fatty acid compositions have not been examined. This study compared a standard chow with a formulated semi-purified energy dense (E-dense) diet on fatty acid composition, Cer concentrations, and apoptosis in plasma and brain regions (cortex, cerebellum, and hippocampus) of pups exposed to EtOH during gestation. Methods: Pregnant Sprague-Dawley rats were randomized into four groups: chow (n = 6), chow + EtOH (20% v/v) (n = 7), E-dense (n = 6), and E-dense + EtOH (n = 8). At postnatal day 7, representing the peak brain growth spurt in rats, lipids, and apoptosis were analyzed by gas chromatography and a fluorometric caspase-3 assay kit, respectively. Results: Maternal E-dense diet increased total fatty acid concentrations (p < 0.0001), including docosahexaenoic acid (DHA) (p < 0.0001) in plasma, whereas DHA concentrations were decreased in the cerebellum (p < 0.03) of pups than those from chow-fed dams. EtOH-induced Cer elevations in the hippocampus of pups born to dams fed chow were reduced by an E-dense diet (p < 0.02). No significant effects of maternal diet quality and EtOH were observed on caspase-3 activity. No significant correlations existed between plasma/brain fatty acids and Cer concentrations. Discussions: Maternal diet quality affected fatty acid compositions and Cer concentrations of pups with prenatal EtOH exposure, differently. Maternal nutrition has the potential to prevent or alleviate some of the adverse effects of prenatal EtOH exposure.


Asunto(s)
Dieta , Etanol , Ácidos Grasos , Efectos Tardíos de la Exposición Prenatal , Animales , Femenino , Embarazo , Ratas , Animales Recién Nacidos , Encéfalo , Caspasa 3 , Ácidos Docosahexaenoicos/farmacología , Etanol/efectos adversos , Ratas Sprague-Dawley
9.
Chem Biol Interact ; 368: 110201, 2022 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-36174738

RESUMEN

PURPOSE: This study aims to further explore cartilage development in prenatal ethanol exposure (PEE) offspring at different times to explore the specific time points and mechanism of ethanol-induced fetal cartilage dysplasia. METHODS: On gestational day (GD)14, GD17, and GD20, PEE fetal cartilage was evaluated by morphological analysis. RT-qPCR, immunohistochemistry, and immunofluorescence were used to detect the expression of cartilage marker genes and their regulatory factors. Bone marrow mesenchymal stem cells (BMSCs) were used to explore the effect of ethanol on the differentiation of chondrocytes. Additionally, we used inhibitors, overexpression plasmids and a luciferase reporter assay on GD17 chondrocytes to verify the mechanism. RESULTS: PEE significantly reduced cartilage matrix content and the expression of marker genes on GD17 and GD20 but had no effect on GD14. The inhibition of chondrogenic differentiation by PEE mainly occurred on GD14-17. Furthermore, the expression of miR-200b-3p was increased, while that of ERG and PTHrP was markedly reduced in PEE fetal cartilage. In vitro, ethanol (30-120 mM) inhibited the differentiation of BMSCs into chondrocytes in a concentration-dependent manner, accompanied by strong expression of miR-200b-3p and low expression of ERG and PTHrP. Moreover, PTHLH and ERG overexpressed, as well as a miR-200b-3p inhibitor reversed the inhibitory effect of ethanol on the differentiation of fetal chondrocytes. Furthermore, miR-200b-3p could target and negatively regulate ERG. CONCLUSIONS: PEE can significantly inhibit the development of articular cartilage, especially during articular cartilage formation. The mechanism is related to the decreased differentiation of fetal cartilage into articular cartilage mediated by the miR-200b-3p/ERG/PTHrP axis.


