Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Neuroendocrinology ; 113(8): 844-858, 2023.
Article in English | MEDLINE | ID: mdl-36948162

ABSTRACT

INTRODUCTION: Early life ethanol exposure is known to program hypothalamic proopiomelanocortin (POMC) neurons to express a reduced level of POMC and its control of stress axis functions throughout the life span. In this study, we tested whether miRNAs contribute to the ethanol-induced suppression of Pomc gene expression during the developmental period. METHODS: In in vivo studies, POMC-EGFP male mice were fed with 2.5 g/kg ethanol using milk formula (AF), pair-fed isocaloric milk formula, or left in the litter during postnatal days (PNDs) 2-6. In in vitro studies, mHypoA-POMC/GFP cells were treated with ethanol (50 mM) for a 24-h period. Hypothalamic tissues or cell extracts were used for measurement of miRNAs and POMC mRNA. RESULTS: Determination of genome-wide microRNA expression profile identified 40 miRNAs significantly altered in hypothalamic tissues of AF mice. In silico analysis further identified miRNA-383, -384, and -488 have putative binding sites at the POMC 3'UTR. However, only miR-383 and miR-384 are identified to be responsive to ethanol. Administration of miR-383 or -384 inhibitor oligos suppressed ethanol-stimulated miR-383 or -384 expression and restored Pomc mRNA and protein expression in AF mice. mHypoA-POMC/GFP cells when treated with ethanol showed elevated levels of miR-383 and miR-384 and reduced level of Pomc mRNA. Treatment with miR-383 or -384 mimic oligos reduced the level of Pomc mRNA, while treatment with miR-383 or -384 inhibitor oligos increased the level of Pomc mRNA. Reporter assay further confirms the binding specificity of miR-383 and miR-384 to Pomc 3'UTR. CONCLUSION: These data suggest that miR-383 and miR-384 suppress Pomc gene expression and may contribute to the ethanol-induced alteration of the stress axis functions.


Subject(s)
Ethanol , Pro-Opiomelanocortin , Mice , Male , Animals , Pro-Opiomelanocortin/metabolism , Ethanol/metabolism , Ethanol/pharmacology , 3' Untranslated Regions , Hypothalamus/metabolism , Gene Expression
2.
Alcohol Clin Exp Res (Hoboken) ; 47(1): 18-35, 2023 01.
Article in English | MEDLINE | ID: mdl-36341762

ABSTRACT

We conducted a systematic review with meta-analytic elements using publicly available Gene Expression Omnibus (GEO) datasets to determine the role of epigenetic mechanisms in prenatal alcohol exposure (PAE)-induced hypothalamic-pituitary-adrenal (HPA) axis dysfunctions in offspring. Several studies have demonstrated that PAE has long-term consequences on HPA axis functions in offspring. Some studies determined that alcohol-induced epigenetic alterations during fetal development persist in adulthood. However, additional research is needed to understand the major epigenetic events leading to alcohol-induced teratogenesis of the HPA axis. Our network analysis of GEO datasets identified key pathways relevant to alcohol-mediated histone modifications, DNA methylation, and miRNA involvement associated with PAE-induced alterations of the HPA axis. Our analysis indicated that PAE perturbated the epigenetic machinery to activate corticotrophin-releasing hormone, while it suppressed opioid, glucocorticoid receptor, and circadian clock genes. These results help to further our understanding of the epigenetic basis of alcohol's effects on HPA axis development.


Subject(s)
Hypothalamo-Hypophyseal System , Prenatal Exposure Delayed Effects , Pregnancy , Female , Humans , Hypothalamo-Hypophyseal System/metabolism , Prenatal Exposure Delayed Effects/chemically induced , Pituitary-Adrenal System/metabolism , Ethanol/adverse effects , Epigenesis, Genetic , Stress, Psychological/metabolism
3.
PLoS One ; 17(2): e0263340, 2022.
Article in English | MEDLINE | ID: mdl-35143549

ABSTRACT

Previously it has been shown that fetal alcohol exposure increases the stress response partly due to lowering stress regulatory proopiomelanocortin (Pomc) gene expression in the hypothalamus via epigenetic mechanisms for multiple generations in mixed-breed rats. In this study we assess the induction of heritable epigenetic changes of Pomc-related variants by fetal alcohol exposure in isogenic Fischer 344 rats. Using transgenerational breeding models and fetal alcohol exposure procedures, we determined changes in hypothalamic Pomc gene expression and its methylation levels, plasma corticosterone hormone response to restraint stress, and anxiety-like behaviors using elevated plus maze tests in fetal alcohol-exposed offspring for multiple generations in isogenic Fischer rats. Fetal alcohol-exposed male and female rat offspring showed significant deficits in POMC neuronal functions with increased Pomc gene methylation and reduced expression. These changes in POMC neuronal functions were associated with increased plasma corticosterone response to restraint stress and increased anxiety-like behavior. These effects of fetal alcohol exposure persisted in the F1, F2, and F3 progeny of the male germline but not of the female germline. These data suggest that fetal alcohol exposure induces heritable changes in Pomc-related variants involving stress hyperresponsiveness and anxiety-like behaviors which perpetuate into subsequent generations through the male germline via epigenetic modifications.


