Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.696
Filter
1.
Int J Mol Sci ; 25(13)2024 Jul 03.
Article in English | MEDLINE | ID: mdl-39000422

ABSTRACT

Maternal obesity, caused by diets rich in fats and sugars during pregnancy, can predispose offspring to metabolic diseases such as diabetes. We hypothesized that obesity during pregnancy leads to increased DNA methylation and reduced protein expression in factors regulating ß-cell function and apoptosis. Female C57BL/6J mice were fed a high-fat diet (HFD; 42% fat content; n = 3) or a control diet (CON; 16% fat content; n = 3) for fourteen weeks before and during pregnancy. Offspring were euthanized at 8 weeks and pancreatic tissue was collected. Isolated DNA was analyzed using whole-genome bisulfite sequencing. Protein expression was quantified using LC-MS. No significant differences in body weight were observed between HFD and control pups (p = 0.10). Whole-genome bisulfite sequencing identified 91,703 and 88,415 differentially methylated regions (DMRs) in CON vs. HFD male and female offspring. A total of 34 and 4 proteins were determined to have changes in expression that correlated with changes in DNA methylation in CON vs. HFD males and females, respectively. The majority of these factors were grouped into the metabolic function category via pathway analyses. This study illustrates the complex relationship between epigenetics, diet, and sex-specific responses, therefore offering insights into potential therapeutic targets and areas for further research.


Subject(s)
DNA Methylation , Diet, High-Fat , Mice, Inbred C57BL , Pancreas , Animals , Female , Diet, High-Fat/adverse effects , Pregnancy , Mice , Male , Pancreas/metabolism , Prenatal Exposure Delayed Effects/genetics , Prenatal Exposure Delayed Effects/metabolism , Obesity/metabolism , Obesity/genetics , Obesity/etiology , Epigenesis, Genetic , Multiomics
2.
Int J Mol Sci ; 25(13)2024 Jun 27.
Article in English | MEDLINE | ID: mdl-39000127

ABSTRACT

The prevalence of prenatal alcohol exposure (PAE) is increasing, with evidence suggesting that PAE is linked to an increased risk of infections. PAE is hypothesized to affect the innate immune system, which identifies pathogens through pattern recognition receptors, of which toll-like receptors (TLRs) are key components. We hypothesized that light-to-moderate PAE would impair immune responses, as measured by a heightened response in cytokine levels following TLR stimulation. Umbilical cord samples (10 controls and 8 PAE) from a subset of the Ethanol, Neurodevelopment, Infant and Child Health Study-2 cohort were included. Peripheral blood mononuclear cells (PMBCs) were stimulated with one agonist (TLR2, TLR3, TLR4, or TLR9). TLR2 agonist stimulation significantly increased pro-inflammatory interleukin-1-beta in the PAE group after 24 h. Pro- and anti-inflammatory cytokines were increased following stimulation with the TLR2 agonists. Stimulation with TLR3 or TLR9 agonists displayed minimal impact overall, but there were significant increases in the percent change of the control compared to PAE after 24 h. The results of this pilot investigation support further work into the impact on TLR2 and TLR4 response following PAE to delineate if alterations in levels of pro- and anti-inflammatory cytokines have clinical significance that could be used in patient management and/or attention to follow-up.


Subject(s)
Fetal Blood , Toll-Like Receptors , Humans , Female , Pregnancy , Fetal Blood/metabolism , Pilot Projects , Toll-Like Receptors/metabolism , Toll-Like Receptors/agonists , Cytokines/metabolism , Cytokines/blood , Adult , Infant, Newborn , Prenatal Exposure Delayed Effects/metabolism , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/drug effects , Male , Ethanol/pharmacology , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 2/agonists
3.
Int J Mol Sci ; 25(13)2024 Jun 27.
Article in English | MEDLINE | ID: mdl-39000130

ABSTRACT

Prenatal stress (PNS), which alters the hypothalamic-pituitary-adrenal axis function in the offspring, predisposes to insulin resistance (IR) in later life and is associated with numerous disorders, including cognitive and memory impairments. At present, our main goal is to assess the effects of chronic piromelatine (Pir) administration, a melatonin analogue, on PNS-provoked IR in the periphery and the hippocampus in male and female offspring. Pregnant Sprague-Dawley rats were exposed to chronic stress (one short-term stressor on a daily basis and one long-term stressor on a nightly basis) from the first gestation week until birth. Vehicle or Pir 20 mg/kg were administered intraperitoneally for 21 days. Plasma glucose, serum insulin levels, and the homeostasis model assessment of insulin resistance (HOMA-IR) were determined as markers of peripheral IR. For the hippocampal IR assessment, insulin receptors (IRs) and glucose transporter 4 (GLUT4) were examined. Prenatally stressed offspring of both sexes indicated enhanced plasma glucose and serum insulin concentrations, increased HOMA-IR, and decreased hippocampal GLUT4 only in male rats. The PNS-induced changes were corrected by chronic treatment with Pir. The present results suggest that the melatoninergic compound Pir exerts beneficial effects on altered glucose/insulin homeostasis in PNS-exposed offspring.


