Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
bioRxiv ; 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38659771

ABSTRACT

Major depressive disorder (MDD) is linked to impaired structural and synaptic plasticity in limbic brain regions. Astrocytes, which regulate synapses and are influenced by chronic stress, likely contribute to these changes. We analyzed astrocyte gene profiles in the nucleus accumbens (NAc) of humans with MDD and mice exposed to chronic stress. Htra1 , which encodes an astrocyte-secreted protease targeting the extracellular matrix (ECM), was significantly downregulated in the NAc of males but upregulated in females in both species. Manipulating Htra1 in mouse NAc astrocytes bidirectionally controlled stress susceptibility in a sex-specific manner. Such Htra1 manipulations also altered neuronal signaling and ECM structural integrity in NAc. These findings highlight astroglia and the brain's ECM as key mediators of sex-specific stress vulnerability, offering new approaches for MDD therapies.

2.
bioRxiv ; 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38464110

ABSTRACT

Drug addiction is a multifactorial syndrome in which genetic predispositions and exposure to environmental stressors constitute major risk factors for the early onset, escalation, and relapse of addictive behaviors. While it is well known that stress plays a key role in drug addiction, the genetic factors that make certain individuals particularly sensitive to stress and thereby more vulnerable to becoming addicted are unknown. In an effort to test a complex set of gene x environment interactions-specifically gene x chronic stress -here we leveraged a systems genetics resource: BXD recombinant inbred mice (BXD5, BXD8, BXD14, BXD22, BXD29, and BXD32) and their parental mouse lines, C57BL/6J and DBA/2J. Utilizing the chronic social defeat stress (CSDS) and chronic variable stress (CVS) paradigms, we first showed sexual dimorphism in the behavioral stress response between the mouse strains. Further, we observed an interaction between genetic background and vulnerability to prolonged exposure to non-social stressors. Finally, we found that DBA/2J and C57BL/6J mice pre-exposed to stress displayed differences in morphine sensitivity. Our results support the hypothesis that genetic variation in predisposition to stress responses influences morphine sensitivity and is likely to modulate the development of drug addiction.

3.
bioRxiv ; 2023 May 08.
Article in English | MEDLINE | ID: mdl-37214877

ABSTRACT

Histone post-translational modifications are critical for mediating persistent alterations in gene expression. By combining unbiased proteomics profiling, and genome-wide approaches, we uncovered a role for mono-methylation of lysine 27 at histone H3 (H3K27me1) in the enduring effects of stress. Specifically, mice exposed to early life stress (ELS) or to chronic social defeat stress (CSDS) in adulthood displayed increased enrichment of H3K27me1, and transient decreases in H3K27me2, in the nucleus accumbens (NAc), a key brain-reward region. Stress induction of H3K27me1 was mediated by the VEFS domain of SUZ12, a core subunit of the polycomb repressive complex-2, which is induced by chronic stress and controls H3K27 methylation patterns. Overexpression of the VEFS domain led to social, emotional, and cognitive abnormalities, and altered excitability of NAc D1 mediums spiny neurons. Together, we describe a novel function of H3K27me1 in brain and demonstrate its role as a "chromatin scar" that mediates lifelong stress susceptibility.

4.
Sci Adv ; 8(48): eabn9494, 2022 Dec 02.
Article in English | MEDLINE | ID: mdl-36449610

ABSTRACT

Women suffer from depression at twice the rate of men, but the underlying molecular mechanisms are poorly understood. Here, we identify marked baseline sex differences in the expression of long noncoding RNAs (lncRNAs), a class of regulatory transcripts, in human postmortem brain tissue that are profoundly lost in depression. One such human lncRNA, RP11-298D21.1 (which we termed FEDORA), is enriched in oligodendrocytes and neurons and up-regulated in the prefrontal cortex (PFC) of depressed females only. We found that virally expressing FEDORA selectively either in neurons or in oligodendrocytes of PFC promoted depression-like behavioral abnormalities in female mice only, changes associated with cell type-specific regulation of synaptic properties, myelin thickness, and gene expression. We also found that blood FEDORA levels have diagnostic implications for depressed women and are associated with clinical response to ketamine. These findings demonstrate the important role played by lncRNAs, and FEDORA in particular, in shaping the sex-specific landscape of the brain and contributing to sex differences in depression.