Asunto(s)
Cartílago Articular , MicroARNs , Femenino , Embarazo , Cartílago Articular/metabolismo , Condrocitos , Etanol/farmacología , Etanol/metabolismo , MicroARNs/metabolismo , Proteína Relacionada con la Hormona Paratiroidea/genética , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Proteína Relacionada con la Hormona Paratiroidea/farmacología , Regulador Transcripcional ERG/metabolismo
10.
Nutrients ; 14(10)2022 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-35631142

RESUMEN

Prenatal ethanol exposure (PNEE) is a leading cause of neurodevelopmental impairments, yet treatments for individuals with PNEE are limited. Importantly, postnatal supplementation with the essential nutrient choline can attenuate some adverse effects of PNEE on cognitive development; however, the mechanisms of action for choline supplementation remain unclear. This study used an animal model to determine if choline supplementation could restore hippocampal synaptic plasticity that is normally impaired by prenatal alcohol. Throughout gestation, pregnant Sprague Dawley rats were fed an ethanol liquid diet (35.5% ethanol-derived calories). Offspring were injected with choline chloride (100 mg/kg/day) from postnatal days (PD) 10-30, and then used for in vitro electrophysiology experiments as juveniles (PD 31-35). High-frequency conditioning stimuli were used to induce long-term potentiation (LTP) in the medial perforant path input to the dentate gyrus of the hippocampus. PNEE altered synaptic transmission in female offspring by increasing excitability, an effect that was mitigated with choline supplementation. In contrast, PNEE juvenile males had decreased LTP compared to controls, and this was rescued by choline supplementation. These data demonstrate sex-specific changes in plasticity following PNEE, and provide evidence that choline-related improvements in cognitive functioning may be due to its positive impact on hippocampal synaptic physiology.


Asunto(s)
Efectos Tardíos de la Exposición Prenatal , Animales , Colina/farmacología , Suplementos Dietéticos , Etanol , Femenino , Humanos , Masculino , Plasticidad Neuronal , Embarazo , Ratas , Ratas Sprague-Dawley
11.
Reprod Toxicol ; 109: 101-108, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35301062

RESUMEN

This study aimed at determining the effect of prenatal ethanol exposure (PEE) on the expression and activity of cytochrome P450 (CYP) isozymes at different life stages of male rat offspring. Pregnant Wistar rats were administered with ethanol (4 g/kg/d) intragastrically from gestational day (GD) 9-20. Male offspring's gene and activity of CYP isozymes were analyzed on GD 20 (only expression), postnatal day (PD) 84 and 196. Using aniline as probe, we compared the enzyme kinetics of hepatic CYP2E1 between two groups. Expression of CYP isozymes was examined in rat primary hepatocytes and human hepatic cell lines treated with ethanol or/and glucocorticoid. Gene level of Cyp1a2, 2b1, 2d1, 2e1, 3a1 and aryl hydrocarbon receptor were increased in PEE group on GD 20 and PD 84 and Cyp2e1 still exhibited an increasing trend on PD 196 compared with the control. PEE inhibited CYP2D1 and 2E1 activities in male offspring on PD 84. CYP activities in two groups became the same level on PD 196. PEE induced an opposite change in gene and protein level of hepatic CYP2E1 before and after birth. In consistent with lower protein level, aniline metabolism in PEE was weaker in liver microsome. Both single and combined use of ethanol or/and glucocorticoid increased CYPs expression in vitro. In conclusion, PEE programmed a higher gene and lower protein level of CYPs in male offspring, which dwindled with age. Impairment of protein levels and enzyme activities of CYPs may affect individual metabolism of endogenous and exogenous substances in early adulthood.