Subject(s)
Hippocampus , Insulin Resistance , Insulin , Prenatal Exposure Delayed Effects , Rats, Sprague-Dawley , Animals , Hippocampus/metabolism , Hippocampus/drug effects , Female , Pregnancy , Male , Rats , Prenatal Exposure Delayed Effects/metabolism , Insulin/metabolism , Insulin/blood , Blood Glucose/metabolism , Stress, Psychological/metabolism , Glucose Transporter Type 4/metabolism , Receptor, Insulin/metabolism , Melatonin/pharmacology
4.
Bull Exp Biol Med ; 177(1): 10-14, 2024 May.
Article in English | MEDLINE | ID: mdl-38954295

ABSTRACT

Spatial learning, memory, and reactivity of the hypothalamic-pituitary-adrenocortical system (HPA axis) were studied in adult male rats, whose mothers during pregnancy were subjected to acute moderate normobaric hypoxia, or repeated injections of buspirone, an agonist of type 1A serotonergic receptors (5HT1A), or their combination. Prenatal treatment with buspirone in rats with prenatal hypoxia impaired learning ability during the first day of 5-day training. A decrease in the effectiveness of long-term memory in comparison with short-term memory was revealed in two groups of rats: prenatal treatment with buspirone in combination with hypoxia and injection of physiological saline without hypoxia. The effectiveness of long-term memory and the level of corticosterone in response to stress did not differ between the groups, which can indicate adaptation of the 5HT1A receptor and the HPA axis to the prenatal buspirone and normobaric hypoxia during ontogeny.


Subject(s)
Buspirone , Hypothalamo-Hypophyseal System , Hypoxia , Prenatal Exposure Delayed Effects , Buspirone/pharmacology , Animals , Pregnancy , Female , Rats , Male , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/physiopathology , Hypoxia/physiopathology , Hypoxia/metabolism , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Corticosterone/blood , Corticosterone/metabolism , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/metabolism , Cognition/drug effects , Cognition/physiology , Rats, Wistar , Receptor, Serotonin, 5-HT1A/metabolism , Maze Learning/drug effects , Memory, Long-Term/drug effects , Memory, Long-Term/physiology , Stress, Physiological/drug effects
5.
Brain Behav ; 14(7): e3614, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38988101

ABSTRACT

PURPOSE: Levothyroxine (LEV) monotherapy cannot completely improve cognitive and behavioral impairments induced by hypothyroidism, whereas a combination therapy of exercise and LEV may ameliorate these deficits. This study aimed to determine the effects of mild-intensity forced exercise and LEV treatment on the anxiety profile and cognitive functions in male offspring of hypothyroid dams. METHOD: Twenty-four female rats (mothers) were randomly divided into sham (healthy) and hypothyroidism groups and then placed with male rats to mate. The presence of vaginal plaque confirmed pregnancy (gestational day, GD 0). 6-propyl-2-thiouracil (PTU, 100 ppm) was added to the drinking water of the hypothyroidism group from GD 6 to the 21st postnatal day (PND). The sham group received tap water. On PND 21, serum T4 levels of mothers, and 10 pups were measured to confirm hypothyroidism. Sixty-four male pups were left undisturbed for 30 days and then were divided into eight groups that received saline or LEV (50 µg/kg, i.p.) with or without forced mild-intensity exercise. After 14 days of interventions, anxiety-like behaviors, spatial learning and memory, and hippocampal brain-derived neurotrophic factor (BDNF) levels were evaluated. FINDING: A pre and postnatal PTU-induced model of hypothyroidism increased anxiety-like behaviors, impaired spatial learning and memory, and decreased hippocampal BDNF levels in male offspring rats. LEV alone increased BDNF levels and improved spatial learning. Exercise alone increased BDNF levels, improved spatial learning and memory, and decreased anxiety-like behaviors. Exercise plus LEV more effectively improved anxiety-like behaviors and spatial learning than exercise or LEV alone. CONCLUSION: Practically, these pre-clinical findings highlight the importance of the combination of exercise and LEV regimen in treating patients with hyperthyroidism.


Subject(s)
Anxiety , Brain-Derived Neurotrophic Factor , Hippocampus , Hypothyroidism , Physical Conditioning, Animal , Thyroxine , Animals , Hypothyroidism/drug therapy , Hypothyroidism/metabolism , Hypothyroidism/therapy , Hypothyroidism/physiopathology , Brain-Derived Neurotrophic Factor/metabolism , Male , Thyroxine/pharmacology , Thyroxine/administration & dosage , Rats , Anxiety/therapy , Anxiety/etiology , Anxiety/drug therapy , Hippocampus/metabolism , Hippocampus/drug effects , Female , Physical Conditioning, Animal/physiology , Pregnancy , Rats, Wistar , Prenatal Exposure Delayed Effects/therapy , Prenatal Exposure Delayed Effects/metabolism , Spatial Learning/drug effects , Spatial Learning/physiology , Combined Modality Therapy , Propylthiouracil/pharmacology , Propylthiouracil/administration & dosage
6.
Toxicol Appl Pharmacol ; 489: 117019, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38950736

