Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(5)2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38474260

ABSTRACT

The rat model of perinatal stress (PRS), in which exposure of pregnant dams to restraint stress reduces maternal behavior, is characterized by a metabolic profile that is reminiscent of the "metabolic syndrome". We aimed to identify plasma metabolomic signatures linked to long-term programming induced by PRS in aged male rats. This study was conducted in the plasma and frontal cortex. We also investigated the reversal effect of postpartum carbetocin (Cbt) on these signatures, along with its impact on deficits in cognitive, social, and exploratory behavior. We found that PRS induced long-lasting changes in biomarkers of secondary bile acid metabolism in the plasma and glutathione metabolism in the frontal cortex. Cbt treatment demonstrated disease-dependent effects by reversing the metabolite alterations. The metabolomic signatures of PRS were associated with long-term cognitive and emotional alterations alongside endocrinological disturbances. Our findings represent the first evidence of how early life stress may alter the metabolomic profile in aged individuals, thereby increasing vulnerability to CNS disorders. This raises the intriguing prospect that the pharmacological activation of oxytocin receptors soon after delivery through the mother may rectify these alterations.


Subject(s)
Adverse Childhood Experiences , Oxytocin , Pregnancy , Female , Humans , Rats , Animals , Male , Oxytocin/metabolism , Mothers , Stress, Psychological/metabolism , Postpartum Period , Brain/metabolism , Metabolome
2.
J Clin Transl Sci ; 7(1): e263, 2023.
Article in English | MEDLINE | ID: mdl-38229904

ABSTRACT

Stress and diabetes coexist in a vicious cycle. Different types of stress lead to diabetes, while diabetes itself is a major life stressor. This was the focus of the Chicago Biomedical Consortium's 19th annual symposium, "Stress and Human Health: Diabetes," in November 2022. There, researchers primarily from the Chicago area met to explore how different sources of stress - from the cells to the community - impact diabetes outcomes. Presenters discussed the consequences of stress arising from mutant proteins, obesity, sleep disturbances, environmental pollutants, COVID-19, and racial and socioeconomic disparities. This symposium showcased the latest diabetes research and highlighted promising new treatment approaches for mitigating stress in diabetes.

3.
Pharmacol Ther ; 240: 108299, 2022 12.
Article in English | MEDLINE | ID: mdl-36323379

ABSTRACT

Alcohol Use Disorder (AUD) is a multifaceted relapsing disorder that is commonly comorbid with psychiatric disorders, including anxiety. Alcohol exposure produces a plethora of effects on neurobiology. Currently, therapeutic strategies are limited, and only a few treatments - disulfiram, acamprosate, and naltrexone - are available. Given the complexity of this disorder, there is a great need for the identification of novel targets to develop new pharmacotherapy. The GABAergic system, the primary inhibitory system in the brain, is one of the well-known targets for alcohol and is responsible for the anxiolytic effects of alcohol. Interestingly, GABAergic neurotransmission is fine-tuned by neuroactive steroids that exert a regulatory role on several endocrine systems involved in neuropsychiatric disorders including AUD. Mounting evidence indicates that alcohol alters the biosynthesis of neurosteroids, whereas acute alcohol increases and chronic alcohol decreases allopregnanolone levels. Our recent work highlighted that chronic alcohol-induced changes in neurosteroid levels are mediated by epigenetic modifications, e.g., DNA methylation, affecting key enzymes involved in neurosteroid biosynthesis. These changes were associated with changes in GABAA receptor subunit expression, suggesting an imbalance between excitatory and inhibitory signaling in AUD. This review will recapitulate the role of neurosteroids in the regulation of the neuroendocrine system, highlight their role in the observed allostatic load in AUD, and develop a framework from mechanisms to potential pharmacotherapy.


Subject(s)
Alcoholism , Neurosteroids , Humans , Pregnanolone/metabolism , Alcoholism/drug therapy , Receptors, GABA-A/metabolism , Anxiety , Ethanol
4.
Transl Psychiatry ; 12(1): 109, 2022 03 16.
Article in English | MEDLINE | ID: mdl-35296634