Subject(s)
Pro-Opiomelanocortin
4.
Front Neurosci ; 15: 608102, 2021.
Article in English | MEDLINE | ID: mdl-33796006

ABSTRACT

Fetal alcohol exposure (FAE) causes various neurodevelopmental deficits in offspring, including reduced expression of the stress regulatory proopiomelanocortin (Pomc) gene and an elevated stress response for multiple generations via the male germline. Male germline-specific effects of FAE on the Pomc gene raises the question if the sex-determining region Y (SRY) may have a role in regulating Pomc gene expression. Using a transgenerational model of FAE in Fischer 344 rats, we determined the role of SRY in the regulation of the Pomc gene. FAEs, like on the Pomc gene, reduced Sry gene expression in sperm and the mediobasal hypothalamus (MBH) in male adult offspring. Fetal alcohol-induced inhibition of Sry gene expression was associated with increased Sry promoter DNA methylation. Additionally, fetal alcohol effects on the Sry gene persisted for three generations in the male germline but not in the female germline. Sry gene knockdown reduced the Pomc gene expression. Sry recruitment onto the Pomc promoter was found to be reduced in the hypothalamus of fetal alcohol-exposed rats compared to control rats. Pomc promoter luciferase activity was increased following Sry overexpression. A site-directed mutagenesis study revealed that SRY binding sites are required for POMC promoter transcription activity. Overall, these findings suggest that SRY plays a stimulatory role in the regulation of Pomc gene expression and may potentially contribute to the fetal alcohol-induced changes in the level of Pomc gene expression for multiple generations.

5.
Clin Epigenetics ; 12(1): 70, 2020 05 24.
Article in English | MEDLINE | ID: mdl-32448218

ABSTRACT

BACKGROUND: Alcohol exposures in utero have been shown to alter immune system functions in the offspring which persists into adulthood. However, it is not apparent why the in utero alcohol effect on the immune system persists into adulthood of fetal alcohol-exposed offspring. The objective of this study was to determine the long-term effects of fetal alcohol exposure on the production of interferon-ϒ (IFN-ϒ), a cytokine known to regulate both innate and adaptive immunity. METHODS: Isogenic Fisher 344 rats were bred to produce pregnant dams, which were fed with a liquid diet containing 6.7% alcohol between gestation days 7 and 21 and pair-fed with an isocaloric liquid diet or fed ad libitum with rat chow; their male and female offspring were used for the study. F1-F3 generation rats were used when they were 2 to 3 months old. Fetal alcohol exposure effects on the Ifn-ɣ gene was determined by measuring the gene promoter methylation and mRNA and protein expression in the spleen. Additionally, transgenerational studies were conducted to evaluate the germline-transmitted effects of fetal alcohol exposure on the Ifn-ɣ gene. RESULTS: Fetal alcohol exposure reduced the expression of Ifn-ɣ mRNA and IFN-ϒ protein while it increased the proximal promoter methylation of the Ifn-ɣ gene in both male and female offspring during the adult period. Transgenerational studies revealed that the reduced levels of Ifn-ɣ expression and increased levels of its promoter methylation persisted only in F2 and F3 generation males derived from the male germ line. CONCLUSION: Overall, these findings provide the evidence that fetal alcohol exposures produce an epigenetic mark on the Ifn-ɣ gene that passes through multiple generations via the male germ line. These data provide the first evidence that the male germ line transmits fetal alcohol exposure's adverse effects on the immune system.


Subject(s)
Fetal Alcohol Spectrum Disorders/genetics , Interferon-gamma/genetics , Animals , DNA Methylation , Epigenesis, Genetic , Female , Fetal Alcohol Spectrum Disorders/immunology , Inheritance Patterns , Interferon-gamma/metabolism , Male , Promoter Regions, Genetic , Rats, Inbred F344 , Spleen/immunology
6.
Alcohol Clin Exp Res ; 43(9): 1887-1897, 2019 09.
Article in English | MEDLINE | ID: mdl-31329297

ABSTRACT

BACKGROUND: We have recently shown that binge or heavy levels of alcohol drinking increase deoxyribonucleic acid (DNA) methylation and reduce gene expression of proopiomelanocortin (POMC) and period 2 (PER2) in adult human subjects (Gangisetty et al., Alcohol Clin Exp Res, 43, 2019, 212). One hypothesis would be that methylation of these 2 genes is consistently associated with alcohol exposure and could be used as biomarkers to predict risk of prenatal alcohol exposure (PAE). Results of the present study provided some support for this hypothesis. METHODS: We conducted a series of studies to determine DNA methylation changes in stress regulatory genes proopiomelanocortin (POMC) and period 2 (PER2) using biological samples from 3 separate cohorts of patients: (i) pregnant women who consumed moderate-to-high levels of alcohol or low/unexposed controls, (ii) children with PAE and non-alcohol-exposed controls, and (iii) children with PAE treated with or without choline. RESULTS: We found pregnant women who consumed moderate-to-high levels of alcohol and gave birth to PAE children had higher DNA methylation of POMC and PER2. PAE children also had increased methylation of POMC and PER2. The differences in the gene methylation of PER2 and POMC between PAE and controls did not differ by maternal smoking status. PAE children had increased levels of stress hormone cortisol and adrenocorticotropic hormone. Choline supplementation reduced DNA hypermethylation and increased expression of POMC and PER2 in children with PAE. CONCLUSIONS: These data suggest that PAE significantly elevates DNA methylation of POMC and PER2 and increases levels of stress hormones. Furthermore, these results suggest the possibility that measuring DNA methylation levels of PER2 and POMC in biological samples from pregnant women or from children may be useful for identification of a woman or a child with PAE.