ABSTRACT

Maternal hypoxia is strongly linked to insulin resistance (IR) in adult offspring, and altered insulin signaling for muscle glucose uptake is thought to play a central role. However, whether the SIRT3/GSK-3ß/GLUT4 axis is involved in maternal hypoxia-induced skeletal muscle IR in old male rat offspring has not been investigated. Maternal hypoxia was established from Days 5 to 21 of pregnancy by continuous infusion of nitrogen and air. The biochemical parameters and levels of key insulin signaling molecules of old male rat offspring were determined through a series of experiments. Compared to the control (Ctrl) old male rat offspring group, the hypoxic (HY) group exhibited elevated fasting blood glucose (FBG) (∼30%), fasting blood insulin (FBI) (∼35%), total triglycerides (TGs), and low-density lipoprotein cholesterol (LDL-C), as well as results showing impairment in the glucose tolerance test (GTT) and insulin tolerance test (ITT). In addition, hematoxylin-eosin (HE) staining and transmission electron microscopy (TEM) revealed impaired cellular structures and mitochondria in the longitudinal sections of skeletal muscle from HY group mice, which might be associated with decreased SIRT3 expression. Furthermore, the expression of insulin signaling molecules, such as GSK-3ß and GLUT4, was also altered. In conclusion, the present results indicate that the SIRT3/GSK-3ß/GLUT4 axis might be involved in maternal hypoxia-induced skeletal muscle IR in old male rat offspring.


Subject(s)
Glucose Transporter Type 4 , Glycogen Synthase Kinase 3 beta , Hypoxia , Insulin Resistance , Muscle, Skeletal , Sirtuin 3 , Animals , Male , Glycogen Synthase Kinase 3 beta/metabolism , Insulin Resistance/physiology , Muscle, Skeletal/metabolism , Female , Glucose Transporter Type 4/metabolism , Pregnancy , Sirtuin 3/metabolism , Rats , Hypoxia/metabolism , Signal Transduction , Prenatal Exposure Delayed Effects/metabolism , Rats, Sprague-Dawley , Insulin/blood , Insulin/metabolism , Blood Glucose/metabolism , Sirtuins
7.
Birth Defects Res ; 116(6): e2369, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38877673

ABSTRACT

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.


Subject(s)
Ethanol , Mice, Inbred C57BL , Myocytes, Cardiac , Prenatal Exposure Delayed Effects , Reactive Oxygen Species , Animals , Female , Pregnancy , Mice , Prenatal Exposure Delayed Effects/metabolism , Reactive Oxygen Species/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Ethanol/adverse effects , Ethanol/toxicity , DNA, Mitochondrial/drug effects , DNA, Mitochondrial/metabolism , Apoptosis/drug effects , Mitochondria, Heart/metabolism , Mitochondria, Heart/drug effects , Membrane Potential, Mitochondrial/drug effects , Myocardium/metabolism , Myocardium/pathology , Oxidative Stress/drug effects , Fetal Alcohol Spectrum Disorders/metabolism , Fetal Alcohol Spectrum Disorders/pathology , Mitochondria/drug effects , Mitochondria/metabolism , Echocardiography , Fibrosis
8.
FASEB J ; 38(12): e23736, 2024 Jun 30.
Article in English | MEDLINE | ID: mdl-38865202

ABSTRACT

Subclinical hypothyroidism (SCH) in pregnancy is the most common form of thyroid dysfunction in pregnancy, which can affect fetal nervous system development and increase the risk of neurodevelopmental disorders after birth. However, the mechanism of the effect of maternal subclinical hypothyroidism on fetal brain development and behavioral phenotypes is still unclear and requires further study. In this study, we constructed a mouse model of maternal subclinical hypothyroidism by exposing dams to drinking water containing 50 ppm propylthiouracil (PTU) during pregnancy and found that its offspring were accompanied by severe cognitive deficits by behavioral testing. Mechanistically, gestational SCH resulted in the upregulation of protein expression and activity of HDAC1/2/3 in the hippocampus of the offspring. ChIP analysis revealed that H3K9ac on the neurogranin (Ng) promoter was reduced in the hippocampus of the offspring of SCH, with a significant reduction in Ng protein, leading to reduced expression levels of synaptic plasticity markers PSD95 (a membrane-associated protein in the postsynaptic density) and SYN (synaptophysin, a specific marker for presynaptic terminals), and impaired synaptic plasticity. In addition, administration of MS-275 (an HDAC1/2/3-specific inhibitor) to SCH offspring alleviated impaired synaptic plasticity and cognitive dysfunction in offspring. Thus, our study suggests that maternal subclinical hypothyroidism may mediate offspring cognitive dysfunction through the HDAC1/2/3-H3K9ac-Ng pathway. Our study contributes to the understanding of the signaling mechanisms underlying maternal subclinical hypothyroidism-mediated cognitive impairment in the offspring.


Subject(s)
Cognitive Dysfunction , Histone Deacetylase 1 , Histone Deacetylase 2 , Hypothyroidism , Neurogranin , Prenatal Exposure Delayed Effects , Animals , Neurogranin/metabolism , Neurogranin/genetics , Hypothyroidism/metabolism , Female , Pregnancy , Mice , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/etiology , Histone Deacetylase 2/metabolism , Histone Deacetylase 2/genetics , Prenatal Exposure Delayed Effects/metabolism , Histone Deacetylase 1/metabolism , Histone Deacetylase 1/genetics , Down-Regulation , Hippocampus/metabolism , Male , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Mice, Inbred C57BL , Neuronal Plasticity
9.
Brain Behav ; 14(6): e3539, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38849974