Asunto(s)
Sistema Enzimático del Citocromo P-450 , Hígado , Animales , Sistema Enzimático del Citocromo P-450/genética , Sistema Enzimático del Citocromo P-450/metabolismo , Etanol/toxicidad , Femenino , Hígado/metabolismo , Masculino , Embarazo , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/farmacología , Ratas , Ratas Wistar
12.
J Toxicol Sci ; 46(9): 413-423, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34470993

RESUMEN

An increased susceptibility to non-alcoholic fatty liver disease (NAFLD) in female rat offspring that experienced prenatal ethanol exposure (PEE) has been previously demonstrated. The present study further investigated the potential mechanism. Based on the results from both fetal and adult studies of offspring rats that experienced PEE (4 g/kg/day), the fetal weight, serum glucose and triglyceride levels decreased significantly and hepatocellular ultra-structure was altered. Fetal livers exhibited inhibited expression and activity of sirtuin 1 (SIRT1), enhanced expression of lipogenic genes: sterol regulatory element binding protein 1c (SREBP1c), fatty acid synthase (FASN), acetyl-coenzyme A carboxylase α (ACCα), stearyl-coenzyme A desaturase 1 (SCD1). In adult offspring fed with high-fat diet, the PEE offspring revealed obviously catch-up growth, increased food intake, elevated serum metabolic phenotypes, suppressed hepatic SIRT1-SREBP1c pathway, and formation of NAFLD. Resveratrol (the chemical activator of SIRT1) could remarkably reverse the serum metabolic phenotypes and alleviate the hepatocyte steatosis in relation to the PEE offspring through activating the hepatic SIRT1-SREBP1c pathway. Therefore, increased susceptibility to diet-induced NAFLD in PEE offspring appears to be mediated by intrauterine programming of hepatic lipogenesis via the SIRT1-SREBP1c pathway. This altered programming effect could partially be reversed by resveratrol intervention after birth in PEE offspring rats.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Animales , Dieta Alta en Grasa , Etanol/toxicidad , Femenino , Hígado , Enfermedad del Hígado Graso no Alcohólico/inducido químicamente , Enfermedad del Hígado Graso no Alcohólico/genética , Embarazo , Ratas , Resveratrol , Sirtuina 1/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética
13.
Toxicol Lett ; 351: 78-88, 2021 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-34454011

RESUMEN

We previously found that prenatal ethanol exposure (PEE) induced adrenal dysplasia in offspring, which was related to intrauterine maternal glucocorticoid overexposure. This study investigated the intergenerational genetic effect and sex differences of PEE-induced changes in the synthetic function of adrenal corticosterone in offspring, and to clarify the intrauterine origin programming mechanism. Wistar pregnant rats were gavaged with ethanol (4 g/kg bw/d) from gestation day (GD) 9-20, and F1 generation was born naturally. The F1 generation female rats in the PEE group were mated with normal male rats to produce F2 generation. Serum and adrenal glands of fetal rats and F1/F2 adult rats were collected at GD20 and postnatal week 28. PEE increased the serum corticosterone level, while diminishing the expression of adrenal steroid synthases of fetal rats. Moreover, PEE enhanced the mRNA expression of GR and HDAC1, but inhibited the mRNA expression of SF1 and reduced the H3K9ac level of P450scc in the fetal adrenal gland. In PEE adult offspring of F1 and F2 generation the serum corticosterone level, the H3K9ac level of P450scc and its expression were decreased in males but were increased in females. In NCI-H295R cells, cortisol reduced the production of endogenous cortisol, down-regulated SF1, and up-regulated HDAC1 expression by activating GR, and decreased H3K9ac level and expression of P450scc. In conclusion, PEE could induce adrenal dysplasia in offspring with sex differences and intergenerational genetic effects, and the adrenal insufficiency in male offspring was related to the induction of low functional genetic programming of P450scc by intrauterine high corticosterone through the GR/SF1/HDAC1 pathway.