Subject(s)
Central Nervous System Depressants/adverse effects , Ethanol/adverse effects , Period Circadian Proteins/metabolism , Prenatal Exposure Delayed Effects , Pro-Opiomelanocortin/metabolism , Adolescent , Adult , Case-Control Studies , Child , Child, Preschool , Choline/pharmacology , Choline/therapeutic use , DNA Methylation/drug effects , Dietary Supplements , Epigenesis, Genetic/drug effects , Female , Fetal Alcohol Spectrum Disorders/metabolism , Fetal Alcohol Spectrum Disorders/prevention & control , Gene Expression Regulation/drug effects , Humans , Lipotropic Agents/pharmacology , Lipotropic Agents/therapeutic use , Male , Pregnancy
7.
Neuropsychopharmacology ; 44(9): 1579-1588, 2019 08.
Article in English | MEDLINE | ID: mdl-30737481

ABSTRACT

Growing evidence has shown that developmental alcohol exposure induces central nervous system inflammation and microglia activation, which may contribute to long-term health conditions, such as fetal alcohol spectrum disorders. These studies sought to investigate whether neonatal alcohol exposure during postnatal days (PND) 2-6 in rats (third trimester human equivalent) leads to long-term disruption of the neuroimmune response by microglia. Exposure to neonatal alcohol resulted in acute increases in activation and inflammatory gene expression in hypothalamic microglia including tumor necrosis factor alpha (TNF-α) and interleukin 6 (IL-6). Adults with neonatal alcohol pre-exposure (alcohol fed; AF) animals showed an exaggerated peripheral stress hormonal response to an immune challenge (lipopolysaccharides; LPS). In addition, there were significantly more microglia present in the hypothalamus of adult AF animals, and their hypothalamic microglia showed more cluster of differentiation molecule 11b (Cd11b) activation, TNF-α expression, and IL-6 expression in response to LPS. Interestingly, blocking microglia activation with minocycline treatment during PND 2-6 alcohol exposure ameliorated the hormonal and microglial hypersensitivity to LPS in AF adult animals. Investigation of possible epigenetic programming mechanisms by alcohol revealed neonatal alcohol decreased several repressive regulators of transcription in hypothalamic microglia, while concomitantly increasing histone H3 acetyl lysine 9 (H3K9ac) enrichment at TNF-α and IL-6 promoter regions. Importantly, adult hypothalamic microglia from AF animals showed enduring increases in H3K9ac enrichment of TNF-α and IL-6 promoters both at baseline and after LPS exposure, suggesting a possible epigenetic mechanism for the long-term immune disruption due to hypothalamic microglial priming.


Subject(s)
Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Gene Expression/drug effects , Hypothalamus/drug effects , Microglia/drug effects , Tumor Necrosis Factor-alpha/drug effects , Animals , Animals, Newborn , Epigenesis, Genetic , Gene Expression/immunology , Histone Code/drug effects , Hypothalamus/cytology , Hypothalamus/immunology , Inflammation/immunology , Interleukin-6/immunology , Lipopolysaccharides/pharmacology , Microglia/immunology , Rats , Tumor Necrosis Factor-alpha/immunology
8.
Alcohol Clin Exp Res ; 43(2): 212-220, 2019 02.
Article in English | MEDLINE | ID: mdl-30597578

ABSTRACT

BACKGROUND: Epigenetic modifications of a gene have been shown to play a role in maintaining a long-lasting change in gene expression. We hypothesize that alcohol's modulating effect on DNA methylation on certain genes in blood is evident in binge and heavy alcohol drinkers and is associated with alcohol motivation. METHODS: Methylation-specific polymerase chain reaction (PCR) assays were used to measure changes in gene methylation of period 2 (PER2) and proopiomelanocortin (POMC) genes in peripheral blood samples collected from nonsmoking moderate, nonbinging, binge, and heavy social drinkers who participated in a 3-day behavioral alcohol motivation experiment of imagery exposure to either stress, neutral, or alcohol-related cues, 1 per day, presented on consecutive days in counterbalanced order. Following imagery exposure on each day, subjects were exposed to discrete alcoholic beer cues followed by an alcohol taste test (ATT) to assess behavioral motivation. Quantitative real-time PCR was used to measure gene expression of PER2 and POMC gene levels in blood samples across samples. RESULTS: In the sample of moderate, binge, and heavy drinkers, we found increased methylation of the PER2 and POMC DNA, reduced expression of these genes in the blood samples of the binge and heavy drinkers relative to the moderate, nonbinge drinkers. Increased PER2 and POMC DNA methylation was also significantly predictive of both increased levels of subjective alcohol craving immediately following imagery (p < 0.0001), and with presentation of the alcohol (2 beers) (p < 0.0001) prior to the ATT, as well as with alcohol amount consumed during the ATT (p < 0.003). CONCLUSIONS: These data establish significant association between binge or heavy levels of alcohol drinking and elevated levels of methylation and reduced levels of expression of POMC and PER2 genes. Furthermore, elevated methylation of POMC and PER2 genes is associated with greater subjective and behavioral motivation for alcohol.