ABSTRACT

BACKGROUND AND OBJECTIVES: Maternal hypoxia disrupts neural development and subsequently leads to cerebral palsy and epilepsy in newborns. Hypoxia plays a role in neurodegeneration by increasing oxidative stress. Pistacia atlantica is known as an important antioxidant, and its anti-inflammatory and antioxidant effects have been shown in various studies. This study aims to investigate the effects of methanolic extract of P. atlantica leaves (MEPaLs) on the oxidative parameters in the serum of rats affected by maternal hypoxia. MATERIAL AND METHODS: In this study, eight pregnant rats were used. The newborns were divided into four groups, including the control and the hypoxia groups, which are affected by maternal hypoxia, hypoxia + MEPaL 100 mg/kg, and hypoxia + MEPaL 150 mg/kg. MEPaL was injected (i.p) for 21 days into the neonatal rats after the lactation period. Hypoxia was induced by keeping pregnant rats in a hypoxic chamber with 7% oxygen and 93% nitrogen intensity for 3 h on the 20th day of pregnancy. Behavioral changes were measured using open-field and rotarod tests. Finally, biomarkers of oxidative stress, nitric oxide (NO), glutathione (GSH), GSSG, TAS, TOS, and oxidative stress index (OSI) were measured in the experimental groups. RESULTS: Behavioral results showed that the anxiety behavior in the hypoxia group increased, but the motor activity (moved distance and movement speed) decreased. Moreover, the amount of time spent maintaining balance on the rotarod rod was significantly decreased in the hypoxia group. The concentration of NO in the group of hypoxia + MEPaL 100 mg/kg showed a significant decrease, and MEPaL 100, and 150 mg/kg + hypoxia also increased the concentration of GSH and decreased GSSG. In addition, MEPaL100 and 150 mg/kg caused a significant increase in the ratio of GSH to GSSG and decreased OSI and total oxidant capacity. CONCLUSIONS: Oxidative stress increased in the rats affected by maternal hypoxia and may be the main mechanism for motor activity impairment and balance disturbance, whereas MELaL improved motor performance by decreasing oxidative stress.


Subject(s)
Antioxidants , Oxidative Stress , Plant Extracts , Animals , Oxidative Stress/drug effects , Female , Pregnancy , Rats , Antioxidants/pharmacology , Plant Extracts/pharmacology , Plant Extracts/administration & dosage , Hypoxia/physiopathology , Rats, Wistar , Animals, Newborn , Prenatal Exposure Delayed Effects/physiopathology , Prenatal Exposure Delayed Effects/metabolism , Motor Activity/drug effects , Motor Activity/physiology , Glutathione/metabolism , Glutathione/blood , Male , Behavior, Animal/drug effects , Behavior, Animal/physiology , Nitric Oxide/metabolism , Nitric Oxide/blood
10.
PLoS Biol ; 22(6): e3002641, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38833481

ABSTRACT

In utero exposure to maternal obesity programs increased obesity risk. Animal models show that programmed offspring obesity is preceded by hyperphagia, but the mechanisms that mediate these changes are unknown. Using a mouse model of maternal obesity, we observed increased intake of a high-fat diet (HFD) in offspring of obese mothers that precedes the development of obesity. Through small RNA sequencing, we identified programmed overexpression of hypothalamic miR-505-5p that is established in the fetus, lasts to adulthood and is maintained in hypothalamic neural progenitor cells cultured in vitro. Metabolic hormones and long-chain fatty acids associated with obesity increase miR-505-5p expression in hypothalamic neurons in vitro. We demonstrate that targets of miR-505-5p are enriched in fatty acid metabolism pathways and overexpression of miR-505-5p decreased neuronal fatty acid metabolism in vitro. miR-505-5p targets are associated with increased BMI in human genetic studies. Intra-cerebroventricular injection of miR-505-5p in wild-type mice increased HFD intake, mimicking the phenotype observed in offspring exposed to maternal obesity. Conversely, maternal exercise intervention in an obese mouse pregnancy rescued the programmed increase of hypothalamic miR-505-5p in offspring of obese dams and reduced HFD intake to control offspring levels. This study identifies a novel mechanism by which maternal obesity programs obesity in offspring via increased intake of high-fat foods.


Subject(s)
Diet, High-Fat , Fatty Acids , Hypothalamus , MicroRNAs , Obesity, Maternal , Animals , Female , Humans , Male , Mice , Pregnancy , Diet, High-Fat/adverse effects , Fatty Acids/metabolism , Hypothalamus/metabolism , Mice, Inbred C57BL , MicroRNAs/metabolism , MicroRNAs/genetics , Neurons/metabolism , Obesity/metabolism , Obesity/genetics , Obesity, Maternal/metabolism , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/genetics
11.
Int J Mol Sci ; 25(11)2024 May 28.
Article in English | MEDLINE | ID: mdl-38892090

ABSTRACT

Fetal hypoxia and maternal stress frequently culminate in neuropsychiatric afflictions in life. To replicate this condition, we employed a model of prenatal severe hypoxia (PSH) during days 14-16 of rat gestation. Subsequently, both control and PSH rats at 3 months old were subjected to episodes of inescapable stress to induce learned helplessness (LH). The results of the open field test revealed an inclination towards depressive-like behavior in PSH rats. Following LH episodes, control (but not PSH) rats displayed significant anxiety. LH induced an increase in glucocorticoid receptor (GR) levels in extrahypothalamic brain structures, with enhanced nuclear translocation in the hippocampus (HPC) observed both in control and PSH rats. However, only control rats showed an increase in GR nuclear translocation in the amygdala (AMG). The decreased GR levels in the HPC of PSH rats correlated with elevated levels of hypothalamic corticotropin-releasing hormone (CRH) compared with the controls. However, LH resulted in a reduction of the CRH levels in PSH rats, aligning them with those of control rats, without affecting the latter. This study presents evidence that PSH leads to depressive-like behavior in rats, associated with alterations in the glucocorticoid system. Notably, these impairments also contribute to increased resistance to severe stressors.


Subject(s)
Anxiety , Depression , Glucocorticoids , Prenatal Exposure Delayed Effects , Receptors, Glucocorticoid , Animals , Rats , Female , Anxiety/metabolism , Pregnancy , Glucocorticoids/metabolism , Depression/metabolism , Depression/etiology , Receptors, Glucocorticoid/metabolism , Prenatal Exposure Delayed Effects/metabolism , Stress, Psychological/metabolism , Male , Corticotropin-Releasing Hormone/metabolism , Hippocampus/metabolism , Hypoxia/metabolism , Phenotype , Behavior, Animal , Helplessness, Learned , Disease Models, Animal , Amygdala/metabolism , Fetal Hypoxia/metabolism , Fetal Hypoxia/complications
12.
Int J Mol Sci ; 25(11)2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38892402

ABSTRACT

In day-to-day living, individuals are exposed to various environmentally hazardous substances that have been associated with diverse diseases. Exposure to air pollutants can occur during breathing, posing a considerable risk to those with environmental health vulnerabilities. Among vulnerable individuals, maternal exposure can negatively impact the mother and child in utero. The developing fetus is particularly vulnerable to environmentally hazardous substances, with potentially greater implications. Among air pollutants, toluene is neurotoxic, and its effects have been widely explored. However, the impact of low-level toluene exposure in daily life remains unclear. Herein, we evaluated 194 mothers and infants from the Growing children's health and Evaluation of Environment (GREEN) cohort to determine the possible effects of early-life toluene exposure on the nervous system. Using Omics experiments, the effects of toluene were confirmed based on epigenetic changes and altered mRNA expression. Various epigenetic changes were identified, with upregulated expression potentially contributing to diseases such as glioblastoma and Alzheimer's, and downregulated expression being associated with structural neuronal abnormalities. These findings were detected in both maternal and infant groups, suggesting that maternal exposure to environmental hazardous substances can negatively impact the fetus. Our findings will facilitate the establishment of environmental health policies, including the management of environmentally hazardous substances for vulnerable groups.


Subject(s)
Maternal Exposure , Toluene , Humans , Toluene/toxicity , Female , Infant , Maternal Exposure/adverse effects , Pregnancy , Adult , Nervous System/drug effects , Nervous System/embryology , Nervous System/metabolism , Nervous System/growth & development , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/metabolism , Epigenesis, Genetic/drug effects , Male , Mothers , Air Pollutants/toxicity , Infant, Newborn
13.
Psychoneuroendocrinology ; 167: 107088, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38924829

ABSTRACT

BACKGROUND: Changes in NR3C1 and IGF2/H19 methylation patterns have been associated with behavioural and psychiatric outcomes. Maternal mental state has been associated with offspring NR3C1 promotor and IGF2/H19 imprinting control region (ICR) methylation patterns. However, there is a lack of prospective studies with long-term follow-up. METHODS: 52 mother-offspring pairs were studied from 12 to 22 weeks of pregnancy and offspring was followed-up until 28-29 years-of-age. During pregnancy, mothers filled in a Life Event Scale and a Daily Hassles Scale measuring perceived stress; i.e., appraisal or subjectively experienced severity of impact of important life events and of daily hassles in several life domains during pregnancy, respectively. Green space was quantified around the residence, using high-resolution (1 m2) map data. Saliva and blood samples were obtained from the adult offspring. Absolute DNA methylation levels were determined in blood and saliva on four NR3C1 amplicons, and one IGF2/H19 ICR amplicon using a bisulfite PCR and sequencing method. Linear mixed effect models were used to test the associations between perceived stress and green spaces during pregnancy, and adult offspring methylation patterns. RESULTS: We found associations between maternal perceived stress during pregnancy and methylation patterns on two out of the four NR3C1 amplicons, measured in blood, from offspring in adulthood, but not with IGF2/H19 methylation. For an interquartile-range (IQR) increase in maternal perceived life event or daily hassles stress scores, absolute methylation levels on several NR3C1 CpG sites were significantly changed (-1.62 % to +5.89 %, p<0.05). Maternal perceived stress scores were not associated with IGF2/H19 methylation, neither in blood nor in saliva. Maternal exposure to green spaces surrounding the residence during the pregnancy was associated with IGF2/H19 ICR methylation (-0.80 % to -1.04 %, p<0.05) in saliva, but not with NR3C1 promotor methylation. CONCLUSION: We observed significant long-term effects of maternal perceived stress during pregnancy on the methylation patterns of the NR3C1 promotor in offspring well into adulthood. This may imply that maternal psychological distress during pregnancy may influence the regulation of the HPA-axis well into adulthood. Additionally, maternal proximity to green spaces was associated with IGF2/H19 ICR methylation patterns, which is a novel finding.


Subject(s)
DNA Methylation , Insulin-Like Growth Factor II , Prenatal Exposure Delayed Effects , RNA, Long Noncoding , Receptors, Glucocorticoid , Stress, Psychological , Humans , Female , Pregnancy , DNA Methylation/genetics , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/metabolism , Pilot Projects , Stress, Psychological/genetics , Stress, Psychological/metabolism , Adult , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Prenatal Exposure Delayed Effects/genetics , Prenatal Exposure Delayed Effects/metabolism , Male , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Adult Children/psychology , Mothers/psychology , Promoter Regions, Genetic/genetics , Genomic Imprinting/genetics , Prospective Studies
14.
Cells ; 13(10)2024 May 14.
Article in English | MEDLINE | ID: mdl-38786059

ABSTRACT

In recent decades, there has been a dramatic rise in the rates of children being born after in utero exposure to drugs of abuse, particularly opioids. Opioids have been shown to have detrimental effects on neurons and glia in the central nervous system (CNS), but the impact of prenatal opioid exposure (POE) on still-developing synaptic circuitry is largely unknown. Astrocytes exert a powerful influence on synaptic development, secreting factors to either promote or inhibit synapse formation and neuronal maturation in the developing CNS. Here, we investigated the effects of the partial µ-opioid receptor agonist buprenorphine on astrocyte synaptogenic signaling and morphological development in cortical cell culture. Acute buprenorphine treatment had no effect on the excitatory synapse number in astrocyte-free neuron cultures. In conditions where neurons shared culture media with astrocytes, buprenorphine attenuated the synaptogenic capabilities of astrocyte-secreted factors. Neurons cultured from drug-naïve mice showed no change in synapses when treated with factors secreted by astrocytes from POE mice. However, this same treatment was synaptogenic when applied to neurons from POE mice, indicating a complex neuroadaptive response in the event of impaired astrocyte signaling. In addition to promoting morphological and connectivity changes in neurons, POE exerted a strong influence on astrocyte development, disrupting their structural maturation and promoting the accumulation of lipid droplets (LDs), suggestive of a maladaptive stress response in the developing CNS.


Subject(s)
Analgesics, Opioid , Astrocytes , Neurons , Prenatal Exposure Delayed Effects , Signal Transduction , Synapses , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Animals , Synapses/metabolism , Synapses/drug effects , Female , Pregnancy , Mice , Analgesics, Opioid/pharmacology , Analgesics, Opioid/adverse effects , Prenatal Exposure Delayed Effects/pathology , Prenatal Exposure Delayed Effects/metabolism , Neurons/metabolism , Neurons/drug effects , Neurons/pathology , Signal Transduction/drug effects , Buprenorphine/pharmacology , Cells, Cultured , Mice, Inbred C57BL
15.
Cell Biol Toxicol ; 40(1): 37, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38777957

ABSTRACT

Bisphenol A (BPA) is a common component in the manufacture of daily plastic consumer goods. Recent studies have suggested that prenatal exposure to BPA can increase the susceptibility of offspring to mental illness, although the underlying mechanisms remain unclear. In this study, we performed transcriptomic and epigenomic profiling in the adult mouse brain following prenatal exposure to low-dose BPA. We observed a sex-specific transcriptional dysregulation in the cortex, with more significant differentially expressed genes was observed in adult cortex from male offspring. Moreover, the upregulated genes primarily influenced neuronal functions, while the downregulated genes were significantly associated with energy metabolism pathways. More evidence supporting impaired mitochondrial function included a decreased ATP level and a reduced number of mitochondria in the cortical neuron of the BPA group. We further investigated the higher-order chromatin regulatory patterns of DEGs by incorporating published Hi-C data. Interestingly, we found that upregulated genes exhibited more distal interactions with multiple enhancers, while downregulated genes displayed relatively short-range interactions among adjacent genes. Our data further revealed decreased H3K9me3 signal on the distal enhancers of upregulated genes, whereas increased DNA methylation and H3K27me3 signals on the promoters of downregulated genes. In summary, our study provides compelling evidence for the potential health risks associated with prenatal exposure to BPA, and uncovers sex-specific transcriptional changes with a complex interplay of multiple epigenetic mechanisms.


Subject(s)
Benzhydryl Compounds , Brain , DNA Methylation , Epigenesis, Genetic , Phenols , Prenatal Exposure Delayed Effects , Animals , Benzhydryl Compounds/toxicity , Phenols/toxicity , Female , Pregnancy , Prenatal Exposure Delayed Effects/genetics , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/metabolism , Epigenesis, Genetic/drug effects , Male , Mice , Brain/metabolism , Brain/drug effects , DNA Methylation/drug effects , Transcriptome/drug effects , Transcriptome/genetics , Mice, Inbred C57BL
16.
Brain Res ; 1838: 148963, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38705555

ABSTRACT

BACKGROUND AND AIM: Autism spectrum disorder (ASD) is a neurodevelopmental disorder with two core behavioral symptoms restricted/repetitive behavior and social-communication deficit. The unknown etiology of ASD makes it difficult to identify potential treatments. Valproic acid (VPA) is an anticonvulsant drug with teratogenic effects during pregnancy in humans and rodents. Prenatal exposure to VPA induces autism-like behavior in both humans and rodents. This study aimed to investigate the protective effects of Diosgenin in prenatal Valproic acid-induced autism in rats. METHOD: pregnant Wister female rats were given a single intraperitoneal injection of VPA (600 mg/kg, i.p.) on gestational day 12.5. The male offspring were given oral Dios (40 mg/kg, p.o.) or Carboxymethyl cellulose (5 mg/kg, p.o.) for 30 days starting from postnatal day 23. On postnatal day 52, behavioral tests were done. Additionally, biochemical assessments for oxidative stress markers were carried out on postnatal day 60. Further, histological evaluations were performed on the prefrontal tissue by Nissl staining and Immunohistofluorescence. RESULTS: The VPA-exposed rats showed increased anxiety-like behavior in the elevated plus maze (EPM). They also demonstrated repetitive and grooming behaviors in the marble burying test (MBT) and self-grooming test. Social interaction was reduced, and they had difficulty detecting the novel object in the novel object recognition (NOR) test. Also, VPA-treated rats have shown higher levels of oxidative stress malondialdehyde (MDA) and lower GPX, TAC, and superoxide dismutase (SOD) levels. Furthermore, the number of neurons decreased and the ERK signaling pathway upregulated in the prefrontal cortex (PFC). On the other hand, treatment with Dios restored the behavioral consequences, lowered oxidative stress, and death of neurons, and rescued the overly activated ERK1/2 signaling in the prefrontal cortex. CONCLUSION: Chronic treatment with Dios restored the behavioral, biochemical, and histological abnormalities caused by prenatal VPA exposure.


Subject(s)
Autistic Disorder , Diosgenin , Disease Models, Animal , Neuroprotective Agents , Oxidative Stress , Prenatal Exposure Delayed Effects , Rats, Wistar , Valproic Acid , Animals , Valproic Acid/pharmacology , Female , Pregnancy , Neuroprotective Agents/pharmacology , Rats , Oxidative Stress/drug effects , Male , Diosgenin/pharmacology , Autistic Disorder/chemically induced , Autistic Disorder/metabolism , Autistic Disorder/drug therapy , Prenatal Exposure Delayed Effects/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Behavior, Animal/drug effects , Anticonvulsants/pharmacology , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/chemically induced , Autism Spectrum Disorder/drug therapy , Anxiety/drug therapy , Anxiety/chemically induced , Social Behavior
17.
Arch Med Res ; 55(4): 103002, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38735235

ABSTRACT

BACKGROUND: Maternal obesity (MO) has been shown to adversely affect metabolic, oxidative, reproductive, and cognitive function in offspring. However, it is unclear whether lifestyle modification can ameliorate the metabolic and organ dysfunction programmed by MO and prevent the effects of metabolic syndrome in adulthood. This study aimed to evaluate whether moderate voluntary exercise in the offspring of rats born to obese mothers can ameliorate the adverse effects of MO programming on metabolism and liver function in mid-adulthood. METHODS: Offspring of control (CF1) and MOF1 mothers were fed with a control diet from weaning. Adult males and females participated in 15 min exercise sessions five days/week. Metabolic parameters were analyzed before and after the exercise intervention. Liver oxidative stress biomarkers and antioxidant enzymes were analyzed before and after the intervention. RESULTS: Males showed that CF1ex ran more than MOF1ex and increased the distance covered. In contrast, females in both groups ran similar distances and remained constant but ran more distance than males. At PND 300 and 450, male and female MOF1 had higher leptin, triglycerides, insulin, and HOMA-IR levels than CF1. However, male MOF1ex had lower triglycerides, insulin, and HOMA-IR levels than MOF1. Improvements in liver fat and antioxidant enzymes were observed in CF1ex and MOF1ex males and females compared to their respective CF1 and MOF1 groups. CONCLUSION: These findings suggest that moderate voluntary exercise, even when started in mid-adulthood, can improve metabolic outcomes and delay accelerated metabolic aging in MO-programmed rats in a sex-dependent manner.


Subject(s)
Aging , Obesity, Maternal , Physical Conditioning, Animal , Animals , Female , Male , Rats , Pregnancy , Aging/metabolism , Obesity, Maternal/metabolism , Oxidative Stress , Rats, Wistar , Liver/metabolism , Prenatal Exposure Delayed Effects/metabolism , Obesity/metabolism , Obesity/therapy , Obesity/physiopathology
18.
Neuroscience ; 551: 17-30, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38777136

ABSTRACT

Exposure to lipopolysaccharide (LPS) during prenatal development leads to various changes in neurobiological and behavioural patterns. Similarly, continuous exposure to constant light (LL) during the critical developmental period of the circadian system affects gene expression in various tissues in adulthood. Given the reciprocal nature of the interaction between the circadian and the immune systems, our study primarily investigated the individual effects of both interventions and, more importantly, their combined effect. We aimed to explore whether there might be a potential synergistic effect on circadian rhythms and their parameters, focussing on the expression of clock genes, immune-related genes, and specific genes in the hippocampus, pineal gland, spleen and adrenal gland of rats at postnatal day 30. Our results show a significant influence of prenatal LPS and postnatal LL on the expression profiles of all genes assessed. However, the combination of prenatal LPS and postnatal LL only revealed an enhanced negative effect in a minority of the comparisons. In most cases, it appeared to attenuate the changes induced by the individual interventions, restoring the measured parameters to values closer to those of the control group. In particular, genes such as Nr1d1, Aanat and Tph1 showed increased amplitude in the pineal gland and spleen, while the kynurenine enzymes Kynu and KatII developed circadian rhythmicity in the adrenal glands only after the combined interventions. Our data suggest that a mild immunological challenge during prenatal development may play a critical role in triggering an adaptive response of the circadian clock later in life.


Subject(s)
Circadian Rhythm , Light , Lipopolysaccharides , Prenatal Exposure Delayed Effects , Spleen , Animals , Female , Prenatal Exposure Delayed Effects/metabolism , Pregnancy , Circadian Rhythm/physiology , Male , Spleen/metabolism , Pineal Gland/metabolism , Rats, Wistar , Hippocampus/metabolism , Rats , Adrenal Glands/metabolism , Transcriptome
19.
Endocrinology ; 165(7)2024 May 27.
Article in English | MEDLINE | ID: mdl-38788194

ABSTRACT

Androgen excess is a hallmark feature of polycystic ovary syndrome (PCOS), the most common form of anovulatory infertility. Clinical and preclinical evidence links developmental or chronic exposure to hyperandrogenism with programming and evoking the reproductive and metabolic traits of PCOS. While critical androgen targets remain to be determined, central GABAergic neurons are postulated to be involved. Here, we tested the hypothesis that androgen signaling in GABAergic neurons is critical in PCOS pathogenesis in 2 well-characterized hyperandrogenic mouse models of PCOS. Using cre-lox transgenics, GABA-specific androgen receptor knockout (GABARKO) mice were generated and exposed to either acute prenatal androgen excess (PNA) or chronic peripubertal androgen excess (PPA). Females were phenotyped for reproductive and metabolic features associated with each model and brains of PNA mice were assessed for elevated GABAergic input to gonadotropin-releasing hormone (GnRH) neurons. Reproductive and metabolic dysfunction induced by PPA, including acyclicity, absence of corpora lutea, obesity, adipocyte hypertrophy, and impaired glucose homeostasis, was not different between GABARKO and wild-type (WT) mice. In PNA mice, acyclicity remained in GABARKO mice while ovarian morphology and luteinizing hormone secretion was not significantly impacted by PNA or genotype. However, PNA predictably increased the density of putative GABAergic synapses to GnRH neurons in adult WT mice, and this PNA-induced plasticity was absent in GABARKO mice. Together, these findings suggest that while direct androgen signaling in GABA neurons is largely not required for the development of PCOS-like traits in androgenized models of PCOS, developmental programming of GnRH neuron innervation is dependent upon androgen signaling in GABA neurons.


Subject(s)
Disease Models, Animal , GABAergic Neurons , Hyperandrogenism , Mice, Knockout , Polycystic Ovary Syndrome , Receptors, Androgen , Animals , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/genetics , Female , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Mice , GABAergic Neurons/metabolism , Hyperandrogenism/metabolism , Hyperandrogenism/genetics , Ovary/metabolism , Androgens/metabolism , Pregnancy , Gonadotropin-Releasing Hormone/metabolism , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/genetics
20.
Biochim Biophys Acta Mol Basis Dis ; 1870(6): 167258, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38788910

ABSTRACT

The increasing prevalence of obesity, type 2 diabetes mellitus (T2DM), and gestational diabetes (GDM) among pregnant women has risen dramatically worldwide. The antihyperglycemic drug metformin is the most common drug for T2DM treatment in non-pregnant individuals; nevertheless, it is increasingly being used for diabetes-complicated pregnancies. Studies on the long-term metabolic effects of this drug in offspring remain scarce. This work aimed to determine the effect of metformin exposure during pregnancy and lactation on the offspring of a model of diet-induced maternal hyperglycemia. Cohorts of pregnant mice were fed a 46% fat diet (HFD) or a control standard diet (SD). A group of dams were exposed to metformin during pregnancy and lactation. After weaning, the offspring were fed SD for 8 weeks and then challenged with a 46% HFD after puberty for 12 weeks. Irrespective of the maternal diet, offspring of metformin-exposed mothers had a lower body weight and reduced inguinal white adipose tissue (iWAT) mass after HFD challenge. This was associated with increased expression of Pparg, Fabp4, Glut4, Srebp1, and Fasn in the iWAT during adulthood in the metabolically impaired dams exposed to metformin, suggesting increased adipogenesis and de novo lipogenesis. Increased expression of Fasn associated with decreased methylation levels at its promoter and proximal coding region in the iWAT was found. These results suggest that metformin modulates gene expression levels by epigenetic mechanisms in maternal metabolic-impaired conditions.


Subject(s)
Body Weight , Diet, High-Fat , Lactation , Metformin , Prenatal Exposure Delayed Effects , Sterol Regulatory Element Binding Protein 1 , Animals , Metformin/pharmacology , Female , Pregnancy , Lactation/drug effects , Mice , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/pathology , Prenatal Exposure Delayed Effects/chemically induced , Diet, High-Fat/adverse effects , Body Weight/drug effects , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , PPAR gamma/metabolism , PPAR gamma/genetics , Glucose Transporter Type 4/metabolism , Glucose Transporter Type 4/genetics , Hypoglycemic Agents/pharmacology , Adipose Tissue, White/metabolism , Adipose Tissue, White/drug effects , Obesity/metabolism , Obesity/pathology , Obesity/chemically induced , Fatty Acid Synthase, Type I/metabolism , Fatty Acid Synthase, Type I/genetics , Male , Mice, Inbred C57BL , Adipose Tissue/metabolism , Adipose Tissue/drug effects , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL
...