Asunto(s)
Glándulas Suprarrenales/efectos de los fármacos , Glándulas Suprarrenales/metabolismo , Corticosterona/biosíntesis , Etanol/toxicidad , Glándulas Suprarrenales/crecimiento & desarrollo , Animales , Línea Celular , Femenino , Desarrollo Fetal/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Hidrocortisona , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal , Factores de Empalme de ARN/genética , Factores de Empalme de ARN/metabolismo , Ratas , Ratas Wistar , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Factores Sexuales , Organismos Libres de Patógenos Específicos
14.
Mol Med Rep ; 23(4)2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33576465

RESUMEN

During embryonic cortical development, radial glial cells (RGCs) are the major source of neurons, and these also serve as a supportive scaffold to guide neuronal migration. Similar to Vimentin, glial fibrillary acidic protein (GFAP) is one of the major intermediate filament proteins present in glial cells. Previous studies confirmed that prenatal ethanol exposure (PEE) significantly affected the levels of GFAP and increased the disassembly of radial glial fibers. GFAPδ is a variant of GFAP that is specifically expressed in RGCs; however, to the best of our knowledge, there are no reports regarding how PEE influences its expression during cortical development. In the present study, the effects of PEE on the expression and distribution of GFAPδ during early cortical development were assessed. It was found that PEE significantly decreased the expression levels of GFAP and GFAPδ. Using double immunostaining, GFAPδ was identified to be specifically expressed in apical and basal RGCs, and was co­localized with other intermediate filament proteins, such as GFAP, Nestin and Vimentin. Additionally, PEE significantly affected the morphology of radial glial fibers and altered the behavior of RGCs. The loss of GFAPδ accelerated the transformation of RGCs into astrocytes. Using co­immunostaining with Ki67 or phospho­histone H3, GFAPδ+ cells were observed to be proliferative or mitotic cells, and ethanol treatment significantly decreased the proliferative or mitotic activities of GFAPδ+ RGCs. Taken together, the results suggested that PEE altered the expression patterns of GFAPδ and impaired the development of radial glial fibers and RGC behavior. The results of the present study provided evidence that GFAPδ may be a promising target to rescue the damage induced by PEE.


Asunto(s)
Astrocitos/efectos de los fármacos , Encéfalo/efectos de los fármacos , Células Ependimogliales/efectos de los fármacos , Etanol/farmacología , Proteína Ácida Fibrilar de la Glía/metabolismo , Animales , Astrocitos/citología , Astrocitos/metabolismo , Encéfalo/embriología , Encéfalo/metabolismo , Proliferación Celular/efectos de los fármacos , Células Ependimogliales/citología , Células Ependimogliales/metabolismo , Femenino , Exposición Materna , Ratones Endogámicos C57BL , Nestina/metabolismo , Neurogénesis/efectos de los fármacos , Embarazo , Vimentina/metabolismo
15.
Reprod Toxicol ; 96: 36-46, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32497709

RESUMEN

Epidemiological investigations have found that maternal alcohol intake increases the risk of mental illness in offspring. Our study investigated changes of depression- and anxiety-like behaviors in adult offspring caused by prenatal ethanol exposure (PEE) and explored the potential mechanism. After Wistar rats were intragastrically administered ethanol at a dose of 4 g/kg·d on the 9-20 t h days of pregnancy, the offspring were given 21 days of chronic unpredictable mild stress (CUMS) starting from the 9th week after birth. Before CUMS, the behavioral results showed that the PEE offspring appeared excited and anxious. After CUMS, the PEE offspring rats were more sensitive to the same intensity of stimulation, and then the behavioral disorders aggravated. In adult offspring from the PEE group, the intercellular space was enlarged in the hippocampus, and there was a loss of pyramidal cells. The expression of brain-derived neurotrophic factor (BDNF) decreased; the mRNA expression of the glucocorticoid receptor and synaptic plasticity-related genes decreased; the apoptosis-related genes expressed disrupted. In order to determine whether hippocampal injury and dysfunction resulted from ethanol directly or indirectly, we performed in vitro study. The outcome was accompanied by disrupted gene expression related to neurogenesis and synaptic plasticity. PEE increases the susceptibility of adult female offspring to depression- and anxiety-like behaviors, and its mechanism may be related to the toxic effects of ethanol, both directly and indirectly. The latter inhibits the hippocampal BDNF pathway, leading to the disruption of hippocampal neurogenesis, apoptosis and decreased synaptic plasticity.