Subject(s)
Alcohol Drinking/metabolism , Alcohol Drinking/psychology , Binge Drinking/metabolism , DNA Methylation/drug effects , Motivation , Period Circadian Proteins/metabolism , Pro-Opiomelanocortin/metabolism , Adult , Craving/drug effects , Cues , Epigenesis, Genetic , Ethanol/pharmacology , Female , Gene Expression/drug effects , Humans , Male , Period Circadian Proteins/blood , Photic Stimulation , Pro-Opiomelanocortin/blood , Young Adult
9.
Front Biosci (Landmark Ed) ; 23(8): 1445-1464, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29293444

ABSTRACT

Epigenetics involves multiple processes such as DNA methylation, histone code modifications, and noncoding RNAs to regulate gene expression. In recent years the implications of epigenetic mechanisms have emerged in the field of neuroscience especially in brain development, memory, learning, and various cognition processes. Epigenetics also plays a pivotal role during the aging process of the brain which has led to various age-related neurodegenerative diseases. This manuscript portrays the findings of various epigenetic mechanisms that play a critical role and their implications in aging as well as  age-related neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, and Huntington's disease.


Subject(s)
Aging/genetics , Brain/metabolism , Epigenesis, Genetic , Neurodegenerative Diseases/genetics , Animals , Brain/pathology , Brain/physiopathology , DNA Methylation , Histone Code , Humans , RNA, Untranslated/genetics
10.
J Endocrinol ; 235(1): 1-14, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28710248

ABSTRACT

Fetal alcohol exposure (FAE) is known to increase prolactin (PRL) secretion from the pituitary lactotropes. In this study, we determined whether microRNAs (miRs) are involved in FAE-induced alteration in PRL release. We employed a rat animal model of FAE involving feeding pregnant Fisher 344 rats with a liquid diet containing 6.7% alcohol between gestational days 7-21 (AF). Both cyclic and estradiol-implanted FAE females showed increased levels of plasma PRL and pituitary Prl mRNA but reduced levels of pituitary dopamine D2 receptor (D2r) and its short spliced form (D2s). FAE increased the expression levels of miR-9 and miR-326 and did not produce any significant changes in miR-153 or miR-200a levels in the pituitary. Effects of FAE on miR-9 and miR-326 were associated with reduced levels of D2r and D2s, increased levels of Prl in the pituitary, and in plasma. These effects of FAE on D2r, D2s and Prl were enhanced following estradiol treatment. In PRL-producing MMQ cells, ethanol increased miR-9 but not miR-326, reduced levels of D2r and D2s and increased levels of Prl Treatment of MMQ cells with an anti-miR-9 oligo reduced ethanol effects on miR-9, D2r, D2s and Prl miR-9 mimic oligos reduced the luciferase activity of reporter vector containing D2r 3'UTR, but failed to reduce the mutant luciferase activity. These data suggest that FAE programs the pituitary to produce increased amounts of miR-9 expression that represses the D2r gene and its spliced variant D2s by targeting its 3'UTR leading to an increase in PRL production and secretion.


Subject(s)
Alcohols/adverse effects , Fetus/drug effects , Maternal Exposure/adverse effects , MicroRNAs/metabolism , Prenatal Exposure Delayed Effects/metabolism , Prolactin/blood , Receptors, Dopamine D2/metabolism , Animals , Female , Fetus/metabolism , Humans , Male , MicroRNAs/genetics , Pituitary Gland/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/etiology , Prenatal Exposure Delayed Effects/genetics , Prolactin/metabolism , Rats , Rats, Inbred F344 , Receptors, Dopamine D2/genetics
11.
Brain Res ; 1659: 142-147, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28137424

ABSTRACT

Progesterone (P) binding to the intracellular progesterone receptors (PRs) plays a key role in epilepsy via modulation of GABA-A receptor plasticity in the brain. This is thought to occur via conversion of P to neurosteroids such as allopregnanolone, an allosteric modulator of GABA-A receptors. In the female brain, the composition of GABA-A receptors is not static and undergoes dynamic spatial changes in response to fluctuations in P and neurosteroid levels. Synaptic α2-containing GABA-A receptors contribute to phasic neuronal excitability and seizure susceptibility. However, the mechanisms underlying α2-subunit plasticity remain unclear. Here, we utilized the neurosteroid synthesis inhibitor finasteride and PR knockout mice to investigate the role of PRs in α2-subunit in the hippocampus. α2-Subunit expression was significantly upregulated during the high-P state of diestrous stage and with P treatment in wildtype and PR knockout mice. In contrast, there was no change in α2-subunit expression when metabolism of P into neurosteroids was blocked by finasteride in both genotypes. These findings suggest that ovarian cycle-related P and neurosteroids regulate α2-GABA-A receptor expression in the hippocampus via a non-PR pathway, which may be relevant to menstrual-cycle related brain conditions.