ABSTRACT

The genomic effects of circulating glucocorticoids are particularly relevant in cortico-limbic structures, which express a high concentration of steroid hormone receptors. To date, no studies have investigated genomic differences in hippocampal subregions, namely the dorsal (dHPC) and ventral (vHPC) hippocampus, in preclinical models treated with exogenous glucocorticoids. Chronic oral corticosterone (CORT) in mouse is a pharmacological approach that disrupts the activity of the hypothalamic-pituitary-adrenal axis, increases affective behavior, and induces genomic changes after stress in the HPC of wildtype (WT) mice and mice heterozygous for the gene coding for brain-derived neurotrophic factor Val66Met (hMet), a variant associated with genetic susceptibility to stress. Using RNA-sequencing, we investigated the genomic signatures of oral CORT in the dHPC and vHPC of WT and hMet male and female mice, and examined sex and genotype differences in response to oral CORT. Males under CORT showed lower glycemia and increased anxiety- and depression-like behavior compared to females that showed instead opposite affective behavior in response to CORT. Rank-rank-hypergeometric overlap (RRHO) was used to identify genes from a continuous gradient of significancy that were concordant across groups. RRHO showed that CORT-induced differentially expressed genes (DEGs) in WT mice and hMet mice converged in the dHPC of males and females, while in the vHPC, DEGs converged in males and diverged in females. The vHPC showed a higher number of DEGs compared to the dHPC and exhibited sex differences related to glucocorticoid receptor (GR)-binding genes and epigenetic modifiers. Methyl-DNA-immunoprecipitation in the vHPC revealed differential methylation of the exons 1C and 1F of the GR gene (Nr3c1) in hMet females. Together, we report behavioral and endocrinological sex differences in response to CORT, as well as epigenetic signatures that i) differ in the dHPC and vHPC,ii) are distinct in males and females, and iii) implicate differential methylation of Nr3c1 selectively in hMet females.


Subject(s)
Corticosterone , Hypothalamo-Hypophyseal System , Animals , Corticosterone/pharmacology , Epigenesis, Genetic , Female , Genotype , Hippocampus/metabolism , Hypothalamo-Hypophyseal System/metabolism , Male , Mice , Pituitary-Adrenal System/metabolism , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism
5.
Transl Psychiatry ; 11(1): 298, 2021 05 20.
Article in English | MEDLINE | ID: mdl-34016951

ABSTRACT

Alcohol use disorder (AUD) is highly comorbid with depression. Withdrawal from chronic alcohol drinking results in depression and understanding brain molecular mechanisms that drive withdrawal-related depression is important for finding new drug targets to treat these comorbid conditions. Here, we performed RNA sequencing of the rat hippocampus during withdrawal from chronic alcohol drinking to discover key signaling pathways involved in alcohol withdrawal-related depressive-like behavior. Data were analyzed by weighted gene co-expression network analysis to identify several modules of co-expressed genes that could have a common underlying regulatory mechanism. One of the hub, or highly interconnected, genes in module 1 that increased during alcohol withdrawal was the transcription factor, signal transducer and activator of transcription 3 (Stat3), a known regulator of immune gene expression. Total and phosphorylated (p)STAT3 protein levels were also increased in the hippocampus during withdrawal after chronic alcohol exposure. Further, pSTAT3 binding was enriched at the module 1 genes Gfap, Tnfrsf1a, and Socs3 during alcohol withdrawal. Notably, pSTAT3 and its target genes were elevated in the postmortem hippocampus of human subjects with AUD when compared with control subjects. To determine the behavioral relevance of STAT3 activation during alcohol withdrawal, we treated rats with the STAT3 inhibitor stattic and tested for sucrose preference as a measure of anhedonia. STAT3 inhibition alleviated alcohol withdrawal-induced anhedonia. These results demonstrate activation of STAT3 signaling in the hippocampus during alcohol withdrawal in rats and in human AUD subjects, and suggest that STAT3 could be a therapeutic target for reducing comorbid AUD and depression.


Subject(s)
STAT3 Transcription Factor , Transcriptome , Anhedonia , Animals , Ethanol , Hippocampus/metabolism , Rats , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism
6.
Schizophr Bull Open ; 2(1): sgab002, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33585819

ABSTRACT

Schizophrenia is a severe neuropsychiatric disorder associated with a wide array of transcriptomic and neurobiochemical changes. Genome-wide transcriptomic profiling conducted in postmortem brain have provided novel insights into the pathophysiology of this disorder, and identified biological processes including immune/inflammatory-related responses, metabolic, endocrine, and synaptic function. However, few studies have investigated whether similar changes are present in peripheral tissue. Here, we used RNA-sequencing to characterize transcriptomic profiles of lymphocytes in 18 nonpsychotic controls and 19 individuals with schizophrenia. We identified 2819 differentially expressed transcripts (P nominal < .05) in the schizophrenia group when compared to controls. Bioinformatic analyses conducted on a subset of 293 genes (P nominal < .01 and |log2 FC| > 0.5) highlighted immune/inflammatory responses as key biological processes in our dataset. Differentially expressed genes in lymphocytes were highly enriched in gene expression profiles associated with cortex layer 5a and immune cells. Thus, we investigated whether the changes in transcripts levels observed in lymphocytes could also be detected in the prefrontal cortex (PFC, BA10) in a second replication cohort of schizophrenia subjects. Remarkably, mRNA levels detected in the PFC and lymphocytes were in strong agreement, and measurements obtained using RNA-sequencing positively correlated with data obtained by reverse transcriptase-quantitative polymerase chain reaction analysis. Collectively, our work supports a role for immune dysfunction in the pathogenesis of schizophrenia and suggests that peripheral markers can be used as accessible surrogates to investigate putative central nervous system disruptions.