5.
Biol Psychiatry ; 92(11): 895-906, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36182529

ABSTRACT

BACKGROUND: Social experiences influence susceptibility to substance use disorder. The adolescent period is associated with the development of social reward and is exceptionally sensitive to disruptions to reward-associated behaviors by social experiences. Social isolation (SI) during adolescence alters anxiety- and reward-related behaviors in adult males, but little is known about females. The medial amygdala (meA) is a likely candidate for the modulation of social influence on drug reward because it regulates social reward, develops during adolescence, and is sensitive to social stress. However, little is known regarding how the meA responds to drugs of abuse. METHODS: We used adolescent SI coupled with RNA sequencing to better understand the molecular mechanisms underlying meA regulation of social influence on reward. RESULTS: We show that SI in adolescence, a well-established preclinical model for addiction susceptibility, enhances preference for cocaine in male but not in female mice and alters cocaine-induced protein and transcriptional profiles within the adult meA particularly in males. To determine whether transcriptional mechanisms within the meA are important for these behavioral effects, we manipulated Crym expression, a sex-specific key driver gene identified through differential gene expression and coexpression network analyses, specifically in meA neurons. Overexpression of Crym, but not another key driver that did not meet our sex-specific criteria, recapitulated the behavioral and transcriptional effects of adolescent SI. CONCLUSIONS: These results show that the meA is essential for modulating the sex-specific effects of social experience on drug reward and establish Crym as a critical mediator of sex-specific behavioral and transcriptional plasticity.


Subject(s)
Cocaine , Animals , Male , Female , Mice , Cocaine/pharmacology , Cocaine/metabolism , mu-Crystallins , Reward , Neurons/metabolism , Amygdala/metabolism
6.
Mol Psychiatry ; 27(11): 4536-4549, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35902629

ABSTRACT

Major depressive disorder (MDD) is the leading cause of disability worldwide. There is an urgent need for objective biomarkers to diagnose this highly heterogeneous syndrome, assign treatment, and evaluate treatment response and prognosis. MicroRNAs (miRNAs) are short non-coding RNAs, which are detected in body fluids that have emerged as potential biomarkers of many disease conditions. The present study explored the potential use of miRNAs as biomarkers for MDD and its treatment. We profiled the expression levels of circulating blood miRNAs from mice that were collected before and after exposure to chronic social defeat stress (CSDS), an extensively validated mouse model used to study depression, as well as after either repeated imipramine or single-dose ketamine treatment. We observed robust differences in blood miRNA signatures between stress-resilient and stress-susceptible mice after an incubation period, but not immediately after exposure to the stress. Furthermore, ketamine treatment was more effective than imipramine at re-establishing baseline miRNA expression levels, but only in mice that responded behaviorally to the drug. We identified the red blood cell-specific miR-144-3p as a candidate biomarker to aid depression diagnosis and predict ketamine treatment response in stress-susceptible mice and MDD patients. Lastly, we demonstrate that systemic knockdown of miR-144-3p, via subcutaneous administration of a specific antagomir, is sufficient to reduce the depression-related phenotype in stress-susceptible mice. RNA-sequencing analysis of blood after such miR-144-3p knockdown revealed a blunted transcriptional stress signature as well. These findings identify miR-144-3p as a novel target for diagnosis of MDD as well as for antidepressant treatment, and enhance our understanding of epigenetic processes associated with depression.


Subject(s)
Depressive Disorder, Major , Ketamine , MicroRNAs , Mice , Animals , Depressive Disorder, Major/diagnosis , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/genetics , MicroRNAs/metabolism , Biomarkers , Epigenesis, Genetic , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Ketamine/pharmacology , Ketamine/therapeutic use
7.
Biol Psychiatry ; 91(1): 81-91, 2022 01 01.
Article in English | MEDLINE | ID: mdl-33896623