Asunto(s)
Consumo de Bebidas Alcohólicas/efectos adversos , Ansiedad/inducido químicamente , Depresión/inducido químicamente , Etanol/toxicidad , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Animales , Ansiedad/genética , Ansiedad/metabolismo , Conducta Animal/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/sangre , Factor Neurotrófico Derivado del Encéfalo/genética , Línea Celular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Depresión/genética , Depresión/metabolismo , Femenino , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Intercambio Materno-Fetal , Embarazo , Efectos Tardíos de la Exposición Prenatal/genética , Efectos Tardíos de la Exposición Prenatal/metabolismo , Ratas Wistar , Receptor trkB/genética , Receptores de Glucocorticoides/genética , Receptores de Mineralocorticoides/genética
16.
Toxicol Lett ; 331: 167-177, 2020 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-32535229

RESUMEN

Prenatal ethanol exposure (PEE) could increase offspring's susceptibility to adult liver lipid-metabolism diseases. This study aimed to confirm intrauterine programming mechanism of glucocorticoid-insulin-like growth factor 1 (GC-IGF1) axis for liver dysfunction in offspring rats induced by PEE. The results showed that levels of hepatic IGF1, lipid metabolism-related enzymes (e.g. FASN and HMGCR) and serum phenotype (TG, TCH, HDL-C, and LDL-C) were low in fetal rats of PEE but high in adult offspring except for HDL-C, meanwhile, hepatic H3K9ac and expression levels of IGF1 were low in fetal rats but high in adult offspring. Furthermore, levels of serum corticosterone and hepatic glucocorticoid-activation system (mainly including expression of 11ß-HSD1, GR, and C/EBPα as well as 11ß-HSD1/11ß-HSD2 ratio) were high in fetal rats of PEE but low or unchanged in adult offspring. The adult F2 generation of PEE maintained the same GC-IGF1 axis programming alteration as the F1 generation despite gender differences. In vitro, cortisol was proved to activate hepatocyte glucocorticoid-activation system and decrease H3K9ac and expression levels of IGF1 by GR. Therefore, PEE has a long-term effect on the offspring's liver functional development, which may be mainly related to the epigenetic programming alteration of the GC-IGF1 axis mediated by the glucocorticoid-activation system.


Asunto(s)
Etanol/toxicidad , Glucocorticoides/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal/metabolismo , Animales , Peso Corporal , Corticosterona/sangre , Femenino , Retardo del Crecimiento Fetal/sangre , Retardo del Crecimiento Fetal/inducido químicamente , Retardo del Crecimiento Fetal/metabolismo , Células Hep G2 , Humanos , Lípidos/sangre , Hígado/embriología , Hígado/metabolismo , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal/sangre , Ratas Wistar , Transducción de Señal
17.
Reprod Toxicol ; 94: 48-54, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32333956

RESUMEN

This study was designed to demonstrate disorder of hypothalamic-pituitary-adrenal (HPA) axis-associated neuroendocrine metabolic programming alteration and dysfunction of glucose and lipid metabolism induced by prenatal ethanol exposure (PEE) in postnatal week 40 (PW40) female offspring rats. Pregnant Wistar rats were administrated 4  g/kg·d ethanol intragastrically from gestational day 11 until term delivery. After weaning, the female offspring were fed with high-fat diet until PW24, and suffered to unpredictable chronic stress (UCS) during PW38-40. Animal serum was collected to examine the changes in hypothalamic-pituitary-adrenal (HPA) axis activity, glucose and lipid metabolic phenotypes before and after UCS. We found that pups in the PEE group manifested a low birthweight at PW1 and an early catch-up growth pattern. Furthermore, a low basal activity of HPA axis continued to PW38 in the PEE group. On the basal condition, serum low-density lipoprotein-cholesterol (LDL-C) level was significantly increased and high-density lipoprotein-cholesterol (HDL-C) level was significantly decreased in the PEE group, while serum triglyceride, total cholesterol (TCH), glucose and insulin levels were not significantly changed. Under unpredictable chronic stress, serum insulin in the PEE group was significantly decreased, while the levels of serum triglyceride, TCH, LDL-C, and the ratio of LDL-C/HDL-C were significantly higher than those in the control. These results suggest that PEE increases the dysfunction of glucose and lipid metabolism in PW40 female offspring, which is related to the disorder of HPA axis-associated neuroendocrine metabolic programming alteration.