Subject(s)
Diestrus/metabolism , Hippocampus/metabolism , Neurotransmitter Agents/metabolism , Progesterone/metabolism , Receptors, GABA-A/metabolism , Receptors, Progesterone/metabolism , Animals , Diestrus/drug effects , Female , Hippocampus/drug effects , Mice, Inbred C57BL , Mice, Knockout , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Neurotransmitter Agents/administration & dosage , Neurotransmitter Agents/antagonists & inhibitors , Progesterone/administration & dosage , RNA, Messenger/metabolism , Receptors, GABA-A/genetics , Receptors, Progesterone/genetics , Up-Regulation/drug effects , Up-Regulation/physiology
12.
Adv Neurobiol ; 12: 1-25, 2016.
Article in English | MEDLINE | ID: mdl-27651245

ABSTRACT

Epigenetic modifications, including DNA methylation, covalent histone modifications, and small noncoding RNAs, play a key role in regulating the gene expression. This regulatory mechanism is important in cellular differentiation and development. Recent advances in the field of epigenetics extended the role of epigenetic mechanisms in controlling key biological processes such as genome imprinting and X-chromosome inactivation. Aberrant epigenetic modifications are associated with the development of many diseases. The role of epigenetic modifications in various neurodegenerative disorders including Alzheimer's disease, Parkinson's disease, Huntington disease, epilepsy, and multiple sclerosis is rapidly emerging. The use of epigenetic modifying drugs to treat these diseases has been the interest in recent years. A number of natural products having diverse mechanism of action are used for drug discovery. For many years, natural compounds have been used to treat various neurodegenerative diseases, but the use of such compounds as epigenetic modulators to reverse or treat neurological diseases are not well studied. In this chapter, we mainly focus on how various epigenetic modifications play a key role in neurodegenerative diseases, their mechanism of action, and how it acts as a potential therapeutic target for epigenetic drugs to treat these diseases will be discussed.


Subject(s)
Biological Products/therapeutic use , Epigenesis, Genetic , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/therapy , DNA Methylation , Epigenomics , Humans
13.
Neuropsychopharmacology ; 41(11): 2782-93, 2016 10.
Article in English | MEDLINE | ID: mdl-27296153

ABSTRACT

The effect of preconception drinking by the mother on the life-long health outcomes of her children is not known, and therefore, in this study using an animal model, we determined the impact of preconception alcohol drinking of the mother on offspring stress response during adulthood. In our preconception alcohol exposure model, adult female rats were fed with 6.7% alcohol in their diet for 4 weeks, went without alcohol for 3 weeks and were bred to generate male and female offspring. Preconception alcohol-exposed offsprings' birth weight, body growth, stress response, anxiety-like behaviors, and changes in stress regulatory gene and protein hormone levels were evaluated. In addition, roles of epigenetic mechanisms in preconception alcohol effects were determined. Alcohol feeding three weeks prior to conception significantly affected pregnancy outcomes of female rats, with respect to delivery period and birth weight of offspring, without affecting maternal care behaviors. Preconception alcohol negatively affected offspring adult health, producing an increased stress hormone response to an immune challenge. In addition, preconception alcohol was associated with changes in expression and methylation profiles of stress regulatory genes in various brain areas. These changes in stress regulatory genes were normalized following treatment with a DNA methylation blocker during the postnatal period. These data highlight the novel possibility that preconception alcohol affects the inheritance of stress-related diseases possibly by epigenetic mechanisms.


Subject(s)
Anxiety/physiopathology , Central Nervous System Depressants/adverse effects , Ethanol/adverse effects , Maternal Behavior/physiology , Prenatal Exposure Delayed Effects/physiopathology , Stress, Psychological/etiology , Animals , Animals, Newborn , Anxiety/etiology , Central Nervous System Depressants/administration & dosage , Central Nervous System Depressants/pharmacology , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , DNA Methylation/drug effects , DNA Methylation/genetics , Ethanol/administration & dosage , Female , Gene Expression Regulation/drug effects , Lipopolysaccharides/adverse effects , Male , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/metabolism , Pregnancy , Pregnancy Outcome , Rats , Rats, Inbred F344 , Receptors, Mineralocorticoid/genetics , Receptors, Mineralocorticoid/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism
14.
PLoS One ; 10(10): e0140699, 2015.
Article in English | MEDLINE | ID: mdl-26509893

ABSTRACT

Recent evidence indicated that alcohol exposure during the fetal period increases the susceptibility to tumor development in mammary and prostate tissues. Whether fetal alcohol exposure increases the susceptibility to prolactin-producing tumor (prolactinoma) development in the pituitary was studied by employing the animal model of estradiol-induced prolactinomas in Fischer 344 female rats. We employed an animal model of fetal alcohol exposure that simulates binge alcohol drinking during the first two trimesters of human pregnancy and involves feeding pregnant rats with a liquid diet containing 6.7% alcohol during gestational day 7 to day 21. Control rats were pair-fed with isocaloric liquid diet or fed ad libitum with rat chow diet. Adult alcohol exposed and control female offspring rats were used in this study on the day of estrus or after estrogen treatment. Results show that fetal alcohol-exposed rats had increased levels of pituitary weight, pituitary prolactin (PRL) protein and mRNA, and plasma PRL. However, these rats show decreased pituitary levels of dopamine D2 receptor (D2R) mRNA and protein and increased pituitary levels of D2R promoter methylation. Also, they show elevated pituitary mRNA levels of DNA methylating genes (DNMT1, DNMT3b, MeCP2) and histone modifying genes (HDAC2, HDAC4, G9a). When fetal alcohol exposed rats were treated neonatally with a DNA methylation inhibitor 5-Aza deoxycytidine and/or a HDAC inhibitor trichostatin-A their pituitary D2R mRNA, pituitary weights and plasma PRL levels were normalized. These data suggest that fetal alcohol exposure programs the pituitary to increase the susceptibility to the development of prolactinomas possibly by enhancing the methylation of the D2R gene promoter and repressing the synthesis and control of D2R on PRL-producing cells.