7.
Int Rev Neurobiol ; 156: 127-183, 2021.
Article in English | MEDLINE | ID: mdl-33461662

ABSTRACT

Chronic exposure to stress throughout lifespan alters brain structure and function, inducing a maladaptive response to environmental stimuli, that can contribute to the development of a pathological phenotype. Studies have shown that hypothalamic-pituitary-adrenal (HPA) axis dysfunction is associated with various neuropsychiatric disorders, including major depressive, alcohol use and post-traumatic stress disorders. Downstream actors of the HPA axis, glucocorticoids are critical mediators of the stress response and exert their function through specific receptors, i.e., the glucocorticoid receptor (GR), highly expressed in stress/reward-integrative pathways. GRs are ligand-activated transcription factors that recruit epigenetic actors to regulate gene expression via DNA methylation, altering chromatin structure and thus shaping the response to stress. The dynamic interplay between stress response and epigenetic modifiers suggest DNA methylation plays a key role in the development of stress surfeit disorders.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Stress, Psychological , Humans , Stress, Psychological/genetics , Stress, Psychological/metabolism
8.
Mol Psychiatry ; 26(3): 1029-1041, 2021 03.
Article in English | MEDLINE | ID: mdl-31239533

ABSTRACT

Environmental factors, including substance abuse and stress, cause long-lasting changes in the regulation of gene expression in the brain via epigenetic mechanisms, such as DNA methylation. We examined genome-wide DNA methylation patterns in the prefrontal cortex (PFC, BA10) of 25 pairs of control and individuals with alcohol use disorder (AUD), using the Infinium® MethylationEPIC BeadChip. We identified 5254 differentially methylated CpGs (pnominal < 0.005). Bioinformatic analyses highlighted biological processes containing genes related to stress adaptation, including the glucocorticoid receptor (encoded by NR3C1). Considering that alcohol is a stressor, we focused our attention on differentially methylated regions of the NR3C1 gene and validated the differential methylation of several genes in the NR3C1 network. Chronic alcohol drinking results in a significant increased methylation of the NR3C1 exon variant 1H, with a particular increase in the levels of 5-hydroxymethylcytosine over 5-methylcytosine. These changes in DNA methylation were associated with reduced NR3C1 mRNA and protein expression levels in PFC, as well as other cortico-limbic regions of AUD subjects when compared with controls. Furthermore, we show that the expression of several stress-responsive genes (e.g., CRF, POMC, and FKBP5) is altered in the PFC of AUD subjects. These stress-response genes were also changed in the hippocampus, a region that is highly susceptible to stress. These data suggest that alcohol-dependent aberrant DNA methylation of NR3C1 and consequent changes in other stress-related genes might be fundamental in the pathophysiology of AUD and lay the groundwork for treatments targeting the epigenetic mechanisms regulating NR3C1 in AUD.


Subject(s)
Alcoholism , Receptors, Glucocorticoid , Alcoholism/genetics , DNA Methylation/genetics , Epigenesis, Genetic/genetics , Hippocampus/metabolism , Humans , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism
9.
Int J Neuropsychopharmacol ; 24(2): 130-141, 2021 02 15.
Article in English | MEDLINE | ID: mdl-32968808

ABSTRACT

BACKGROUND: Alcohol use disorder (AUD) is a chronic relapsing brain disorder. GABAA receptor (GABAAR) subunits are a target for the pharmacological effects of alcohol. Neurosteroids play an important role in the fine-tuning of GABAAR function in the brain. Recently, we have shown that AUD is associated with changes in DNA methylation mechanisms. However, the role of DNA methylation in the regulation of neurosteroid biosynthesis and GABAergic neurotransmission in AUD patients remains under-investigated. METHODS: In a cohort of postmortem brains from 20 male controls and AUD patients, we investigated the expression of GABAAR subunits and neurosteroid biosynthetic enzymes and their regulation by DNA methylation mechanisms. Neurosteroid levels were quantified by gas chromatography-mass spectrometry. RESULTS: The α 2 subunit expression was reduced due to increased DNA methylation at the gene promoter region in the cerebellum of AUD patients, a brain area particularly sensitive to the effects of alcohol. Alcohol-induced alteration in GABAAR subunits was also observed in the prefrontal cortex. Neurosteroid biosynthesis was also affected with reduced cerebellar expression of the 18kDa translocator protein and 3α-hydroxysteroid dehydrogenase mRNAs. Notably, increased DNA methylation levels were observed at the promoter region of 3α-hydroxysteroid dehydrogenase. These changes were associated with markedly reduced levels of allopregnanolone and pregnanolone in the cerebellum. CONCLUSION: Given the key role of neurosteroids in modulating the strength of GABAAR-mediated inhibition, our data suggest that alcohol-induced impairments in GABAergic neurotransmission might be profoundly impacted by reduced neurosteroid biosynthesis most likely via DNA hypermethylation.


Subject(s)
Alcoholism/metabolism , Cerebellum/metabolism , DNA Methylation/physiology , Epigenesis, Genetic/physiology , Neurosteroids/metabolism , Prefrontal Cortex/metabolism , Pregnanolone/metabolism , Receptors, GABA-A/metabolism , gamma-Aminobutyric Acid/metabolism , Adult , Autopsy , Cohort Studies , Humans , Male , Protein Biosynthesis/physiology , Synaptic Transmission/physiology
10.
Nitric Oxide ; 100-101: 50-56, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32278831

ABSTRACT

The cerebellum is widely known as a motor structure because it regulates and controls motor learning, coordination, and balance. However, it is also critical for non-motor functions such as cognitive processing, sensory discrimination, addictive behaviors and mental disorders. The cerebellum has the highest relative abundance of neuronal nitric oxide synthase (nNos) and is sensitive to ethanol. Although it has been demonstrated that the interaction of γ-aminobutyric acid (GABA) and nitric oxide (NO) might play an important role in the regulation of ethanol-induced cerebellar ataxia, the molecular mechanisms through which ethanol regulates nNos function to elicit this behavioral effect have not been studied extensively. Here, we investigated the dose-dependent effects of acute ethanol treatment on motor impairment using the rotarod behavioral paradigm and the alterations of nNos mRNA expression in cerebellum, frontal cortex (FC), hippocampus and striatum. We also examined the link between acute ethanol-induced motor impairment and nNos by pharmacological manipulation of nNos function. We found that acute ethanol induced a dose-dependent elevation of ethanol blood levels which was associated with the impairment of motor coordination performance and decreased expression of cerebellar nNos. In contrast, acute ethanol increased nNos expression in FC but did not to change the expression for this enzyme in striatum and hippocampus. The effects of acute ethanol were attenuated by l-arginine, a precursor for NO and potentiated by 7-nitroindazole (7-NI), a selective inhibitor of nNos. Our data suggests that differential regulation of nNos mRNA expression in cerebellum and frontal cortex might be involved in acute ethanol-induced motor impairment.


Subject(s)
Alcohol-Induced Disorders, Nervous System/metabolism , Cerebellar Ataxia/metabolism , Ethanol/adverse effects , Nitric Oxide Synthase Type I/metabolism , Psychomotor Disorders/metabolism , Alcohol-Induced Disorders, Nervous System/chemically induced , Animals , Arginine/pharmacology , Cerebellar Ataxia/chemically induced , Cerebellum/drug effects , Cerebellum/metabolism , Enzyme Inhibitors/pharmacology , Frontal Lobe/drug effects , Frontal Lobe/metabolism , Indazoles/pharmacology , Male , Nitric Oxide Synthase Type I/antagonists & inhibitors , Psychomotor Disorders/chemically induced , Rats, Sprague-Dawley
11.
Front Mol Neurosci ; 12: 89, 2019.
Article in English | MEDLINE | ID: mdl-31118884

ABSTRACT

Stress and the circadian systems play a major role in an organism's adaptation to environmental changes. The adaptive value of the stress system is reactive while that of the circadian system is predictive. Dysfunctions in these two systems may account for many clinically relevant disorders. Despite the evidence that interindividual differences in stress sensitivity and in the functioning of the circadian system are related, there is limited integrated research on these topics. Moreover, sex differences in these systems are poorly investigated. We used the perinatal stress (PRS) rat model, a well-characterized model of maladaptive programming of reactive and predictive adaptation, to monitor the running wheel behavior in male and female adult PRS rats, under a normal light/dark cycle as well as in response to a chronobiological stressor (6-h phase advance/shift). We then analyzed across different time points the expression of genes involved in circadian clocks, stress response, signaling, and glucose metabolism regulation in the suprachiasmatic nucleus (SCN). In the unstressed control group, we found a sex-specific profile that was either enhanced or inverted by PRS. Also, PRS disrupted circadian wheel-running behavior by inducing a phase advance in the activity of males and hypoactivity in females and increased vulnerability to chronobiological stress in both sexes. We also observed oscillations of several genes in the SCN of the unstressed group in both sexes. PRS affected males to greater extent than females, with PRS males displaying a pattern similar to unstressed females. Altogether, our findings provide evidence for a specific profile of dysmasculinization induced by PRS at the behavioral and molecular level, thus advocating the necessity to include sex as a biological variable to study the set-up of circadian system in animal models.

12.
Alcohol ; 78: 79-87, 2019 08.
Article in English | MEDLINE | ID: mdl-30851364

ABSTRACT

Withdrawal from chronic alcohol drinking can cause depression, leading to an inability to function in daily life and an increased risk for relapse to harmful drinking. Understanding the causes of alcohol withdrawal-related depression may lead to new therapeutic targets for treatment. Epigenetic factors have recently emerged as important contributors to both depression and alcohol use disorder (AUD). Specifically, acetylation of the N-terminal tails of histone proteins that package DNA into nucleosomes is altered in stress-induced models of depression and during alcohol withdrawal. The goal of this study was to examine depression-like behavior during alcohol withdrawal and associated changes in histone acetylation and expression of histone deacetylase 2 (HDAC2) in the hippocampus, a brain region critical for mood regulation and depression. Male Sprague-Dawley rats were treated with the Lieber-DeCarli ethanol liquid diet for 15 days and then underwent withdrawal. Rats were treated with the HDAC inhibitor, suberoylanilide hydroxamic acid (SAHA), during withdrawal and were tested for depression-like behavior. In a separate group of rats, the hippocampus was analyzed for mRNA and protein expression of HDAC2 and levels of histone H3 lysine 9 acetylation (H3K9ac) during chronic ethanol exposure and withdrawal. Rats undergoing ethanol withdrawal exhibited depression-like behavior and had increased HDAC2 and decreased H3K9ac levels in specific structures of the hippocampus. Treatment with SAHA during withdrawal ameliorated depression-like behavior and normalized changes in hippocampal HDAC2 and H3K9ac levels. These results demonstrate that ethanol withdrawal causes an altered epigenetic state in the hippocampus. Treatment with an HDAC inhibitor can correct this state and alleviate depression-like symptoms developed during withdrawal. Targeting histone acetylation may be a novel strategy to reduce ethanol withdrawal-induced depression.


Subject(s)
Depression/drug therapy , Hippocampus/metabolism , Histone Deacetylase 2/chemistry , Histone Deacetylase Inhibitors/therapeutic use , Substance Withdrawal Syndrome/drug therapy , Vorinostat/therapeutic use , Acetylation , Animals , Epigenesis, Genetic , Histones/drug effects , Male , Rats , Rats, Sprague-Dawley
13.
Mol Pharmacol ; 95(1): 62-69, 2019 01.
Article in English | MEDLINE | ID: mdl-30397000

ABSTRACT

Schizophrenia (SZ), schizoaffective (SZA), and bipolar (BP) disorder are neurodevelopmental psychopathological conditions related, in part, to genetic load and, in part, to environmentally induced epigenetic dysregulation of chromatin structure and function in neocortical GABAergic, glutamatergic, and monoaminergic neurons. To test the above hypothesis, we targeted our scientific efforts on identifying whether the molecular epigenetic signature of postmortem brains of patients with SZ, SZA, and BP disorder are also present in the brains of adult mice born from dams prenatally restraint stressed (PRS) during gestation. The brains of PRS mice, which are similar to the brains of patients with SZ and BP disorder, show an ∼2-fold increased binding of DNMT1 to psychiatric candidate promoters (glutamic acid decarboxylase 67, Reelin, and brain-derived neurotrophic factor), leading to their hypermethylation, reduced expression, as well as the behavioral endophenotypes reminiscent of those observed in the above psychiatric disorders. To establish whether clozapine (CLO) produces its behavioral and molecular action through a causal involvement of DNA methylation/demethylation processes, we compared the epigenetic action of CLO with that of the DNMT1 competitive inhibitor N-phthalyl-l-tryptophan (RG108). The intracerebroventricular injection of RG108 (20 nmol/day per 5 days), similar to the systemic administration of CLO, corrects the altered behavioral and molecular endophenotypes that are typical of PRS mice. These results are consistent with an epigenetic etiology underlying the behavioral endophenotypic profile in PRS mice. Further, it suggests that PRS mice may be useful in the preclinical screening of antipsychotic drugs acting to correct altered epigenetic mechanisms.


Subject(s)
Brain/drug effects , Chromatin Assembly and Disassembly/drug effects , Chromatin/drug effects , Clozapine/pharmacology , Mental Disorders/genetics , Phthalimides/pharmacology , Tryptophan/analogs & derivatives , Animals , Antipsychotic Agents/pharmacology , Brain-Derived Neurotrophic Factor/genetics , Cell Adhesion Molecules, Neuronal/genetics , Chromatin/genetics , Chromatin Assembly and Disassembly/genetics , DNA Methylation/drug effects , DNA Methylation/genetics , Disease Models, Animal , Epigenesis, Genetic/drug effects , Extracellular Matrix Proteins/genetics , Female , Glutamate Decarboxylase/genetics , Male , Mice , Nerve Tissue Proteins/genetics , Pregnancy , Prenatal Exposure Delayed Effects/genetics , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , Reelin Protein , Serine Endopeptidases/genetics , Tryptophan/pharmacology
14.
Pharmacol Res Perspect ; 6(4): e00416, 2018 07.
Article in English | MEDLINE | ID: mdl-29951207

ABSTRACT

Corroborating evidence indicate that the downregulation of GABAA receptor subunit expression may underlie tolerance to the anticonvulsant and anxiolytic actions of benzodiazepine (BZ) ligands that act as full allosteric modulators (FAMs) of GABA actions at a variety of GABAA receptor subtypes. We and others have shown that 10-14 days treatment with increasing doses of diazepam (a FAM) resulted in anticonvulsant tolerance and decreased the expression of the α1 GABAA receptor subunit mRNA and protein in frontal cortex. In addition, we have also shown that long-term treatment with imidazenil, a partial allosteric modulator of GABA action at selective GABAA receptor subtypes, fail to change the expression of the α1 subunit mRNA or induce tolerance to its anticonvulsant or anxiolytic action. However, little is known regarding the potential role of epigenetic mechanisms on long-term BZ-induced downregulation of GABAA receptor subunit. Therefore, we examined the role of histone acetylation and DNA methylation mechanisms on long-term diazepam-induced downregulation of the α1 subunit mRNA expression in rat frontal cortex. We found that 10 days treatment with increasing doses of diazepam but not imidazenil decreased the expression of the α1 GABAA receptor subunit mRNA and promoter acetylation in frontal cortex. In addition, we also found that 10 days treatment with diazepam but not imidazenil increased the expression of histone deacetylase (HDAC) 1 and 2 in frontal cortex. Thus, the increased expression of HDAC1 and HDAC2 (class 1 HDACs) and consequently increased histone deacetylation mechanism of this class 1 HDACs, may underlie long-term diazepam-induced decreased expression of the α1 GABAA receptor subunit mRNA in frontal cortex.


Subject(s)
Anti-Anxiety Agents/pharmacology , Anticonvulsants/pharmacology , Diazepam/pharmacology , Histones/metabolism , Prefrontal Cortex/drug effects , Receptors, GABA-A/genetics , Acetylation , Animals , DNA Methylation , Down-Regulation , Histone Deacetylase 1/genetics , Histone Deacetylase 2/genetics , Male , Prefrontal Cortex/metabolism , Rats, Sprague-Dawley
15.
Psychoneuroendocrinology ; 93: 45-55, 2018 07.
Article in English | MEDLINE | ID: mdl-29689422

ABSTRACT

The interplay between experiences during critical developmental periods and later adult life is crucial in shaping individual variability in stress coping strategies. Exposure to stressful events in early life has strongly programs an individual's phenotype and adaptive capabilities. Until now, studies on programming and reversal strategies in early life stress animal models have been essentially limited to males. By using the perinatal stress (PRS) rat model (a model more sensitive to aging changes) in middle-aged females, we investigated the behavioral and endocrine responses following exposure in later life to an unpredictable chronic mild stress (uCMS) condition for six weeks. PRS by itself accelerated the ageing-related-disruption in the estrous cycle and led to reductions in the levels of estradiol. It also reduced motivational and risk-taking behavior in later life, with PRS females being characterized by a reduction in self-grooming in the splash test, in the exploration of the light compartment in the light/dark box test and in the time spent eating a palatable food in the novelty-induced suppression feeding test. PRS females showed impaired regulation of plasma glucose and insulin levels following a glucose challenge, with a hyperglycemic phenotype, and disrupted feedback of the HPA axis after acute stress with respect to controls. Remarkably, all PRS-induced alterations were modified by exposure to the uCMS procedure, thus resulting in a disease-dependent intervention; controls were not affected by uCMS, except for a slight and transient reduction in body weight, while PRS females displayed a reduced body weight gain for the entire duration of the uCMS procedure. Interestingly, the effects of uCMS on PRS females were still observed up to two months after its termination and the females displayed heightened rhythms of locomotor activity and enhanced sensitivity to reward with respect to controls exposed to uCMS. Our findings indicate that many parameters of the PRS female adult phenotype are shaped by both early and later life experiences in a non-additive way. As a consequence, early stressed individuals may be programmed with a more dynamic phenotype than non-stressed individuals.


Subject(s)
Adaptation, Psychological/physiology , Stress, Psychological/physiopathology , Age Factors , Animals , Behavior, Animal/physiology , Corticosterone/blood , Endocrine System , Female , Hypothalamo-Hypophyseal System/physiology , Parturition , Pituitary-Adrenal System/physiology , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Rats , Rats, Sprague-Dawley , Stress Disorders, Post-Traumatic/physiopathology , Stress, Psychological/metabolism
16.
Neurotoxicology ; 66: 138-149, 2018 05.
Article in English | MEDLINE | ID: mdl-29630914

ABSTRACT

Exposure of the mother to adverse events during pregnancy is known to induce pathological programming of the HPA axis in the progeny, thereby increasing the vulnerability to neurobehavioral disorders. Maternal care plays a crucial role in the programming of the offspring, and oxytocin plays a key role in mother/pup interaction. Therefore, we investigated whether positive modulation of maternal behavior by activation of the oxytocinergic system could reverse the long-term alterations induced by perinatal stress (PRS; gestational restraint stress 3 times/day during the last ten days of gestation) on HPA axis activity, risk-taking behavior in the elevated-plus maze, hippocampal mGlu5 receptor and gene expression in Sprague-Dawley rats. Stressed and control unstressed dams were treated during the first postpartum week with an oxytocin receptor agonist, carbetocin (1 mg/kg, i.p.). Remarkably, reduction of maternal behavior was predictive of behavioral disturbances in PRS rats as well as of the impairment of the oxytocin and its receptor gene expression. Postpartum carbetocin corrected the reduction of maternal behavior induced by gestational stress as well as the impaired oxytocinergic system in the PRS progeny, which was associated with reduced risk-taking behavior. Moreover, postpartum carbetocin had an anti-stress effect on HPA axis activity in the adult PRS progeny and increased hippocampal mGlu5 receptor expression in aging. In conclusion, the activation of the oxytocinergic system in the early life plays a protective role against the programming effect by adverse experiences and could be considered as a novel and powerful potential therapeutic target for stress-related disorders.


Subject(s)
Gene Expression , Maternal Behavior , Oxytocin/physiology , Risk-Taking , Stress, Psychological/metabolism , Stress, Psychological/psychology , Animals , Female , Gestational Age , Hippocampus/metabolism , Oxytocin/administration & dosage , Oxytocin/analogs & derivatives , Postpartum Period , Rats, Sprague-Dawley , Receptor, Metabotropic Glutamate 5/metabolism , Receptors, Oxytocin/metabolism , Stress, Psychological/genetics
17.
Int J Neuropsychopharmacol ; 20(12): 1013-1026, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29020412

ABSTRACT

Background: Cerebellum is an area of the brain particularly sensitive to the effects of acute and chronic alcohol consumption. Alcohol exposure decreases cerebellar Purkinje cell output by increasing GABA release from Golgi cells onto extrasynaptic α6/δ-containing GABAA receptors located on glutamatergic granule cells. Here, we studied whether chronic alcohol consumption induces changes in GABAA receptor subunit expression and whether these changes are associated with alterations in epigenetic mechanisms via DNA methylation. Methods: We used a cohort of postmortem cerebellum from control and chronic alcoholics, here defined as alcohol use disorders subjects (n=25/group). S-adenosyl-methionine/S-adenosyl-homocysteine were measured by high-performance liquid chromatography. mRNA levels of various genes were assessed by reverse transcriptase-quantitative polymerase chain reaction. Promoter methylation enrichment was assessed using methylated DNA immunoprecipitation and hydroxy-methylated DNA immunoprecipitation assays. Results: mRNAs encoding key enzymes of 1-carbon metabolism that determine the S-adenosyl-methionine/S-adenosyl-homocysteine ratio were increased, indicating higher "methylation index" in alcohol use disorder subjects. We found that increased methylation of the promoter of the δ subunit GABAA receptor was associated with reduced mRNA and protein levels in the cerebellum of alcohol use disorder subjects. No changes were observed in α1- or α6-containing GABAA receptor subunits. The expression of DNA-methyltransferases (1, 3A, and 3B) was unaltered, whereas the mRNA level of TET1, which participates in the DNA demethylation pathway, was decreased. Hence, increased methylation of the δ subunit GABAA receptor promoter may result from alcohol-induced reduction of DNA demethylation. Conclusion: Together, these results support the hypothesis that aberrant DNA methylation pathways may be involved in cerebellar pathophysiology of alcoholism. Furthermore, this work provides novel evidence for a central role of DNA methylation mechanisms in the alcohol-induced neuroadaptive changes of human cerebellar GABAA receptor function.


Subject(s)
Alcoholism/pathology , Carbon/metabolism , Cerebellum/metabolism , DNA Methylation/genetics , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Adenosine/analogs & derivatives , Adenosine/metabolism , Alcoholism/genetics , Chromatography, High Pressure Liquid , Cohort Studies , Female , Gene Expression/physiology , Humans , Immunoprecipitation , Male , Middle Aged , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism , Postmortem Changes , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Messenger/metabolism , S-Adenosylhomocysteine/metabolism , S-Adenosylmethionine/metabolism , Signal Transduction/genetics
18.
Pharmacol Res ; 105: 186-97, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26816085