Asunto(s)
Etanol/toxicidad , Glucosa/metabolismo , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal , Animales , Femenino , Intercambio Materno-Fetal , Embarazo , Ratas Wistar , Estrés Fisiológico , Estrés Psicológico/metabolismo
18.
Alcohol Clin Exp Res ; 44(4): 866-879, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32020622

RESUMEN

BACKGROUND: Prenatal exposure to ethanol (EtOH) has lasting effects on neuropeptide and neuroimmune systems in the brain alongside detrimental alcohol-related behaviors. At low-to-moderate doses, prenatal EtOH stimulates neurogenesis in lateral hypothalamus (LH) and increases neurons that express the orexigenic peptides hypocretin/orexin (Hcrt/OX) and melanin-concentrating hormone (MCH), and the proinflammatory chemokine CCL2, which through its receptor CCR2 stimulates cell differentiation and movement. Our recent studies demonstrated that CCL2 and CCR2 colocalize with MCH neurons and are involved in EtOH's stimulatory effect on their development but show no relation to Hcrt/OX. Here, we investigated another chemokine, CXCL12, and its receptor, CXCR4, which promote neurogenesis and neuroprogenitor cell proliferation, to determine if they also exhibit peptide specificity in their response to EtOH exposure. METHODS: Pregnant rats were intraorally administered a moderate dose of EtOH (2 g/kg/d) from embryonic day 10 (E10) to E15. Their embryos and postnatal offspring were examined using real-time quantitative PCR and immunofluorescence histochemistry, to determine if EtOH affects CXCL12 and CXCR4 and the colocalization of CXCR4 with Hcrt/OX and MCH neurons in the LH and with radial glia neuroprogenitor cells in the hypothalamic neuroepithelium (NEP). RESULTS: Prenatal EtOH strongly stimulated CXCL12 and CXCR4 in LH neurons of embryos and postnatal offspring. This stimulation was significantly stronger in Hcrt/OX than MCH neurons in LH and also occurred in radial glia neuroprogenitor cells dense in the NEP. These effects were sexually dimorphic, consistently stronger in females than males. CONCLUSIONS: While showing prenatal EtOH exposure to have a sexually dimorphic, stimulatory effect on CXCL12 and CXCR4 in LH similar to CCL2 and its receptor, these results reveal their distinct relationship to the peptide neurons, with the former closely related to Hcrt/OX and the latter to MCH, and they link EtOH's actions in LH to a stimulatory effect on neuroprogenitor cells in the NEP.


Asunto(s)
Depresores del Sistema Nervioso Central/farmacología , Quimiocina CXCL12/efectos de los fármacos , Células Ependimogliales/efectos de los fármacos , Etanol/farmacología , Área Hipotalámica Lateral/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Neuronas/efectos de los fármacos , Receptores CXCR4/efectos de los fármacos , Animales , Animales Recién Nacidos , Proliferación Celular/efectos de los fármacos , Quimiocina CXCL12/metabolismo , Embrión de Mamíferos , Células Ependimogliales/metabolismo , Área Hipotalámica Lateral/citología , Área Hipotalámica Lateral/metabolismo , Hormonas Hipotalámicas/metabolismo , Hipotálamo/citología , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Inmunohistoquímica , Melaninas/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis/efectos de los fármacos , Neuronas/metabolismo , Orexinas/metabolismo , Hormonas Hipofisarias/metabolismo , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores CXCR4/metabolismo
19.
Front Neurosci ; 14: 12, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32038156