Subject(s)
Epigenesis, Genetic , Pituitary Gland/pathology , Prenatal Exposure Delayed Effects/blood , Prolactin/biosynthesis , Receptors, Dopamine D2/metabolism , Animals , Cell Proliferation , CpG Islands/genetics , DNA Methylation/genetics , Estrogens/pharmacology , Female , Histones/metabolism , Lactotrophs/drug effects , Lactotrophs/metabolism , Lactotrophs/pathology , Mitogens/pharmacology , Organ Size , Pituitary Gland/metabolism , Pregnancy , Prolactin/blood , Promoter Regions, Genetic , Protein Processing, Post-Translational , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Inbred F344 , Receptors, Dopamine D2/genetics , Transcription, Genetic
15.
PLoS One ; 9(11): e113228, 2014.
Article in English | MEDLINE | ID: mdl-25409090

ABSTRACT

Proopiomelanocortin (POMC) is a precursor gene of the neuropeptide ß-endorphin in the hypothalamus and is known to regulate various physiological functions including stress response. Several recent reports showed that fetal alcohol exposure programs the hypothalamus to produce lower levels of POMC gene transcripts and to elevate the hypothalamic-pituitary-adrenal (HPA) axis response to stressful stimuli. We investigated the role of methyl CpG binding protein (MeCP2) in the effects of prenatal ethanol on POMC gene expression and hypothalamic-pituitary-adrenal (HPA) axis function. Pregnant Sprague Dawley rats were fed between GD 7 and 21 with a liquid diet containing 6.7% alcohol, pair-fed with isocaloric liquid diet, or fed ad libitum with rat chow, and their male offsprings were used at 60 days after birth in this study. Fetal alcohol exposure reduced the level of POMC mRNA, but increased the level of DNA methylation of this gene in the arcuate nucleus (ARC) of the hypothalamus where the POMC neuronal cell bodies are located. Fetal alcohol exposed rats showed a significant increase in MeCP2 protein levels in POMC cells, MeCP2 gene transcript levels as well as increased MeCP2 protein binding on the POMC promoter in the arcuate nucleus. Lentiviral delivery of MeCP2 shRNA into the third ventricle efficiently reduced MeCP2 expression and prevented the effect of prenatal ethanol on POMC gene expression in the arcuate nucleus. MeCP2-shRNA treatment also normalized the prenatal ethanol-induced increase in corticotropin releasing hormone (CRH) gene expression in the hypothalamus and elevated plasma adrenocorticotrophic hormone (ACTH) and corticosterone hormone responses to lipopolysaccharide (LPS) challenge. These results suggest that fetal alcohol programming of POMC gene may involve recruitment of MeCP2 on to the methylated promoter of the POMC gene to suppress POMC transcript levels and contribute to HPA axis dysregulation.


Subject(s)
Ethanol/adverse effects , Hypothalamo-Hypophyseal System/metabolism , Methyl-CpG-Binding Protein 2/metabolism , Pituitary-Adrenal System/metabolism , Prenatal Exposure Delayed Effects/genetics , Pro-Opiomelanocortin/genetics , Animals , Arcuate Nucleus of Hypothalamus/metabolism , DNA Methylation , Female , Gene Expression Regulation , Male , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/metabolism , Pro-Opiomelanocortin/metabolism , Promoter Regions, Genetic , Rats , Rats, Sprague-Dawley
16.
J Neurosci ; 34(43): 14181-97, 2014 Oct 22.
Article in English | MEDLINE | ID: mdl-25339733

ABSTRACT

Neurosteroids are endogenous regulators of neuronal excitability and seizure susceptibility. Neurosteroids, such as allopregnanolone (AP; 3α-hydroxy-5α-pregnan-20-one), exhibit enhanced anticonvulsant activity in perimenstrual catamenial epilepsy, a neuroendocrine condition in which seizures are clustered around the menstrual period associated with neurosteroid withdrawal (NSW). However, the molecular mechanisms underlying such enhanced neurosteroid sensitivity remain unclear. Neurosteroids are allosteric modulators of both synaptic (αßγ2-containing) and extrasynaptic (αßδ-containing) GABAA receptors, but they display greater sensitivity toward δ-subunit receptors in dentate gyrus granule cells (DGGCs). Here we report a novel plasticity of extrasynaptic δ-containing GABAA receptors in the dentate gyrus in a mouse perimenstrual-like model of NSW. In molecular and immunofluorescence studies, a significant increase occurred in δ subunits, but not α1, α2, ß2, and γ2 subunits, in the dentate gyrus of NSW mice. Electrophysiological studies confirmed enhanced sensitivity to AP potentiation of GABA-gated currents in DGGCs, but not in CA1 pyramidal cells, in NSW animals. AP produced a greater potentiation of tonic currents in DGGCs of NSW animals, and such enhanced AP sensitivity was not evident in δ-subunit knock-out mice subjected to a similar withdrawal paradigm. In behavioral studies, mice undergoing NSW exhibited enhanced seizure susceptibility to hippocampus kindling. AP has enhanced anticonvulsant effects in fully kindled wild-type mice, but not δ-subunit knock-out mice, undergoing NSW-induced seizures, confirming δ-linked neurosteroid sensitivity. These results indicate that perimenstrual NSW is associated with striking upregulation of extrasynaptic, δ-containing GABAA receptors that mediate tonic inhibition and neurosteroid sensitivity in the dentate gyrus. These findings may represent a molecular rationale for neurosteroid therapy of catamenial epilepsy.


Subject(s)
Menstrual Cycle/physiology , Neural Inhibition/physiology , Receptors, GABA-A/physiology , Synapses/physiology , Animals , Female , Hippocampus/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurotransmitter Agents/physiology , Organ Culture Techniques
17.
J Pharmacol Exp Ther ; 346(1): 146-60, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23667248

ABSTRACT

The ovarian cycle affects susceptibility to behavioral and neurologic conditions. The molecular mechanisms underlying these changes are poorly understood. Deficits in cyclical fluctuations in steroid hormones and receptor plasticity play a central role in physiologic and pathophysiologic menstrual conditions. It has been suggested that synaptic GABA(A) receptors mediate phasic inhibition in the hippocampus and extrasynaptic receptors mediate tonic inhibition in the dentate gyrus. Here we report a novel role of extrasynaptic δ-containing GABA(A) receptors as crucial mediators of the estrous cycle-related changes in neuronal excitability in mice, with hippocampus subfield specificity. In molecular and immunofluorescence studies, a significant increase occurred in δ-subunit, but not α4- and γ2-subunits, in the dentate gyrus during diestrus. However, δ-subunit upregulation was not evident in the CA1 region. The δ-subunit expression was undiminished by age and ovariectomy and in mice lacking progesterone receptors, but it was significantly reduced by finasteride, a neurosteroid synthesis inhibitor. Electrophysiologic studies confirmed greater potentiation of GABA currents by progesterone-derived neurosteroid allopregnanolone in dissociated dentate gyrus granule cells in diestrus than in CA1 pyramidal cells. The baseline conductance and allopregnanolone potentiation of tonic currents in dentate granule cells from hippocampal slices were higher than in CA1 pyramidal cells. In behavioral studies, susceptibility to hippocampus kindling epileptogenesis was lower in mice during diestrus. These results demonstrate the estrous cycle-related plasticity of neurosteroid-sensitive, δ-containing GABA(A) receptors that mediate tonic inhibition and seizure susceptibility. These findings may provide novel insight on molecular cascades of menstrual disorders like catamenial epilepsy, premenstrual syndrome, and migraine.


Subject(s)
Dentate Gyrus/metabolism , Epilepsy/etiology , Estrous Cycle , GABAergic Neurons/metabolism , Nerve Tissue Proteins/metabolism , Neural Inhibition , Receptors, GABA-A/metabolism , 5-alpha Reductase Inhibitors/pharmacology , Animals , Behavior, Animal , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/pathology , Dentate Gyrus/cytology , Dentate Gyrus/drug effects , Dentate Gyrus/pathology , Disease Susceptibility , Epilepsy/blood , Epilepsy/metabolism , Epilepsy/pathology , Female , GABAergic Neurons/cytology , GABAergic Neurons/drug effects , GABAergic Neurons/pathology , Gene Expression Regulation , In Vitro Techniques , Kindling, Neurologic , Mice , Mice, Knockout , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neural Inhibition/drug effects , Neuronal Plasticity , Pregnanolone/metabolism , Progesterone/blood , Protein Subunits/metabolism , Receptors, GABA-A/biosynthesis , Receptors, GABA-A/genetics , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism
18.
Neuropharmacology ; 59(7-8): 573-81, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20804775

ABSTRACT

Progesterone (P) is an endogenous anticonvulsant hormone. P is being evaluated as a treatment for epilepsy, traumatic brain injury, and other complex neurological conditions. Preclinical and clinical studies suggest that P appears to interrupt epileptogenic events. However, the potential disease-modifying effect of P in epileptogenic models is not widely investigated. In this study, we examined the effects of P on the development of hippocampus kindling in female mice. In addition, we determined the role of progesterone receptors (PR) in the P's effect on the kindling epileptogenesis utilizing PR knockout (PRKO) mice. P, at 25 mg/kg, did not affect seizures and did not exert sedative/motor effects in fully-kindled mice. P treatment (25 mg/kg, twice daily for 2 weeks) significantly suppressed the rate of development of behavioral kindled seizure activity evoked by daily hippocampus stimulation in wild-type (WT) mice, indicating a disease-modifying effect of P on limbic epileptogenesis. There was a significant increase in the rate of 'rebound or withdrawal' kindling during drug-free stimulation sessions following abrupt discontinuation of P treatment. A washout period after termination of P treatment prevented such acceleration in kindling. PRKO mice were kindled significantly slower than WT mice, indicating a modulatory role of PRs in seizure susceptibility. P's effects on early kindling progression was partially decreased in PRKO mice, but the overall (˜2-fold) delay in the rate of kindling for the induction of stage 5 seizures was unchanged in PRKO mice. Moreover, the acute anticonvulsant effect of P was undiminished in fully-kindled PRKO mice. These studies suggest that P exerts disease-modifying effects in the hippocampus kindling model at doses that do not significantly affect seizure expression and motor performance, and the kindling-retarding effects of P may occur partly through a complex PR-dependent and PR-independent mechanism.


Subject(s)
Epilepsy/metabolism , Kindling, Neurologic , Progesterone/physiology , Receptors, Progesterone/genetics , Animals , Anticonvulsants/blood , Anticonvulsants/pharmacology , Epilepsy/etiology , Epilepsy/physiopathology , Female , Hippocampus/drug effects , Hippocampus/physiopathology , Kindling, Neurologic/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Progesterone/blood , Progesterone/pharmacology , Seizures/etiology , Seizures/physiopathology
19.
Mol Cancer Res ; 7(6): 851-62, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19531572

ABSTRACT

The H3K9me2 histone methyltransferases G9a and GLP repress Mage-a class cancer germ-line (CG) antigen gene expression in murine embryonic stem (ES) cells, but the role of these enzymes in CG antigen gene regulation in human cancer cells is unknown. Here we show that whereas independent or dual knockdown of G9a and GLP in human cancer cells leads to reduced global and CG antigen promoter-associated H3K9me2 levels, it does not activate CG antigen gene expression. Moreover, CG antigen gene repression is maintained following pharmacologic targeting of G9a or treatment of G9a knockdown cells with the histone deacetylase inhibitor trichostatin A. However, G9a knockdown cells display increased sensitivity to CG antigen gene activation mediated by the DNA methyltransferase inhibitor decitabine. To account for these findings, we examined DNA methylation at CG antigen gene promoters in both cell types. We found robust DNA hypomethylation in G9a/GLP targeted murine ES cells but a lack of DNA methylation changes in G9a/GLP targeted human cancer cells; intriguingly, this distinction also extended to markers of global DNA methylation. These data reveal that G9a/GLP is required for DNA methylation of CG antigen genes and genomic DNA in murine ES cells, but not human cancer cells, and implicate DNA methylation status as the key epigenetic mechanism involved in CG antigen gene repression.


Subject(s)
Antigens, Neoplasm/metabolism , Embryonic Stem Cells/metabolism , Histocompatibility Antigens/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Animals , Antigens, Neoplasm/genetics , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Line , Cell Line, Tumor , DNA Methylation , Decitabine , Embryonic Stem Cells/immunology , Gene Expression/drug effects , Gene Expression Regulation , Histocompatibility Antigens/genetics , Histone Deacetylase Inhibitors , Histone Deacetylases/pharmacology , Histone-Lysine N-Methyltransferase/genetics , Humans , Hydroxamic Acids/pharmacology , Mice , Mice, Knockout
20.
J Neurosci Methods ; 181(1): 58-66, 2009 Jun 30.
Article in English | MEDLINE | ID: mdl-19406150

ABSTRACT

The GABA-A receptor plays a critical role in inhibitory neurotransmission in the brain. Quantitation of GABA-A receptor subunits in various brain regions is essential to understand their role in plasticity and brain disorders. However, conventional RNA assays are tedious and less sensitive for use in studies of subunit plasticity. Here we describe optimization of a sensitive assay of GABA-A receptor subunit gene expression by TaqMan real-time PCR. For each subunit gene, a set of primers and TaqMan fluorogenic probe were designed to specifically amplify the target template. The TaqMan methodology was optimized for quantification of mouse GABA-A receptor subunits (alpha(1-6), beta(1-3), gamma(2), and delta) and GAPDH. The TaqMan reaction detected very low levels of gene expression ( approximately 100 template copies of cDNA). A standard curve for GAPDH and one of the target genes, constructed using the cDNA, revealed slopes around -3.4 (r(2)=0.990), reflecting similar optimum PCR efficiencies. The methodology was utilized for quantification of the GABA-A receptor alpha(4)-subunit, which is known to upregulate following withdrawal from chronic progesterone or neurosteroids. Our results show that the alpha(4)-subunit expression increased threefold in the hippocampus following neurosteroid withdrawal in mice. The TaqMan PCR assay allows sensitive, high-throughput transcriptional profiling of complete GABA-A receptor subunit family, and thus provides specific tool for studies of GABA-A receptor subunit plasticity in neurological and psychiatric animal models.


Subject(s)
Brain/metabolism , Gene Expression Profiling/methods , Polymerase Chain Reaction/methods , Receptors, GABA-A/genetics , Animals , Female , Mice , Mice, Inbred C57BL , Protein Subunits/genetics , Protein Subunits/metabolism , RNA, Messenger/metabolism , Receptors, GABA-A/metabolism , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...