ABSTRACT

BACKGROUND: Major depressive disorder is a pervasive and debilitating syndrome characterized by mood disturbances, anhedonia, and alterations in cognition. While the prevalence of major depressive disorder is twice as high for women as men, little is known about the molecular mechanisms that drive sex differences in depression susceptibility. METHODS: We discovered that SLIT1, a secreted protein essential for axonal navigation and molecular guidance during development, is downregulated in the adult ventromedial prefrontal cortex (vmPFC) of women with depression compared with healthy control subjects, but not in men with depression. This sex-specific downregulation of Slit1 was also observed in the vmPFC of mice exposed to chronic variable stress. To identify a causal, sex-specific role for SLIT1 in depression-related behavioral abnormalities, we performed knockdown (KD) of Slit1 expression in the vmPFC of male and female mice. RESULTS: When combined with stress exposure, vmPFC Slit1 KD reflected the human condition by inducing a sex-specific increase in anxiety- and depression-related behaviors. Furthermore, we found that vmPFC Slit1 KD decreased the dendritic arborization of vmPFC pyramidal neurons and decreased the excitability of the neurons in female mice, effects not observed in males. RNA sequencing analysis of the vmPFC after Slit1 KD in female mice revealed an augmented transcriptional stress signature. CONCLUSIONS: Together, our findings establish a crucial role for SLIT1 in regulating neurophysiological and transcriptional responses to stress within the female vmPFC and provide mechanistic insight into novel signaling pathways and molecular factors influencing sex differences in depression susceptibility.


Subject(s)
Depressive Disorder, Major , Anhedonia , Animals , Anxiety , Female , Male , Mice , Prefrontal Cortex , Sex Characteristics
8.
J Neurosci ; 2021 Jun 07.
Article in English | MEDLINE | ID: mdl-34099514

ABSTRACT

Paternal stress can induce long-lasting changes in germ cells potentially propagating heritable changes across generations. To date, no studies have investigated differences in transmission patterns between stress-resilient and -susceptible mice. We tested the hypothesis that transcriptional alterations in sperm during chronic social defeat stress (CSDS) transmit increased susceptibility to stress phenotypes to the next generation. We demonstrate differences in offspring from stressed fathers that depend upon paternal category (resilient vs susceptible) and offspring sex. Importantly, artificial insemination reveals that sperm mediates some of the behavioral phenotypes seen in offspring. Using RNA-sequencing we report substantial and distinct changes in the transcriptomic profiles of sperm following CSDS in susceptible vs resilient fathers, with alterations in long noncoding RNAs (lncRNAs) predominating especially in susceptibility. Correlation analysis revealed that these alterations were accompanied by a loss of regulation of protein-coding genes by lncRNAs in sperm of susceptible males. We also identify several co-expression gene modules that are enriched in differentially expressed genes in sperm from either resilient or susceptible fathers. Taken together, these studies advance our understanding of intergenerational epigenetic transmission of behavioral experience.SIGNIFICANCE STATEMENTThis manuscript contributes to the complex factors that influence the paternal transmission of stress phenotypes. By leveraging the segregation of males exposed to chronic social defeat stress into either resilient or susceptible categories we were able to identify the phenotypic differences in the paternal transmission of stress phenotypes across generations between the two lineages. Importantly, this work also alludes to the significance of both long noncoding RNAs and protein coding genes mediating the paternal transmission of stress. The knowledge gained from these data is of particular interest in understanding the risk for the development of psychiatric disorders such as anxiety and depression.

10.
Nat Neurosci ; 24(5): 667-676, 2021 05.
Article in English | MEDLINE | ID: mdl-33723435

ABSTRACT

Animals susceptible to chronic social defeat stress (CSDS) exhibit depression-related behaviors, with aberrant transcription across several limbic brain regions, most notably in the nucleus accumbens (NAc). Early life stress (ELS) promotes susceptibility to CSDS in adulthood, but associated enduring changes in transcriptional control mechanisms in the NAc have not yet been investigated. In this study, we examined long-lasting changes to histone modifications in the NAc of male and female mice exposed to ELS. Dimethylation of lysine 79 of histone H3 (H3K79me2) and the enzymes (DOT1L and KDM2B) that control this modification are enriched in D2-type medium spiny neurons and are shown to be crucial for the expression of ELS-induced stress susceptibility. We mapped the site-specific regulation of this histone mark genome wide to reveal the transcriptional networks it modulates. Finally, systemic delivery of a small molecule inhibitor of DOT1L reversed ELS-induced behavioral deficits, indicating the clinical relevance of this epigenetic mechanism.


Subject(s)
Histone Demethylases/metabolism , Neurons/metabolism , Nucleus Accumbens/metabolism , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Stress, Psychological/metabolism , Animals , F-Box Proteins/metabolism , Gene Expression Regulation , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Male , Mice
11.
Neuron ; 106(6): 912-926.e5, 2020 06 17.
Article in English | MEDLINE | ID: mdl-32304628

ABSTRACT

Depression is a common disorder that affects women at twice the rate of men. Here, we report that long non-coding RNAs (lncRNAs), a recently discovered class of regulatory transcripts, represent about one-third of the differentially expressed genes in the brains of depressed humans and display complex region- and sex-specific patterns of regulation. We identified the primate-specific, neuronal-enriched gene LINC00473 as downregulated in prefrontal cortex (PFC) of depressed females but not males. Using viral-mediated gene transfer to express LINC00473 in adult mouse PFC neurons, we mirrored the human sex-specific phenotype by inducing stress resilience solely in female mice. This sex-specific phenotype was accompanied by changes in synaptic function and gene expression selectively in female mice and, along with studies of human neuron-like cells in culture, implicates LINC00473 as a CREB effector. Together, our studies identify LINC00473 as a female-specific driver of stress resilience that is aberrant in female depression.


Subject(s)
Depressive Disorder, Major/genetics , Prefrontal Cortex/metabolism , RNA, Long Noncoding/genetics , Resilience, Psychological , Stress, Psychological/genetics , Adult , Aged , Aged, 80 and over , Animals , Behavior, Animal , Depression/genetics , Depression/metabolism , Depressive Disorder, Major/metabolism , Down-Regulation , Female , Humans , Male , Mice , Mice, Transgenic , Middle Aged , Neurons/metabolism , RNA, Long Noncoding/metabolism , RNA-Seq , Sex Factors , Stress, Psychological/metabolism , Young Adult
12.
Nat Neurosci ; 22(9): 1413-1423, 2019 09.
Article in English | MEDLINE | ID: mdl-31427770

ABSTRACT

Understanding the transcriptional changes that are engaged in stress resilience may reveal novel antidepressant targets. Here we use gene co-expression analysis of RNA-sequencing data from brains of resilient mice to identify a gene network that is unique to resilience. Zfp189, which encodes a previously unstudied zinc finger protein, is the highest-ranked key driver gene in the network, and overexpression of Zfp189 in prefrontal cortical neurons preferentially activates this network and promotes behavioral resilience. The transcription factor CREB is a predicted upstream regulator of this network and binds to the Zfp189 promoter. To probe CREB-Zfp189 interactions, we employ CRISPR-mediated locus-specific transcriptional reprogramming to direct CREB or G9a (a repressive histone methyltransferase) to the Zfp189 promoter in prefrontal cortex neurons. Induction of Zfp189 with site-specific CREB is pro-resilient, whereas suppressing Zfp189 expression with G9a increases susceptibility. These findings reveal an essential role for Zfp189 and CREB-Zfp189 interactions in mediating a central transcriptional network of resilience.


Subject(s)
Adaptation, Psychological/physiology , Stress, Psychological/genetics , Zinc Fingers/genetics , Animals , Gene Regulatory Networks/genetics , Mice , Mice, Inbred C57BL , Prefrontal Cortex/metabolism , Transcription, Genetic
13.
Sci Rep ; 9(1): 4615, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30874581

ABSTRACT

Animal studies using chronic social defeat stress (CSDS) in mice showed that brain-derived neurotrophic factor (BDNF) signaling in the mesolimbic dopamine (DA) circuit is important for the development of social aversion. However, the downstream molecular targets after BDNF release from ventral tegmental area (VTA) DA terminals are unknown. Here, we show that depressive-like behaviors induced by CSDS are mediated in part by Gadd45b downstream of BDNF signaling in the nucleus accumbens (NAc). We show that Gadd45b mRNA levels are increased in susceptible but not resilient mice. Intra-NAc infusion of BDNF or optical stimulation of VTA DA terminals in NAc enhanced Gadd45b expression levels in the NAc. Importantly, Gadd45b downregulation reversed social avoidance in susceptible mice. Together, these data suggest that Gadd45b in NAc contributes to susceptibility to social stress. In addition, we investigated the function of Gadd45b in demethylating CpG islands of representative gene targets, which have been associated with a depressive phenotype in humans and animal models. We found that Gadd45b downregulation changes DNA methylation levels in a phenotype-, gene-, and locus-specific fashion. Together, these results highlight the contribution of Gadd45b and changes in DNA methylation in mediating the effects of social stress in the mesolimbic DA circuit.


Subject(s)
Antigens, Differentiation/metabolism , DNA Demethylation/drug effects , Depression/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , DNA/metabolism , Dopamine/metabolism , Male , Mice , Mice, Inbred C57BL , Neural Pathways/physiology , Nucleus Accumbens/metabolism , Social Behavior , Stress, Psychological/physiopathology , Ventral Tegmental Area/metabolism
14.
Dialogues Clin Neurosci ; 21(4): 341-357, 2019 12.
Article in English | MEDLINE | ID: mdl-31949402

ABSTRACT

Depression is a devastating psychiatric disorder caused by a combination of genetic predisposition and life events, mainly exposure to stress. Early life stress (ELS) in particular is known to "scar" the brain, leading to an increased susceptibility to developing depression later in life via epigenetic mechanisms. Epigenetic processes lead to changes in gene expression that are not due to changes in DNA sequence, but achieved via modulation of chromatin modifications, DNA methylation, and noncoding RNAs. Here we review common epigenetic mechanisms including the enzymes that take part in reading, writing, and erasing specific epigenetic marks. We then describe recent developments in understanding how ELS leads to changes in the epigenome that are manifested in increased susceptibility to depression-like abnormalities in animal models. We conclude with highlighting the need for future studies that will potentially enable the utilisation of the understanding of epigenetic changes linked to ELS for the development of much-needed novel therapeutic strategies and biomarker discovery.
.


La depresión es un trastorno psiquiátrico devastador causado por una combinación de una predisposición genética y de acontecimientos vitales, en que destaca la exposición al estrés. Se sabe que el estrés en los inicios de la vida (EIV) deja una "cicatriz" en el cerebro, lo que lleva a un aumento de la susceptibilidad para desarrollar una depresión en años posteriores a través de mecanismos epigenéticos. Los procesos epigenéticos conducen a cambios en la expresión génica que no se deben a cambios en la secuencia de ADN, sino que ocurren mediante la modulación de las modificaciones de la cromatina, de la metilación del ADN y de los ARNs no codificantes. En este artículo se revisan los mecanismos epigenéticos comunes, incluyendo las enzimas que participan en la lectura, la escritura y el borrado de marcas epigenéticas específicas. Luego se describen los desarrollos recientes en la comprensión de cómo el EIV produce cambios en el epigenoma, lo que se manifiesta -en modelos animales- en una mayor susceptibilidad a anormalidades similares a la depresión. Se finaliza destacando la necesidad de futuros estudios que potencialmente permitan utilizar la comprensión de los cambios epigenéticos relacionados con el EIV para el desarrollo de muy necesarias nuevas estrategias terapéuticas y el descubrimiento de biomarcadores.


La dépression, trouble psychiatrique destructeur, est provoquée par une association de prédisposition génétique et d'événements de vie, principalement l'exposition au stress. Le stress de début de vie est particulièrement connu pour laisser une « cicatrice ¼ au cerveau, le rendant plus susceptible de développer une dépression ultérieure par le biais de mécanismes épigénétiques. Les processus épigénétiques entraînent des modifications de l'expression génique qui ne sont pas dues à des changements de séquence ADN mais qui apparaissent par modulation des modifications de la chromatine, par méthylation de l'ADN et par des ARN non codants. Nous analysons dans cet article les mécanismes épigénétiques courants, dont les enzymes impliquées dans la lecture, l'écriture et l'effacement de marques épigénétiques spécifiques. Ainsi que nous le décrivons ensuite, des avancées récentes nous permettent de comprendre comment le stress de début de vie modifie l'épigénome en augmentant la susceptibilité aux anomalies de la dépression dans les modèles animaux. Nous en concluons que de nouvelles études sont nécessaires qui pourraient utiliser cette compréhension des modifications épigénétiques liées au stress de début de vie pour développer d'indispensables nouvelles stratégies thérapeutiques et la découverte de biomarqueurs.


Subject(s)
Depression/genetics , Depressive Disorder/genetics , Genetic Predisposition to Disease/genetics , Stress, Psychological/genetics , Animals , DNA Methylation/genetics , Depression/drug therapy , Depressive Disorder/drug therapy , Epigenomics/methods , Humans
16.
Nat Commun ; 9(1): 1116, 2018 03 16.
Article in English | MEDLINE | ID: mdl-29549264

ABSTRACT

Most people exposed to stress do not develop depression. Animal models have shown that stress resilience is an active state that requires broad transcriptional adaptations, but how this homeostatic process is regulated remains poorly understood. In this study, we analyze upstream regulators of genes differentially expressed after chronic social defeat stress. We identify estrogen receptor α (ERα) as the top regulator of pro-resilient transcriptional changes in the nucleus accumbens (NAc), a key brain reward region implicated in depression. In accordance with these findings, nuclear ERα protein levels are altered by stress in male and female mice. Further, overexpression of ERα in the NAc promotes stress resilience in both sexes. Subsequent RNA-sequencing reveals that ERα overexpression in NAc reproduces the transcriptional signature of resilience in male, but not female, mice. These results indicate that NAc ERα is an important regulator of pro-resilient transcriptional changes, but with sex-specific downstream targets.


Subject(s)
Adaptation, Psychological/physiology , Behavior, Animal/physiology , Depression/physiopathology , Estrogen Receptor alpha/metabolism , Nucleus Accumbens/metabolism , Stress, Psychological/physiopathology , Animals , Estrogen Receptor alpha/genetics , Female , Gene Expression Profiling , Male , Mice , Mice, Inbred C57BL , Models, Animal , Sex Factors , Transcriptome/genetics
17.
Nat Med ; 23(9): 1102-1111, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28825715

ABSTRACT

Major depressive disorder (MDD) is a leading cause of disease burden worldwide. While the incidence, symptoms and treatment of MDD all point toward major sex differences, the molecular mechanisms underlying this sexual dimorphism remain largely unknown. Here, combining differential expression and gene coexpression network analyses, we provide a comprehensive characterization of male and female transcriptional profiles associated with MDD across six brain regions. We overlap our human profiles with those from a mouse model, chronic variable stress, and capitalize on converging pathways to define molecular and physiological mechanisms underlying the expression of stress susceptibility in males and females. Our results show a major rearrangement of transcriptional patterns in MDD, with limited overlap between males and females, an effect seen in both depressed humans and stressed mice. We identify key regulators of sex-specific gene networks underlying MDD and confirm their sex-specific impact as mediators of stress susceptibility. For example, downregulation of the female-specific hub gene Dusp6 in mouse prefrontal cortex mimicked stress susceptibility in females, but not males, by increasing ERK signaling and pyramidal neuron excitability. Such Dusp6 downregulation also recapitulated the transcriptional remodeling that occurs in prefrontal cortex of depressed females. Together our findings reveal marked sexual dimorphism at the transcriptional level in MDD and highlight the importance of studying sex-specific treatments for this disorder.


Subject(s)
Brain/metabolism , Depressive Disorder, Major/genetics , Stress, Psychological/genetics , Transcriptome , Adult , Aged , Animals , Blotting, Western , Case-Control Studies , Cerebral Cortex/metabolism , Disease Models, Animal , Down-Regulation , Female , Hippocampus/metabolism , Humans , Immunohistochemistry , Male , Mice , Middle Aged , Nucleus Accumbens/metabolism , Patch-Clamp Techniques , Prefrontal Cortex/metabolism , Pyramidal Cells/metabolism , Sequence Analysis, RNA , Sex Characteristics , Sex Factors
18.
Science ; 356(6343): 1185-1188, 2017 06 16.
Article in English | MEDLINE | ID: mdl-28619944

ABSTRACT

Early life stress increases risk for depression. Here we establish a "two-hit" stress model in mice wherein stress at a specific postnatal period increases susceptibility to adult social defeat stress and causes long-lasting transcriptional alterations that prime the ventral tegmental area (VTA)-a brain reward region-to be in a depression-like state. We identify a role for the developmental transcription factor orthodenticle homeobox 2 (Otx2) as an upstream mediator of these enduring effects. Transient juvenile-but not adult-knockdown of Otx2 in VTA mimics early life stress by increasing stress susceptibility, whereas its overexpression reverses the effects of early life stress. This work establishes a mechanism by which early life stress encodes lifelong susceptibility to stress via long-lasting transcriptional programming in VTA mediated by Otx2.


Subject(s)
Depression/genetics , Gene Expression Regulation , Otx Transcription Factors/genetics , Stress, Physiological/genetics , Ventral Tegmental Area/physiopathology , Age Factors , Animals , Depression/physiopathology , Female , Gene Knockdown Techniques , Male , Mice , Mice, Inbred C57BL , Protein Binding
19.
Biol Psychiatry ; 82(11): 794-805, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-28577753

ABSTRACT

BACKGROUND: Exposure to drugs of abuse alters the epigenetic landscape of the brain's reward regions, such as the nucleus accumbens. We investigated how combinations of chromatin modifications affect genes that regulate responses to cocaine. We focused on Auts2, a gene linked to human evolution and cognitive disorders, which displays strong clustering of cocaine-induced chromatin modifications in this brain region. METHODS: We combined chromosome conformation capture, circularized chromosome conformation capture, and related approaches with behavioral paradigms relevant to cocaine phenotypes. Cell type-specific functions were assessed by fluorescence-activated cell sorting and viral-mediated overexpression in Cre-dependent mouse lines. RESULTS: We observed that Auts2 gene expression is increased by repeated cocaine administration specifically in D2-type medium spiny neurons in the nucleus accumbens, an effect seen in male but not female mice. Auts2 messenger RNA expression was also upregulated postmortem in the nucleus accumbens of male human cocaine addicts. We obtained evidence that chromosomal looping, bypassing 1524 kb of linear genome, connects Auts2 to the Caln1 gene locus under baseline conditions. This looping was disrupted after repeated cocaine exposure, resulting in increased expression of both genes in D2-type medium spiny neurons. Cocaine exposure reduces binding of CCCTC-binding factor, a chromosomal scaffolding protein, and increases histone and DNA methylation at the Auts-Caln1 loop base in the nucleus accumbens. Cell type-specific overexpression of Auts2 or Caln1 in D2-type medium spiny neurons demonstrated that both genes promote cocaine reward. CONCLUSIONS: These findings suggest that cocaine-induced alterations of neuronal three-dimensional genome organization destabilize higher order chromatin at specific loci that regulate responses to the drug.


Subject(s)
Chromatin/drug effects , Cocaine/administration & dosage , Dopamine Uptake Inhibitors/administration & dosage , Gene Expression Regulation/drug effects , Nuclear Proteins/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Cohort Studies , Conditioning, Operant/drug effects , Cytoskeletal Proteins , DNA Methylation/drug effects , Gene Expression Regulation/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Models, Molecular , Molecular Conformation , Neuroblastoma/pathology , Nuclear Proteins/genetics , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D1/genetics , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/metabolism , Transcription Factors , Young Adult
20.
Neuropsychopharmacology ; 42(8): 1657-1669, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28074830

ABSTRACT

Depression is a leading cause of disease burden, yet current therapies fully treat <50% of affected individuals. Increasing evidence implicates epigenetic mechanisms in depression and antidepressant action. Here we examined a possible role for the DNA dioxygenase, ten-eleven translocation protein 1 (TET1), in depression-related behavioral abnormalities. We applied chronic social defeat stress, an ethologically validated mouse model of depression-like behaviors, and examined Tet1 expression changes in nucleus accumbens (NAc), a key brain reward region. We show decreased Tet1 expression in NAc in stress-susceptible mice only. Surprisingly, selective knockout of Tet1 in NAc neurons of adult mice produced antidepressant-like effects in several behavioral assays. To identify Tet1 targets that mediate these actions, we performed RNAseq on NAc after conditional deletion of Tet1 and found that immune-related genes are the most highly dysregulated. Moreover, many of these genes are also upregulated in the NAc of resilient mice after chronic social defeat stress. These findings reveal a novel role for TET1, an enzyme important for DNA hydroxymethylation, in the brain's reward circuitry in modulating stress responses in mice. We also identify a subset of genes that are regulated by TET1 in this circuitry. These findings provide new insight into the pathophysiology of depression, which can aid in future antidepressant drug discovery efforts.


Subject(s)
Anxiety/physiopathology , DNA-Binding Proteins/physiology , Depression/physiopathology , Nucleus Accumbens/metabolism , Proto-Oncogene Proteins/physiology , Stress, Psychological/physiopathology , Animals , Anxiety/genetics , Behavior, Animal , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Depression/genetics , Disease Models, Animal , Gene Expression/genetics , Male , Mice , Mice, Knockout , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...