ABSTRACT

Intracellular accumulation of hyperphosphorylated tau protein is linked to neuronal degeneration in Alzheimer's disease (AD). Mounting evidence suggests that tau phosphorylation and O-N-acetylglucosamine glycosylation (O-GlcNAcylation) are mutually exclusive post-translational modifications. O-GlcNAcylation depends on 3-5% of intracellular glucose that enters the hexosamine biosynthetic pathway. To our knowledge, the existence of an imbalance between tau phosphorylation and O-GlcNAcylation has not been reported in animal models of AD, as yet. Here, we used triple transgenic (3xTg-AD) mice at 12 months, an age at which hyperphosphorylated tau is already detected and associated with cognitive decline. In these mice, we showed that tau was hyperphosphorylated on both Ser396 and Thr205 in the hippocampus, and to a lower extent and exclusively on Thr205 in the frontal cortex. Tau O-GlcNAcylation, assessed in tau immunoprecipitates, was substantially reduced in the hippocampus of 3xTg-AD mice, with no changes in the frontal cortex or in the cerebellum. No changes in the expression of the three major enzymes involved in O-GlcNAcylation, i.e., glutamine fructose-6-phosphate amidotransferase, O-linked ß-N-acetylglucosamine transferase, and O-GlcNAc hydrolase were found in the hippocampus of 3xTg-AD mice. These data demonstrate that an imbalance between tau phosphorylation and O-GlcNAcylation exists in AD mice, and strengthens the hypothesis that O-GlcNAcylation might be targeted by disease modifying drugs in AD.


Subject(s)
Acetylglucosamine/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Hippocampus/metabolism , Hippocampus/pathology , tau Proteins/metabolism , Acylation , Animals , Brain/metabolism , Brain/pathology , Disease Models, Animal , Glycosylation , Humans , Male , Mice , Mice, Transgenic , Phosphorylation
19.
Psychoneuroendocrinology ; 62: 36-46, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26231445

ABSTRACT

Oxytocin receptors are known to modulate synaptic transmission and network activity in the hippocampus, but their precise function has been only partially elucidated. Here, we have found that activation of presynaptic oxytocin receptor with the potent agonist, carbetocin, enhanced depolarization-evoked glutamate release in the ventral hippocampus with no effect on GABA release. This evidence paved the way for examining the effect of carbetocin treatment in "prenatally restraint stressed" (PRS) rats, i.e., the offspring of dams exposed to repeated episodes of restraint stress during pregnancy. Adult PRS rats exhibit an anxious/depressive-like phenotype associated with an abnormal glucocorticoid feedback regulation of the hypothalamus-pituitary-adrenal (HPA) axis, and, remarkably, with a reduced depolarization-evoked glutamate release in the ventral hippocampus. Chronic systemic treatment with carbetocin (1mg/kg, i.p., once a day for 2-3 weeks) in PRS rats corrected the defect in glutamate release, anxiety- and depressive-like behavior, and abnormalities in social behavior, in the HPA response to stress, and in the expression of stress-related genes in the hippocampus and amygdala. Of note, carbetocin treatment had no effect on these behavioral and neuroendocrine parameters in prenatally unstressed (control) rats, with the exception of a reduced expression of the oxytocin receptor gene in the amygdala. These findings disclose a novel function of oxytocin receptors in the hippocampus, and encourage the use of oxytocin receptor agonists in the treatment of stress-related psychiatric disorders in adult life.


Subject(s)
Glutamic Acid/metabolism , Hippocampus/drug effects , Oxytocin/analogs & derivatives , Prenatal Exposure Delayed Effects/metabolism , Presynaptic Terminals/drug effects , Receptors, Oxytocin/agonists , Stress, Psychological/metabolism , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Female , Hippocampus/metabolism , Hippocampus/physiopathology , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/physiopathology , Oxytocin/pharmacology , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/physiopathology , Pregnancy , Prenatal Exposure Delayed Effects/physiopathology , Presynaptic Terminals/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Presynaptic/metabolism , Restraint, Physical , Social Behavior
20.
Adv Neurobiol ; 10: 27-44, 2015.
Article in English | MEDLINE | ID: mdl-25287534

ABSTRACT

Prenatal restraint stress (PRS) can induce persisting changes in individual's development. PRS increases anxiety and depression-like behaviors and induces changes in the hypothalamo-pituitary-adrenal (HPA) axis in adult PRS rats after exposure to stress. Since adaptive capabilities also depend on temporal organization and synchronization with the external environment, we studied the effects of PRS on circadian rhythms, including the sleep-wake cycle, that are parameters altered in depression. Using a restraint stress during gestation, we showed that PRS induced phase advances in hormonal/behavioral circadian rhythms in adult rats, and an increase in the amount of paradoxical sleep, positively correlated to plasma corticosterone levels. Plasma corticosterone levels were also correlated with immobility in the forced swimming test, indicating a depressive-like profile in the PRS rats. We observed comorbidity with anxiety-like profile on PRS rats that was correlated with a reduced release of glutamate in the ventral hippocampus. Pharmacological approaches aimed at modulating glutamate release may represent a novel therapeutic strategy to treat stress-related disorders. Finally, since depressed patients exhibit changes in HPA axis activity and in circadian rhythmicity as well as in the paradoxical sleep regulation, we suggest that PRS could represent an original animal model of depression.

SELECTION OF CITATIONS
SEARCH DETAIL
...