RESUMEN

BACKGROUND: Prenatal ethanol exposure (PE) causes multiple behavioral and cognitive deficits, collectively referred to as fetal alcohol spectrum disorders (FASD). Studies show that 49-94% of FASD children exhibit attention deficits, even when they have normal IQs or lack severe facial deformities, suggesting that attention deficits could be caused by even moderate prenatal exposure to alcohol, of which the underlying neural mechanisms are still unclear. A valid rodent model could help elucidate this phenomenon. MATERIALS AND METHODS: A second-trimester equivalent binge drinking PE model was utilized. Pregnant Sprague Dawley rats were administered with 15% (w/v) ethanol (6 g/kg/day, via gastric gavage) during gestational days 8-20, and their offspring were the subjects in the present study. A modified 2-choice reaction time (2-CRT) task was used to illustrate possible attention deficits, including increased action impulsivity and lapses of attention. Enhanced impulsivity was reflected by more premature responses while increased lapses of attention were manifested as more incorrect responses and/or greater variability of reaction time, demonstrated by more skewed distributions of reaction time. Ten-week-old male and female rats were tested for three sessions following 16-19 days of training. RESULTS: Our PE paradigm caused no major teratogenic effects. PE led to increased impulsivity exhibited as greater premature responses and augmented lapses of attention shown by greater skewnesses of reaction time distributions, relative to controls. The deficits were observed in both PE male and female rats. Interestingly, in males, the attention deficits were detected only when the 2-CRT task was relatively difficult whereas in females they were detected even when the task was at a less demanding level. CONCLUSION: We show that the binge drinking pattern of PE led to attention deficits in both sexes of rats even though no major teratogenic effects were observed. Therefore, this rodent model can be used to study neural mechanisms underlying attention deficits caused by PE and to explore effective intervention approaches for FASD.

20.
Neuropharmacology ; 165: 107917, 2020 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-31926456

RESUMEN

Prenatal ethanol exposure (PEE) promotes ethanol consumption in the adolescent offspring accompanied by the transcriptional regulation of kappa opioid receptor (KOR) system genes. This study analysed if environmental enrichment (EE, from gestational day 20 to postnatal day 26) exerts protective effects upon PEE-modulation of gene expression, ethanol intake and anxiety responses. Pregnant rats were exposed to PEE (0.0 or 2.0 g/kg ethanol, gestational days 17-20) and subsequently the dam and offspring were reared under EE or standard conditions. PEE upregulated KOR mRNA levels in amygdala (AMY) and prodynorphin (PDYN) mRNA levels in ventral tegmental area (VTA) with the latter effect associated with lower DNA methylation at the gene promoter. These effects were normalized by exposure to EE. PEE modulated BDNF mRNA levels in VTA and Nucleus accumbens (AcbN), and EE mitigated the changes in AcbN. EE induced a protective effect on ethanol intake and preference, an effect more noticeable in males than in females, and in prenatal vehicle-treated (PV) than in PEE rats. The male offspring drank significantly less ethanol than the female offspring. The latter result suggests that the protective effect of EE on ethanol drinking may only emerge at lower levels of drinking. In the dams, PEE induced an upregulation of PDYN and KOR in AcbN. PDYN gene expression was normalized by exposure to EE. These results suggest that EE is a promising treatment to inhibit the effects of PEE. The results confirm that PEE effects are mediated by alterations in the transcriptional regulation of KOR system genes.


Asunto(s)
Consumo de Bebidas Alcohólicas , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Ambiente , Etanol/administración & dosificación , Regulación de la Expresión Génica , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Receptores Opioides kappa/metabolismo , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/metabolismo , Animales , Ansiedad/inducido químicamente , Encefalinas/metabolismo , Femenino , Masculino , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Precursores de Proteínas/metabolismo , ARN Mensajero/metabolismo , Ratas Wistar , Área Tegmental Ventral/efectos de los fármacos , Área Tegmental Ventral/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA