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










Publication year range
1.
Alcohol ; 2024 Jan 28.
Article in English | MEDLINE | ID: mdl-38290696

ABSTRACT

Ceftriaxone is an antibiotic that increases central nervous system (CNS) protein expression of the glutamate transporters GLT-1 and xCT and ameliorates pathological behaviors in rodent models of neurological disease and substance use disorder. However, little ceftriaxone passes through the blood-brain-barrier, the CNS binding partner of ceftriaxone is unknown, and ceftriaxone does not consistently upregulate GLT-1 and xCT in cell culture. Ceftriaxone alters the gut microbiome composition in rodents and humans, and the microbiome-gut-brain axis regulates drug-seeking. Thus, here we test the hypothesis that ceftriaxone reduces alcohol intake while ameliorating alcohol-induced disruption of the gut microbiome composition. Male and female Sprague-Dawley rats received intermittent access to alcohol (IAA) while controls received access to only water. Following 17 IAA sessions, ceftriaxone/vehicle treatment was given for 5 days. Analysis of the gut microbiome composition was assessed by 16S rRNA gene amplicon sequencing conducted on fecal pellets collected prior to and after alcohol consumption and following ceftriaxone treatment. Male rats displayed escalated alcohol intake and preference over the course of the 17 sessions; however, total alcohol intake did not differ between the sexes. Ceftriaxone reduced alcohol intake and preference in male and female rats. While alcohol affected a diverse set of amplicon sequencing variants (ASV), ceftriaxone markedly reduced the diversity of microbial communities reflected by a blooming of the Enterococcaceae family. The remaining effects of ceftriaxone, however, encompassed families both affected and unaffected by prior alcohol drinking and highlight the Ruminococcaceae and Muribaculaceae families as bidirectionally modulated by alcohol and ceftriaxone. Altogether, our study confirms that ceftriaxone reduces alcohol intake in rats and partially reverses alcohol-induced dysbiosis.

2.
BMC Genomics ; 23(1): 679, 2022 Oct 01.
Article in English | MEDLINE | ID: mdl-36183097

ABSTRACT

BACKGROUND: The importance of fathers' engagement in care and its critical role in the offspring's cognitive and emotional development is now well established. Yet, little is known on the underlying neurobiology due to the lack of appropriate animal models. In the socially monogamous and bi-parental prairie vole (Microtus ochrogaster), while 60-80% of virgin males show spontaneous paternal behaviors (Paternal), others display pup-directed aggression (Attackers). Here we took advantage of this phenotypic dichotomy and used RNA-sequencing in three important brain areas to characterize gene expression associated with paternal behaviors of Paternal males and compare it to experienced Fathers and Mothers. RESULTS: While Paternal males displayed the same range and extent of paternal behaviors as experienced Fathers, we observed structure-specific transcriptomic differences between parental behaviors phenotypes. Using differential expression, gene set expression, as well as co-expression network analyses, we found that phenotypic differences between Paternal males and Attackers were mainly reflected by the lateral septum (LS), and to a lower extent, the nucleus accumbens (NAc), transcriptomes. In the medial preoptic area (MPOA), the profiles of gene expression mainly reflected differences between females and males regardless of their parental behaviors phenotype. Functional enrichment analyses of those gene sets associated with Paternal males or Attackers in the LS and the NAc revealed the involvement of processes related to the mitochondria, RNA translation, protein degradation processes, as well as epigenetic regulation of gene expression. CONCLUSIONS: By leveraging the natural phenotypic differences in parental behaviors in virgin male prairie voles alongside fathers and mothers, we identified a marked structure- and phenotype-specific pattern of gene expression associated with spontaneous paternal behaviors independently from fatherhood and pair-bonding. The LS transcriptome related to the mitochondria, RNA translation, and protein degradation processes was thus highlighted as a primary candidate associated with the spontaneous display of paternal behaviors. Altogether, our observations further characterize the behavioral and transcriptomic signature of parental behaviors in the socially monogamous prairie vole and lay the groundwork to further our understanding of the molecular underpinnings of paternal behavior.


Subject(s)
Paternal Behavior , Transcriptome , Animals , Arvicolinae/genetics , Epigenesis, Genetic , Female , Grassland , Male , Paternal Behavior/physiology , RNA/metabolism
3.
Biol Psychiatry ; 91(1): 141-151, 2022 01 01.
Article in English | MEDLINE | ID: mdl-33549315

ABSTRACT

BACKGROUND: The ability to form enduring social bonds is characteristic of human nature, and impairments in social affiliation are central features of severe neuropsychiatric disorders including autism spectrum disorder and schizophrenia. Owing to its ability to form long-term pair-bonds, the socially monogamous prairie vole has emerged as an excellent model to study the neurobiology of social attachment. Despite the enduring nature of the bond, however, surprisingly few genes have been implicated in the pair-bonding process in either sex. METHODS: Male and female prairie voles (Microtus ochrogaster) were cohabitated with an opposite-sex partner for 24 hours or 3 weeks, and transcriptomic regulations in the nucleus accumbens were measured by RNA sequencing. RESULTS: We found sex-specific response patterns despite similar behavioral indicators of pair-bond establishment. Indeed, 24 hours of cohabitation with an opposite-sex partner induced widespread transcriptomic changes that remained sustained to some extent in females after 3 weeks but returned to baseline before a second set of regulations in males. This led to a highly sexually biased nucleus accumbens transcriptome at 3 weeks related to processes such as neurotransmission, protein turnover, and DNA transcription. In particular, we found sex-specific alterations of mitochondrial dynamics following cohabitation, with a shift toward fission in males. CONCLUSIONS: In addition to identifying the genes, networks, and pathways involved in the pair-bonding process in the nucleus accumbens, our work illustrates the vast extent of sex differences in the molecular mechanisms underlying pair-bonding in prairie voles and paves the way to further our understanding of the complex social bonding process.


Subject(s)
Autism Spectrum Disorder , Transcriptome , Animals , Arvicolinae , Female , Grassland , Humans , Male , Pair Bond , Sexual Behavior, Animal , Social Behavior
4.
Curr Top Behav Neurosci ; 54: 283-310, 2022.
Article in English | MEDLINE | ID: mdl-34595741

ABSTRACT

Aggression is a complex behavioral trait modulated by both genetic and environmental influences on gene expression. By controlling gene expression in a reversible yet potentially lasting manner in response to environmental stimulation, epigenetic mechanisms represent prime candidates in explaining both individual differences in aggression and the development of elevated aggressive behaviors following life adversity. In this manuscript, we review the evidence for an epigenetic basis in the development and expression of aggression in both humans and related preclinical animal models. In particular, we discuss reports linking DNA methylation, histone post-translational modifications, as well as non-coding RNA, to the regulation of a variety of genes implicated in the neurobiology of aggression including neuropeptides, the serotoninergic and dopaminergic systems, and stress response related systems. While clinical reports do reveal interesting patterns of DNA methylation underlying individual differences and experience-induced aggressive behaviors, they do, in general, face the challenge of linking peripheral observations to central nervous system regulations. Preclinical studies, on the other hand, provide detailed mechanistic insights into the epigenetic reprogramming of gene expression following life adversities. Although the functional link to aggression remains unclear in most, these studies together do highlight the involvement of epigenetic events driven by DNA methylation, histone modifications, and non-coding RNA in the neuroadaptations underlying the development and expression of aggression.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Aggression/physiology , Animals , DNA Methylation/genetics , Histone Code , RNA, Untranslated
5.
Neurosci Biobehav Rev ; 105: 305-317, 2019 10.
Article in English | MEDLINE | ID: mdl-29020607

ABSTRACT

Growing evidence has begun to elucidate the contribution of epigenetic mechanisms in the modulation and maintenance of gene expression and behavior. Histone acetylation is one such epigenetic mechanism, which has been shown to profoundly alter gene expression and behaviors. In this review, we begin with an overview of the major epigenetic mechanisms including histones acetylation. We next focus on recent evidence about the influence of environmental stimuli on various motivated behaviors through histone acetylation and highlight how histone deacetylase inhibitors can correct some of the pathologies linked to motivated behaviors including substance abuse, feeding and social attachments. Particularly, we emphasize that the effects of histone deacetylase inhibitors on motivated behaviors are time and context-dependent.


Subject(s)
Aggression/drug effects , Epigenesis, Genetic/drug effects , Feeding and Eating Disorders/drug therapy , Histone Deacetylase Inhibitors/pharmacology , Maternal Behavior/drug effects , Motivation/drug effects , Object Attachment , Social Behavior , Substance-Related Disorders/drug therapy , Humans
6.
PLoS One ; 13(4): e0195095, 2018.
Article in English | MEDLINE | ID: mdl-29614089

ABSTRACT

BACKGROUND: Understanding molecular mechanisms underlying the induction of learning and memory impairments remains a challenge. Recent investigations have shown that the activation of group I mGluRs (mGluR1 and mGluR5) in cultured hippocampal neurons by application of (S)-3,5-Dihydroxyphenylglycine (DHPG) causes the regulated internalization of N-methyl-D-aspartate receptors (NMDARs), which subsequently activates protein kinase D1 (PKD1). Through phosphorylating the C-terminals of the NMDAR GluN2 subunits, PKD1 down-regulates the activity of remaining (non-internalized) surface NMDARs. The knockdown of PKD1 does not affect the DHPG-induced inhibition of AMPA receptor-mediated miniature excitatory post-synaptic currents (mEPSCs) but prevents the DHPG-induced inhibition of NMDAR-mediated mEPSCs in vitro. Thus, we investigated the in vivo effects of bilateral infusions of DHPG into the hippocampal CA1 area of rats in the Morris water maze (MWM) and the novel object discrimination (NOD) tests. METHODS: A total of 300 adult male Sprague Dawley rats (250-280 g) were used for behavioral tests. One hundred ninety four were used in MWM test and the other 106 rats in the NOD test. Following one week of habituation to the vivarium, rats were bilaterally implanted under deep anesthesia with cannulas aimed at the CA1 area of the hippocampus (CA1 coordinates in mm from Bregma: AP -3.14; lateral +/-2; DV -3.0). Through implanted cannulas artificial cerebrospinal fluid (ACSF), the group1 mGluR antagonist 6-Methyl-2-(phenylethynyl)pyridine (MPEP), the dynamin-dependent internalization inhibitor Dynasore, or the PKD1 inhibitor CID755673 were infused into the bilateral hippocampal CA1 areas (2 µL per side, over 5 min). The effects of these infusions and the effects of PKD1 knockdown were examined in MWM or NOD test. RESULTS: DHPG infusion increased the latency to reach the platform in the MWM test and reduced the preference for the novel object in the NOD task. We found that the DHPG effects were dose-dependent and could be maintained for up to 2 days. Notably, these effects could be prevented by pre-infusion of the group1 mGluR antagonist MPEP, the dynamin-dependent internalization inhibitor Dynasore, the PKD1 inhibitor CID755673, or by PKD1 knockdown in the hippocampal CA1 area. CONCLUSION: Altogether, these findings provide direct evidence that PKD1-mediated signaling may play a critical role in the induction of learning and memory impairments by DHPG infusion into the hippocampal CA1 area.


Subject(s)
Hippocampus/metabolism , Hippocampus/physiopathology , Learning , Memory , Protein Kinase C/genetics , Animals , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Knockout Techniques , Learning Disabilities/etiology , Learning Disabilities/physiopathology , Locomotion , Male , Maze Learning , Memory Disorders/etiology , Memory Disorders/physiopathology , Methoxyhydroxyphenylglycol/adverse effects , Methoxyhydroxyphenylglycol/analogs & derivatives , Protein Kinase C/metabolism , Rats , Spatial Memory
7.
Curr Opin Behav Sci ; 14: 19-26, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28584860

ABSTRACT

In major depressive disorder, women exhibit higher lifetime prevalence and different antidepressant response rates than men, which illustrates the importance of examining individual differences in the pathophysiology of depression and therapeutic response. In recent years, the consideration of sex in related preclinical research has thus gained interest-particularly in light of novel evidence for rapid-acting antidepressants. Notably, the literature recently revealed a higher sensitivity of females to the antidepressant effects of the N-methyl-D-aspartate receptor antagonist ketamine, in both baseline and preclinical conditions. Combined with its fast-acting and relatively sustained properties, this evidence highlights ketamine as a particularly interesting therapeutic alternative for this sensitive population, and supports the value in considering sex as a critical factor for improved individualized therapeutic strategies.

8.
Front Behav Neurosci ; 11: 35, 2017.
Article in English | MEDLINE | ID: mdl-28321184

ABSTRACT

It is now clearly established that complex interactions between genes and environment are involved in multiple aspects of neuropsychiatric disorders, from determining an individual's vulnerability to onset, to influencing its response to therapeutic intervention. In this perspective, it appears crucial to better understand how the organism reacts to environmental stimuli and provide a coordinated and adapted response. In the central nervous system, neuronal plasticity and neurotransmission are among the major processes integrating such complex interactions between genes and environmental stimuli. In particular, immediate early genes (IEGs) are critical components of these interactions as they provide the molecular framework for a rapid and dynamic response to neuronal activity while opening the possibility for a lasting and sustained adaptation through regulation of the expression of a wide range of genes. As a result, IEGs have been tightly associated with neuronal activity as well as a variety of higher order processes within the central nervous system such as learning, memory and sensitivity to reward. The immediate early gene and transcription factor early growth response 1 (EGR1) has thus been revealed as a major mediator and regulator of synaptic plasticity and neuronal activity in both physiological and pathological conditions. In this review article, we will focus on the role of EGR1 in the central nervous system. First, we will summarize the different factors influencing its activity. Then, we will analyze the amount of data, including genome-wide, that has emerged in the recent years describing the wide variety of genes, pathways and biological functions regulated directly or indirectly by EGR1. We will thus be able to gain better insights into the mechanisms underlying EGR1's functions in physiological neuronal activity. Finally, we will discuss and illustrate the role of EGR1 in pathological states with a particular interest in cognitive functions and neuropsychiatric disorders.

10.
Neuropharmacology ; 109: 293-305, 2016 10.
Article in English | MEDLINE | ID: mdl-27343386

ABSTRACT

Only a portion of the population exposed to trauma will develop persistent emotional alterations characteristic of posttraumatic stress disorder (PTSD), which illustrates the necessity for identifying vulnerability factors and novel pharmacotherapeutic alternatives. Interestingly, clinical evidence suggests that novelty seeking is a good predictor for vulnerability to the development of excessive and persistent fear. Here, we first tested this hypothesis by analyzing contextual and cued fear responses of rats selected for their high (high responders, HR) or low (low responders, LR) exploration of a novel environment, indicator of novelty seeking. While HR and LR rats exhibited similar sensitivity to the shock and cued fear memory retention, fewer extinction sessions were required in HR than LR animals to reach extinction, indicating faster contextual and cued memory extinction. In a second part, we found an effective disruption of contextual fear reconsolidation by the N-methyl-d-aspartate receptor antagonist ketamine, associated with a down-regulation of early growth response 1 (Egr1) in the hippocampal CA1 area, and up-regulation of brain-derived neurotrophic factor (Bdnf) mRNA levels in the prelimbic and infralimbic cortices. Altogether, these data demonstrate a link between novelty seeking and conditioned fear extinction, and highlight a promising novel role of ketamine in affecting established fear memory.


Subject(s)
Exploratory Behavior/drug effects , Fear/drug effects , Fear/psychology , Individuality , Ketamine/pharmacology , Memory Consolidation/drug effects , Animals , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/physiology , Exploratory Behavior/physiology , Fear/physiology , Forecasting , Locomotion/drug effects , Locomotion/physiology , Male , Memory Consolidation/physiology , Rats , Rats, Sprague-Dawley
11.
Genome Biol ; 16: 256, 2015 Dec 02.
Article in English | MEDLINE | ID: mdl-26628058

ABSTRACT

BACKGROUND: Males and females differ in cognitive functions and emotional processing, which in part have been associated with baseline sex differences in gene expression in the medial prefrontal cortex. Nevertheless, a growing body of evidence suggests that sex differences in medial prefrontal cortex-dependent cognitive functions are attenuated by hormonal fluctuations within the menstrual cycle. Despite known genomic effects of ovarian hormones, the interaction of the estrous cycle with sex differences in gene expression in the medial prefrontal cortex remains unclear and warrants further investigations. RESULTS: We undertake a large-scale characterization of sex differences and their interaction with the estrous cycle in the adult medial prefrontal cortex transcriptome and report that females with high and low ovarian hormone levels exhibited a partly opposed sexually biased transcriptome. The extent of regulation within females vastly exceeds sex differences, and supports a multi-level reorganization of synaptic function across the estrous cycle. Genome-wide analysis of the transcription factor early growth response 1 binding highlights its role in controlling the synapse-related genes varying within females. CONCLUSIONS: We uncover a critical influence of the estrous cycle on the adult rat medial prefrontal cortex transcriptome resulting in partly opposite sex differences in proestrus when compared to diestrus females, and we discovered a direct role for Early Growth Response 1 in this opposite regulation. In addition to illustrating the importance of accounting for the estrous cycle in females, our data set the ground for a better understanding of the female specificities in cognition and emotional processing.


Subject(s)
Early Growth Response Protein 1/metabolism , Estrous Cycle , Prefrontal Cortex/metabolism , Sex Characteristics , Transcriptome , Animals , Early Growth Response Protein 1/genetics , Female , Genome , Male , Rats , Rats, Sprague-Dawley
12.
Mol Brain ; 8(1): 75, 2015 Nov 19.
Article in English | MEDLINE | ID: mdl-26584860

ABSTRACT

BACKGROUND: Constitutive and regulated internalization of cell surface proteins has been extensively investigated. The regulated internalization has been characterized as a principal mechanism for removing cell-surface receptors from the plasma membrane, and signaling to downstream targets of receptors. However, so far it is still not known whether the functional properties of remaining (non-internalized) receptor/channels may be regulated by internalization of the same class of receptor/channels. The N-methyl-D-aspartate receptor (NMDAR) is a principal subtype of glutamate-gated ion channel and plays key roles in neuronal plasticity and memory functions. NMDARs are well-known to undergo two types of regulated internalization - homologous and heterologous, which can be induced by high NMDA/glycine and DHPG, respectively. In the present work, we investigated effects of regulated NMDAR internalization on the activity of residual cell-surface NMDARs and neuronal functions. RESULTS: In electrophysiological experiments we discovered that the regulated internalization of NMDARs not only reduced the number of cell surface NMDARs but also caused an inhibition of the activity of remaining (non-internalized) surface NMDARs. In biochemical experiments we identified that this functional inhibition of remaining surface NMDARs was mediated by increased serine phosphorylation of surface NMDARs, resulting from the activation of protein kinase D1 (PKD1). Knockdown of PKD1 did not affect NMDAR internalization but prevented the phosphorylation and inhibition of remaining surface NMDARs and NMDAR-mediated synaptic functions. CONCLUSION: These data demonstrate a novel concept that regulated internalization of cell surface NMDARs not only reduces the number of NMDARs on the cell surface but also causes an inhibition of the activity of remaining surface NMDARs through intracellular signaling pathway(s). Furthermore, modulating the activity of remaining surface receptors may be an effective approach for treating receptor internalization-induced changes in neuronal functions of the CNS.


Subject(s)
Cell Membrane/metabolism , Endocytosis , Protein Kinase C/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Down-Regulation/drug effects , Endocytosis/drug effects , Enzyme Activation/drug effects , Gene Knockdown Techniques , Glycine/pharmacology , HEK293 Cells , Humans , Methoxyhydroxyphenylglycol/analogs & derivatives , Methoxyhydroxyphenylglycol/pharmacology , Mice , Models, Biological , N-Methylaspartate/pharmacology , Phosphorylation/drug effects , Phosphoserine/metabolism , Rats, Wistar
13.
J Neurosci ; 35(23): 8948-58, 2015 Jun 10.
Article in English | MEDLINE | ID: mdl-26063926

ABSTRACT

Epigenetic mechanisms, such as histone modifications, regulate responsiveness to drugs of abuse, such as cocaine, but relatively little is known about the regulation of addictive-like behaviors by DNA methylation. To investigate the influence of DNA methylation on the locomotor-activating effects of cocaine and on drug-seeking behavior, rats receiving methyl supplementation via chronic l-methionine (MET) underwent either a sensitization regimen of intermittent cocaine injections or intravenous self-administration of cocaine, followed by cue-induced and drug-primed reinstatement. MET blocked sensitization to the locomotor-activating effects of cocaine and attenuated drug-primed reinstatement, with no effect on cue-induced reinstatement or sucrose self-administration and reinstatement. Furthermore, upregulation of DNA methyltransferase 3a and 3b and global DNA hypomethylation were observed in the nucleus accumbens core (NAc), but not in the medial prefrontal cortex (mPFC), of cocaine-pretreated rats. Glutamatergic projections from the mPFC to the NAc are critically involved in the regulation of cocaine-primed reinstatement, and activation of both brain regions is seen in human addicts when reexposed to the drug. When compared with vehicle-pretreated rats, the immediate early gene c-Fos (a marker of neuronal activation) was upregulated in the NAc and mPFC of cocaine-pretreated rats after cocaine-primed reinstatement, and chronic MET treatment blocked its induction in both regions. Cocaine-induced c-Fos expression in the NAc was associated with reduced methylation at CpG dinucleotides in the c-Fos gene promoter, effects reversed by MET treatment. Overall, these data suggest that drug-seeking behaviors are, in part, attributable to a DNA methylation-dependent process, likely occurring at specific gene loci (e.g., c-Fos) in the reward pathway.


Subject(s)
Brain/drug effects , Cocaine-Related Disorders/physiopathology , Cocaine/adverse effects , DNA Methylation/drug effects , Dopamine Uptake Inhibitors/adverse effects , Drug-Seeking Behavior/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Animals , Body Weight/drug effects , Brain/metabolism , Cocaine-Related Disorders/etiology , Conditioning, Operant/drug effects , DNA Methyltransferase 3A , Disease Models, Animal , Drinking/drug effects , Eating/drug effects , Extinction, Psychological , Male , Proto-Oncogene Proteins c-fos/genetics , Rats , Rats, Sprague-Dawley , Reinforcement, Psychology , Self Administration , Sucrose/administration & dosage , Sweetening Agents/administration & dosage
14.
Biol Psychiatry ; 78(4): 259-69, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-25683735

ABSTRACT

BACKGROUND: While the influence of testosterone levels on vulnerability to affective disorders is not straightforward, research suggests this hormone may confer some degree of resiliency in men. We recently demonstrated a role for the dentate gyrus in mediating testosterone's protective effects on depressive-like behavior in gonadectomized male rats. Here, testosterone may exert its effects through androgen receptor-mediated mechanisms or via local aromatization to estradiol. METHODS: Gonadectomized male rats were implanted with a placebo, testosterone, or estradiol pellet, and subsequent protective anxiolytic- and antidepressant-like effects of testosterone and its aromatized metabolite, estradiol, were then investigated in the open field and sucrose preference tests, respectively. Moreover, their influence on gene expression in the hippocampus was analyzed by genome-wide complementary DNA microarray analysis. Finally, the contribution of testosterone's aromatization within the dentate gyrus was assessed by local infusion of the aromatase inhibitor fadrozole, whose efficacy was confirmed by liquid chromatography-tandem mass spectrometry. RESULTS: Both hormones had antidepressant-like effects associated with a substantial overlap in transcriptional regulation, particularly in synaptic plasticity- and mitogen-activated protein kinase pathway-related genes. Further, chronic aromatase inhibition within the dentate gyrus blocked the protective effects of testosterone. CONCLUSIONS: Both testosterone and estradiol exhibit anxiolytic- and antidepressant-like effects in gonadectomized male rats, while similarly regulating critical mediators of these behaviors, suggesting common underlying mechanisms. Accordingly, we demonstrated that testosterone's protective effects are mediated, in part, by its aromatization in the dentate gyrus. These findings thus provide further insight into a role for estradiol in mediating the protective anxiolytic- and antidepressant-like effects of testosterone.


Subject(s)
Anxiety/metabolism , Anxiety/physiopathology , Depression/metabolism , Depression/physiopathology , Estrogens/physiology , Testosterone/physiology , Animals , Anti-Anxiety Agents/administration & dosage , Antidepressive Agents/administration & dosage , Anxiety/genetics , Aromatase Inhibitors/pharmacology , Castration , Depression/genetics , Estrogens/administration & dosage , Fadrozole/pharmacology , Gene Expression/drug effects , Hippocampus/drug effects , Hippocampus/enzymology , Hippocampus/metabolism , Male , Motor Activity/drug effects , Rats , Rats, Sprague-Dawley , Testosterone/administration & dosage , Testosterone/metabolism
15.
J Exp Biol ; 218(Pt 1): 21-31, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25568448

ABSTRACT

Major depressive disorder (MDD) is a devastating neuropsychiatric disorder encompassing a wide range of cognitive and emotional dysfunctions. The prevalence of MDD is expected to continue its growth to become the second leading cause of disease burden (after HIV) by 2030. Despite an extensive research effort, the exact etiology of MDD remains elusive and the diagnostics uncertain. Moreover, a marked inter-individual variability is observed in the vulnerability to develop depression, as well as in response to antidepressant treatment, for nearly 50% of patients. Although a genetic component accounts for some cases of MDD, it is now clearly established that MDD results from strong gene and environment interactions. Such interactions could be mediated by epigenetic mechanisms, defined as chromatin and DNA modifications that alter gene expression without changing the DNA structure itself. Some epigenetic mechanisms have recently emerged as particularly relevant molecular substrates, promoting vulnerability or resilience to the development of depressive-like symptoms. Although the role of brain-derived neurotrophic factor (BDNF) in the pathophysiology of MDD remains unclear, its modulation of the efficacy of antidepressants is clearly established. Therefore, in this review, we focus on the epigenetic mechanisms regulating the expression of BDNF in humans and in animal models of depression, and discuss their role in individual differences in vulnerability to depression and response to antidepressant drugs.


Subject(s)
Antidepressive Agents/therapeutic use , Brain-Derived Neurotrophic Factor/genetics , Depression/drug therapy , Depression/genetics , Epigenesis, Genetic , Animals , Antidepressive Agents/pharmacology , Disease Models, Animal , Epigenesis, Genetic/drug effects , Genetic Predisposition to Disease , Humans
16.
J Neurosci ; 33(27): 11048-60, 2013 Jul 03.
Article in English | MEDLINE | ID: mdl-23825410

ABSTRACT

Some personality traits, including novelty seeking, are good predictors of vulnerability to stress-related mood disorders in both humans and rodents. While high-novelty-seeking rats [high responders (HRs)] are vulnerable to the induction of depressive-like symptoms by social defeat stress, low-novelty-seeking rats [low responders (LRs)] are not. Here, we show that such individual differences are critically regulated by hippocampal BDNF. While LR animals exhibited an increase in BDNF levels following social defeat, HR individuals did not. This difference in hippocampal BDNF expression promoted the vulnerability of HR and the resilience of LR rats. Indeed, preventing activation of BDNF signaling by infusing the BDNF scavenger TrkB-Fc into the dentate gyrus of the hippocampus of LR rats led to social defeat-induced social avoidance, whereas its activation in HR rats by the TrkB agonist 7,8-dihydroxyflavone promoted social approach. Along with the changes in BDNF expression following defeat, we report in LR animals a downregulation of the inactive BDNF receptor TrkB.T1, associated with an activation of CREB through Akt-mediated signaling, but not MSK1-mediated signaling. In HR animals, none of these molecules were affected by social defeat. Importantly, the BDNF upregulation involved an epigenetically controlled transcription of bdnf exon VI, associated with a coherent regulation of relevant epigenetic factors. Altogether, our data support the importance of hippocampal BDNF regulation in response to stressful events. Moreover, we identify a specific and adaptive regulation of bdnf exon VI in the hippocampus as a critical regulator of stress resilience, and strengthen the importance of epigenetic factors in mediating stress-induced adaptive and maladaptive responses in different individuals.


Subject(s)
Brain-Derived Neurotrophic Factor/biosynthesis , Epigenesis, Genetic/physiology , Exploratory Behavior/physiology , Gene Expression Regulation , Individuality , Motor Activity/physiology , Animals , Brain-Derived Neurotrophic Factor/genetics , Forecasting , Hippocampus/physiology , Male , Random Allocation , Rats , Rats, Sprague-Dawley , Social Behavior
17.
Nat Neurosci ; 16(7): 919-24, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23727821

ABSTRACT

In the socially monogamous prairie vole (Microtus ochrogaster), mating induces enduring pair-bonds that are initiated by partner preference formation and regulated by a variety of neurotransmitters, including oxytocin, vasopressin and dopamine. We examined potential epigenetic mechanisms mediating pair-bond regulation and found that the histone deacetylase inhibitors sodium butyrate and trichostatin A (TSA) facilitated partner preference formation in female prairie voles in the absence of mating. This was associated with a specific upregulation of oxytocin receptor (OTR, oxtr) and vasopressin V1a receptor (V1aR, avpr1a) in the nucleus accumbens (NAcc), through an increase in histone acetylation at their respective promoters. Furthermore, TSA-facilitated partner preference was prevented by OTR or V1aR blockade in the NAcc. Notably, mating-induced partner preference triggered the same epigenetic regulation of oxtr and avpr1a gene promoters as TSA. These observations indicate that TSA and mating facilitate partner preference through epigenetic events, providing, to the best of our knowledge, the first direct evidence for epigenetic regulation of pair-bonding.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Pair Bond , Sexual Behavior, Animal/drug effects , Animals , Arvicolinae/physiology , Dose-Response Relationship, Drug , Female , Injections, Intraventricular , Male , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , RNA, Messenger/metabolism , Receptors, Oxytocin/genetics , Receptors, Oxytocin/metabolism , Receptors, Vasopressin/genetics , Receptors, Vasopressin/metabolism , Time Factors , Up-Regulation/drug effects
18.
Physiol Behav ; 104(2): 296-305, 2011 Aug 03.
Article in English | MEDLINE | ID: mdl-21172365

ABSTRACT

Most neuropsychiatric disorders, including stress-related mood disorders, are complex multi-parametric syndromes. Diagnoses are therefore hard to establish and current therapeutic strategies suffer from significant variability in effectiveness, making the understanding of inter-individual variations crucial to unveiling effective new treatments. In rats, such individual differences are observed during exposure to a novel environment, where individuals will exhibit either high or low locomotor activity and can thus be separated into high (HR) and low (LR) responders, respectively. In rodents, a long-lasting, psychosocial, stress-induced depressive state can be triggered by exposure to a social defeat procedure. We therefore analyzed the respective vulnerabilities of HR and LR animals to long-lasting, social defeat-induced behavioral alterations relevant to mood disorders. Two weeks after four daily consecutive social defeat exposures, HR animals exhibit higher anxiety levels, reduced body weight gain, sucrose preference, and a marked social avoidance. LR animals, however, remain unaffected. Moreover, while repeated social defeat exposure induces long-lasting contextual fear memory in both HR and LR animals, only HR individuals exhibit marked freezing behavior four weeks after a single social defeat. Combined, these findings highlight the critical involvement of inter-individual variations in novelty-seeking behavior in the vulnerability to stress-related mood disorders, and uncover a promising model for posttraumatic stress disorder.


Subject(s)
Exploratory Behavior/physiology , Individuality , Stress, Psychological/physiopathology , Analysis of Variance , Animals , Avoidance Learning , Conditioning, Classical , Disease Models, Animal , Dominance-Subordination , Fear , Female , Food Preferences , Male , Memory , Motor Activity/physiology , Orchiectomy , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Sucrose/administration & dosage , Time Factors
19.
Neurosci Lett ; 475(3): 179-83, 2010 May 21.
Article in English | MEDLINE | ID: mdl-20371377

ABSTRACT

P300/CBP associated factor (PCAF) acts as an acetyltransferase that acetylates specific lysine residues in histones, thereby remodelling chromatin structure. The possible involvement of PCAF in learning and memory processes or mood disorders was recently assessed by characterizing the behavioural phenotype of PCAF KO mice bred on a CD1 background and revealed short-term memory deficits that evolved with age towards long-term memory alteration and an exaggerated response to stress [10]. PCAF KO mice have been backcrossed on a C57BL/6j strain for 15 generations and we report here the first data regarding their behavioural phenotype. PCAF KO mice bred on a C57 background showed short-term memory deficits in terms of decreased spontaneous alternation and absence of acquisition of a daily changing platform position in the water-maze. Acquisition of a fixed platform location or passive avoidance response was preserved. PCAF KO mice showed no difference with WT C57BL/6j controls in their performances in the forced swimming and light/dark exploration box, suggesting no particular phenotype on anxiety and stress responses. We therefore evidenced marked phenotypic differences in PCAF KO mice depending on the genetic background strain confirming that PCAF histone acetyltransferase is involved lifelong in the chromatin remodelling necessary for memory formation but differentially involved in anxiety and response to stress.


Subject(s)
Anxiety/psychology , Histone Acetyltransferases/genetics , Memory, Short-Term , Stress, Psychological/psychology , p300-CBP Transcription Factors/genetics , Animals , Anxiety/genetics , Avoidance Learning , Female , Histone Acetyltransferases/physiology , Male , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Knockout , Reaction Time , Spatial Behavior , Stress, Psychological/genetics , p300-CBP Transcription Factors/physiology
20.
Neuropsychopharmacology ; 33(7): 1584-602, 2008 Jun.
Article in English | MEDLINE | ID: mdl-17805310

ABSTRACT

Chromatin remodeling by posttranslational modification of histones plays an important role in brain plasticity, including memory, response to stress and depression. The importance of H3/4 histones acetylation by CREB-binding protein (CBP) or related histone acetyltransferase, including p300, was specifically demonstrated using knockout (KO) mouse models. The physiological role of a related protein that also acts as a transcriptional coactivator with intrinsic histone acetylase activity, the p300/CBP-associated factor (PCAF), is poorly documented. We analyzed the behavioral phenotype of homozygous male and female PCAF KO mice and report a marked impact of PCAF deletion on memory processes and stress response. PCAF KO animals showed short-term memory deficits at 2 months of age, measured using spontaneous alternation, object recognition, or acquisition of a daily changing platform position in the water maze. Acquisition of a fixed platform location was delayed, but preserved, and no passive avoidance deficit was noted. No gender-related difference was observed. These deficits were associated with hippocampal alterations in pyramidal cell layer organization, basal levels of Fos immunoreactivity, and MAP kinase activation. PCAF KO mice also showed an exaggerated response to acute stress, forced swimming, and conditioned fear, associated with increased plasma corticosterone levels. Moreover, learning and memory impairments worsened at 6 and 12 months of age, when animals failed to acquire the fixed platform location in the water maze and showed passive avoidance deficits. These observations demonstrate that PCAF histone acetylase is involved lifelong in the chromatin remodeling necessary for memory formation and response to stress.


Subject(s)
Memory/physiology , Stress, Psychological/genetics , Stress, Psychological/physiopathology , p300-CBP Transcription Factors/deficiency , Age Factors , Analysis of Variance , Animals , Avoidance Learning/physiology , Behavior, Animal , Conditioning, Psychological/physiology , Corticosterone/blood , Disease Models, Animal , Emotions/physiology , Exploratory Behavior/physiology , Fear , Female , Hippocampus/pathology , Male , Maze Learning/physiology , Memory Disorders/genetics , Memory Disorders/physiopathology , Mice , Mice, Knockout , Pattern Recognition, Visual/physiology , Sex Factors , Stress, Psychological/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...