Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Neurobiol Stress ; 26: 100565, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37664876

ABSTRACT

Repeated, long-term (weeks to months) exposure to standard antidepressant medications is required to achieve treatment efficacy. In contrast, acute ketamine quickly improves mood for an extended time. Recent work implicates that hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are involved in mediating ketamine's antidepressant effects. In this study, we directly targeted HCN channels and achieved ketamine-like rapid and sustained antidepressant efficacy. Our in vitro electrophysiological recordings first showed that HCN inhibitor DK-AH 269 (also called cilobradine) decreased the pathological HCN-mediated current (Ih) and abnormal hyperactivity of ventral tegmental area (VTA) dopamine (DA) neurons in a depressive-like model produced by chronic social defeat stress (CSDS). Our in vivo studies further showed that acute intra-VTA or acute systemic administration of DK-AH 269 normalized social behavior and rescued sucrose preference in CSDS-susceptible mice. The single-dose of DK-AH 269, both by intra-VTA microinfusion and intraperitoneal (ip) approaches, could produce an extended 13-day duration of antidepressant-like efficacy. Animals treated with acute DK-AH 269 spent less time immobile than vehicle-treated mice during forced swim test. A social behavioral reversal lasted up to 13 days following the acute DK-AH 269 ip injection, and this rapid and sustained antidepressant-like response is paralleled with a single-dose treatment of ketamine. This study provides a novel ion channel target for acutely acting, long-lasting antidepressant-like effects.

2.
Nat Commun ; 13(1): 1532, 2022 03 22.
Article in English | MEDLINE | ID: mdl-35318315

ABSTRACT

Anxiety disorders are complex diseases, and often co-occur with depression. It is as yet unclear if a common neural circuit controls anxiety-related behaviors in both anxiety-alone and comorbid conditions. Here, utilizing the chronic social defeat stress (CSDS) paradigm that induces singular or combined anxiety- and depressive-like phenotypes in mice, we show that a ventral tegmental area (VTA) dopamine circuit projecting to the basolateral amygdala (BLA) selectively controls anxiety- but not depression-like behaviors. Using circuit-dissecting ex vivo electrophysiology and in vivo fiber photometry approaches, we establish that expression of anxiety-like, but not depressive-like, phenotypes are negatively correlated with VTA → BLA dopamine neuron activity. Further, our optogenetic studies demonstrate a causal link between such neuronal activity and anxiety-like behaviors. Overall, these data establish a functional role for VTA → BLA dopamine neurons in bi-directionally controlling anxiety-related behaviors not only in anxiety-alone, but also in anxiety-depressive comorbid conditions in mice.


Subject(s)
Basolateral Nuclear Complex , Animals , Anxiety , Anxiety Disorders , Dopaminergic Neurons/metabolism , Mesencephalon , Mice , Stress, Psychological , Ventral Tegmental Area/physiology
3.
Biol Psychiatry ; 85(3): 226-236, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30336931

ABSTRACT

BACKGROUND: Homeostatic plasticity in mesolimbic dopamine (DA) neurons plays an essential role in mediating resilience to social stress. Recent evidence implicates an association between stress resilience and projections from the locus coeruleus (LC) to the ventral tegmental area (VTA) (LC→VTA) DA system. However, the precise circuitry and molecular mechanisms of the homeostatic plasticity in mesolimbic DA neurons mediated by the LC→VTA circuitry, and its role in conferring resilience to social defeat stress, have not been described. METHODS: In a well-established chronic social defeat stress model of depression, using projection-specific electrophysiological recordings and optogenetic, pharmacological, and molecular profiling techniques, we investigated the functional role and molecular basis of an LC→VTA circuit in conferring resilience to social defeat stress. RESULTS: We found that LC neurons projecting to the VTA exhibit enhanced firing activity in resilient, but not susceptible, mice. Optogenetically mimicking this firing adaptation in susceptible mice reverses their depression-related behaviors, and induces reversal of cellular hyperactivity and homeostatic plasticity in VTA DA neurons projecting to the nucleus accumbens. Circuit-specific molecular profiling studies reveal that α1- and ß3-adrenergic receptors are highly expressed in VTA→nucleus accumbens DA neurons. Pharmacologically activating these receptors induces similar proresilient effects at the ion channel and cellular and behavioral levels, whereas antagonizing these receptors blocks the proresilient effect of optogenetic activation of LC→VTA circuit neurons in susceptible mice. CONCLUSIONS: These findings reveal a key role of the LC→VTA circuit in mediating homeostatic plasticity in stress resilience and reveal α1- and ß3-adrenergic receptors as new molecular targets for therapeutically promoting resilience.


Subject(s)
Locus Coeruleus/physiology , Receptors, Adrenergic, alpha-1/physiology , Receptors, Adrenergic, beta-3/physiology , Resilience, Psychological , Ventral Tegmental Area/physiology , Adrenergic alpha-1 Receptor Agonists/pharmacology , Adrenergic alpha-1 Receptor Antagonists/pharmacology , Adrenergic beta-3 Receptor Agonists/pharmacology , Adrenergic beta-3 Receptor Antagonists/pharmacology , Animals , Behavior, Animal/physiology , Dopaminergic Neurons/physiology , Homeostasis/physiology , Locus Coeruleus/drug effects , Male , Mice , Neural Pathways/physiology , Neuronal Plasticity/physiology , Resilience, Psychological/drug effects , Stress, Psychological/physiopathology , Ventral Tegmental Area/drug effects
4.
Nat Commun ; 9(1): 3149, 2018 08 08.
Article in English | MEDLINE | ID: mdl-30089879

ABSTRACT

The role of somatostatin interneurons in nucleus accumbens (NAc), a key brain reward region, remains poorly understood due to the fact that these cells account for < 1% of NAc neurons. Here, we use optogenetics, electrophysiology, and RNA-sequencing to characterize the transcriptome and functioning of NAc somatostatin interneurons after repeated exposure to cocaine. We find that the activity of somatostatin interneurons regulates behavioral responses to cocaine, with repeated cocaine reducing the excitability of these neurons. Repeated cocaine also induces transcriptome-wide changes in gene expression within NAc somatostatin interneurons. We identify the JUND transcription factor as a key regulator of cocaine action and confirmed, by use of viral-mediated gene transfer, that JUND activity in somatostatin interneurons influences behavioral responses to cocaine. Our results identify alterations in NAc induced by cocaine in a sparse population of somatostatin interneurons, and illustrate the value of studying brain diseases using cell type-specific whole transcriptome RNA-sequencing.


Subject(s)
Adaptation, Physiological/drug effects , Cocaine/pharmacology , Interneurons/drug effects , Interneurons/metabolism , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Somatostatin/metabolism , Transcriptome , Animals , Brain/metabolism , Gene Expression Profiling , Gene Expression Regulation/drug effects , Gene Transfer Techniques , Locomotion , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Optogenetics/methods , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Reward , Sequence Analysis, RNA , Somatostatin/pharmacology , Transcription Factors/drug effects
5.
Nat Commun ; 9(1): 653, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29422549

ABSTRACT

The original version of this Article contained an error in the spelling of the author Scott Edwards, which was incorrectly given as Scott Edward. This has now been corrected in both the PDF and HTML versions of the Article.

6.
Neuroscience ; 376: 108-116, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29476894

ABSTRACT

Women are twice as likely to be diagnosed with major depressive disorder. However, fewer studies in rodent models of depression have used female animals, leading to a relative lack of understanding of the female brain's response to stress, especially at a neural circuit level. In this study, we utilized a 6-day subchronic variable stress (SCVS) mouse model and measured novelty suppressed feeding as behavioral criteria to evaluate susceptibility to SCVS in male and female mice. First, we showed that SCVS induced a decrease in latency to eat (susceptible phenotype) in female mice, but not in males (resilient phenotype). After determining behavioral phenotypes, we investigated the firing activities of dopamine (DA) neurons in the ventral tegmental area (VTA), as well as the neurons that project from lateral habenula (LHb) to the VTA and from locus coeruleus (LC) to the VTA. Utilizing retrograding lumafluor fluorescent tracers and electrophysiology techniques, we performed cell type- and circuit-specific measures of neuronal firing rates. Our data show that SCVS significantly increased the firing rate of LHb-VTA circuit neurons in female mice when compared to that of their female controls, an effect that was absent in SCVS-exposed males. Interestingly, SCVS did not induce significant firing alterations in VTA DA neurons and LC-VTA circuit neurons in either female mice or male mice when compared to their stress-naïve controls. Overall, our data show sex differences in the LHb-VTA circuit responses to SCVS, and implicates a potential role of this projection in mediating vulnerability of female mice to stress-induced depression.


Subject(s)
Depressive Disorder/physiopathology , Neuronal Plasticity/physiology , Neurons/physiology , Reward , Sex Characteristics , Stress, Psychological/physiopathology , Action Potentials , Animals , Brain/physiopathology , Disease Susceptibility , Female , Male , Mice, Inbred C57BL , Tissue Culture Techniques
7.
Nat Commun ; 8(1): 2220, 2017 12 20.
Article in English | MEDLINE | ID: mdl-29263389

ABSTRACT

Alcohol-use disorder (AUD) is the most prevalent substance-use disorder worldwide. There is substantial individual variability in alcohol drinking behaviors in the population, the neural circuit mechanisms of which remain elusive. Utilizing in vivo electrophysiological techniques, we find that low alcohol drinking (LAD) mice have dramatically higher ventral tegmental area (VTA) dopamine neuron firing and burst activity. Unexpectedly, VTA dopamine neuron activity in high alcohol drinking (HAD) mice does not differ from alcohol naive mice. Optogenetically enhancing VTA dopamine neuron burst activity in HAD mice decreases alcohol drinking behaviors. Circuit-specific recordings reveal that spontaneous activity of nucleus accumbens-projecting VTA (VTA-NAc) neurons is selectively higher in LAD mice. Specifically activating this projection is sufficient to reduce alcohol consumption in HAD mice. Furthermore, we uncover ionic and cellular mechanisms that suggest unique neuroadaptations between the alcohol drinking groups. Together, these data identify a neural circuit responsible for individual alcohol drinking behaviors.


Subject(s)
Alcohol Drinking/physiopathology , Behavior, Animal/physiology , Dopaminergic Neurons/metabolism , Nucleus Accumbens/physiopathology , Ventral Tegmental Area/physiopathology , Alcohol Drinking/metabolism , Animals , Mesencephalon/metabolism , Mesencephalon/physiopathology , Mice , Neural Pathways/physiology , Nucleus Accumbens/metabolism , Optogenetics , Ventral Tegmental Area/metabolism
8.
Neurotherapeutics ; 14(3): 698-715, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28560710

ABSTRACT

Major depressive disorder (MDD) is a chronic and potentially life threatening illness that carries a staggering global burden. Characterized by depressed mood, MDD is often difficult to diagnose and treat owing to heterogeneity of syndrome and complex etiology. Contemporary antidepressant treatments are based on improved monoamine-based formulations from serendipitous discoveries made > 60 years ago. Novel antidepressant treatments are necessary, as roughly half of patients using available antidepressants do not see long-term remission of depressive symptoms. Current development of treatment options focuses on generating efficacious antidepressants, identifying depression-related neural substrates, and better understanding the pathophysiological mechanisms of depression. Recent insight into the brain's mesocorticolimbic circuitry from animal models of depression underscores the importance of ionic mechanisms in neuronal homeostasis and dysregulation, and substantial evidence highlights a potential role for ion channels in mediating depression-related excitability changes. In particular, hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are essential regulators of neuronal excitability. In this review, we describe seminal research on HCN channels in the prefrontal cortex and hippocampus in stress and depression-related behaviors, and highlight substantial evidence within the ventral tegmental area supporting the development of novel therapeutics targeting HCN channels in MDD. We argue that methods targeting the activity of reward-related brain areas have significant potential as superior treatments for depression.


Subject(s)
Antidepressive Agents/pharmacology , Brain/physiopathology , Depressive Disorder, Major/physiopathology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Animals , Brain/drug effects , Brain/metabolism , Depressive Disorder, Major/metabolism , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/drug effects
9.
Nat Commun ; 8: 15497, 2017 05 22.
Article in English | MEDLINE | ID: mdl-28530238

ABSTRACT

Antidepressants (ADs) are the most common treatment for major depressive disorder (MDD). However, only ∼30% of patients experience adequate response after a single AD trial, and this variability remains poorly understood. Here, we investigated microRNAs (miRNAs) as biomarkers of AD response using small RNA-sequencing in paired samples from MDD patients enrolled in a large, randomized placebo-controlled trial of duloxetine collected before and 8 weeks after treatment. Our results revealed differential expression of miR-146a-5p, miR-146b-5p, miR-425-3p and miR-24-3p according to treatment response. These results were replicated in two independent clinical trials of MDD, a well-characterized animal model of depression, and post-mortem human brains. Furthermore, using a combination of bioinformatics, mRNA studies and functional in vitro experiments, we showed significant dysregulation of genes involved in MAPK/Wnt signalling pathways. Together, our results indicate that these miRNAs are consistent markers of treatment response and regulators of the MAPK/Wnt systems.


Subject(s)
Depressive Disorder, Major/drug therapy , Duloxetine Hydrochloride/therapeutic use , MicroRNAs/genetics , Adult , Aged , Animals , Antidepressive Agents/therapeutic use , Biomarkers , Brain/pathology , Computational Biology , Depressive Disorder, Major/genetics , Female , Gene Expression Regulation , HEK293 Cells , Humans , MAP Kinase Signaling System , Male , Mice , Mice, Inbred C57BL , Middle Aged , Wnt Signaling Pathway , Young Adult
10.
Nat Neurosci ; 19(10): 1321-30, 2016 10.
Article in English | MEDLINE | ID: mdl-27526204

ABSTRACT

Normal brain function depends on the interaction between highly specialized neurons that operate within anatomically and functionally distinct brain regions. Neuronal specification is driven by transcriptional programs that are established during early neuronal development and remain in place in the adult brain. The fidelity of neuronal specification depends on the robustness of the transcriptional program that supports the neuron type-specific gene expression patterns. Here we show that polycomb repressive complex 2 (PRC2), which supports neuron specification during differentiation, contributes to the suppression of a transcriptional program that is detrimental to adult neuron function and survival. We show that PRC2 deficiency in striatal neurons leads to the de-repression of selected, predominantly bivalent PRC2 target genes that are dominated by self-regulating transcription factors normally suppressed in these neurons. The transcriptional changes in PRC2-deficient neurons lead to progressive and fatal neurodegeneration in mice. Our results point to a key role of PRC2 in protecting neurons against degeneration.


Subject(s)
Gene Silencing , Nerve Degeneration/genetics , Polycomb Repressive Complex 2/metabolism , Animals , Cell Death/genetics , Cell Survival/genetics , Down-Regulation , Female , Histone-Lysine N-Methyltransferase/metabolism , Male , Mice , Mice, Knockout , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurons/metabolism , Polycomb Repressive Complex 2/deficiency , Polycomb Repressive Complex 2/genetics
11.
Nat Commun ; 7: 11671, 2016 05 24.
Article in English | MEDLINE | ID: mdl-27216573

ABSTRACT

Less than half of patients suffering from major depressive disorder, a leading cause of disability worldwide, achieve remission with current antidepressants, making it imperative to develop more effective treatment. A new therapeutic direction is emerging from the increased understanding of natural resilience as an active stress-coping process. It is known that potassium (K(+)) channels in the ventral tegmental area (VTA) are an active mediator of resilience. However, no druggable targets have been identified to potentiate active resilience mechanisms. In the chronic social defeat stress model of depression, we report that KCNQ-type K(+) channel openers, including FDA-approved drug retigabine (ezogabine), show antidepressant efficacy. We demonstrate that overexpression of KCNQ channels in the VTA dopaminergic neurons and either local infusion or systemic administration of retigabine normalized neuronal hyperactivity and depressive behaviours. These findings identify KCNQ as a target for conceptually novel antidepressants that function through the potentiation of active resilience mechanisms.


Subject(s)
Depressive Disorder, Major/drug therapy , KCNQ3 Potassium Channel/metabolism , Membrane Transport Modulators/pharmacology , Resilience, Psychological/drug effects , Stress, Psychological/drug therapy , Adaptation, Psychological/drug effects , Adaptation, Psychological/physiology , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Behavior, Animal/drug effects , Behavior, Animal/physiology , Carbamates/pharmacology , Carbamates/therapeutic use , Depressive Disorder, Major/metabolism , Depressive Disorder, Major/physiopathology , Depressive Disorder, Major/psychology , Disease Models, Animal , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Electrophysiological Phenomena , Humans , Male , Membrane Transport Modulators/therapeutic use , Mice , Mice, Inbred C57BL , Phenylenediamines/pharmacology , Phenylenediamines/therapeutic use , Stress, Psychological/metabolism , Stress, Psychological/psychology , Ventral Tegmental Area/cytology , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism , Ventral Tegmental Area/physiology
12.
Science ; 344(6181): 313-9, 2014 Apr 18.
Article in English | MEDLINE | ID: mdl-24744379

ABSTRACT

Typical therapies try to reverse pathogenic mechanisms. Here, we describe treatment effects achieved by enhancing depression-causing mechanisms in ventral tegmental area (VTA) dopamine (DA) neurons. In a social defeat stress model of depression, depressed (susceptible) mice display hyperactivity of VTA DA neurons, caused by an up-regulated hyperpolarization-activated current (I(h)). Mice resilient to social defeat stress, however, exhibit stable normal firing of these neurons. Unexpectedly, resilient mice had an even larger I(h), which was observed in parallel with increased potassium (K(+)) channel currents. Experimentally further enhancing Ih or optogenetically increasing the hyperactivity of VTA DA neurons in susceptible mice completely reversed depression-related behaviors, an antidepressant effect achieved through resilience-like, projection-specific homeostatic plasticity. These results indicate a potential therapeutic path of promoting natural resilience for depression treatment.


Subject(s)
Depression/physiopathology , Dopaminergic Neurons/physiology , Resilience, Psychological , Stress, Psychological/physiopathology , Ventral Tegmental Area/physiology , Animals , Behavior, Animal/drug effects , Electrophysiological Phenomena , Homeostasis , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Lamotrigine , Male , Membrane Potentials/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Optogenetics , Patch-Clamp Techniques , Potassium Channels/metabolism , Social Behavior , Triazines/pharmacology
13.
J Neurosci ; 34(11): 3878-87, 2014 Mar 12.
Article in English | MEDLINE | ID: mdl-24623766

ABSTRACT

Decreased medial prefrontal cortex (mPFC) neuronal activity is associated with social defeat-induced depression- and anxiety-like behaviors in mice. However, the molecular mechanisms underlying the decreased mPFC activity and its prodepressant role remain unknown. We show here that induction of the transcription factor ΔFosB in mPFC, specifically in the prelimbic (PrL) area, mediates susceptibility to stress. ΔFosB induction in PrL occurred selectively in susceptible mice after chronic social defeat stress, and overexpression of ΔFosB in this region, but not in the nearby infralimbic (IL) area, enhanced stress susceptibility. ΔFosB produced these effects partly through induction of the cholecystokinin (CCK)-B receptor: CCKB blockade in mPFC induces a resilient phenotype, whereas CCK administration into mPFC mimics the anxiogenic- and depressant-like effects of social stress. We previously found that optogenetic stimulation of mPFC neurons in susceptible mice reverses several behavioral abnormalities seen after chronic social defeat stress. Therefore, we hypothesized that optogenetic stimulation of cortical projections would rescue the pathological effects of CCK in mPFC. After CCK infusion in mPFC, we optogenetically stimulated mPFC projections to basolateral amygdala or nucleus accumbens, two subcortical structures involved in mood regulation. Stimulation of corticoamygdala projections blocked the anxiogenic effect of CCK, although no effect was observed on other symptoms of social defeat. Conversely, stimulation of corticoaccumbens projections reversed CCK-induced social avoidance and sucrose preference deficits but not anxiogenic-like effects. Together, these results indicate that social stress-induced behavioral deficits are mediated partly by molecular adaptations in mPFC involving ΔFosB and CCK through cortical projections to distinct subcortical targets.


Subject(s)
Anxiety Disorders/physiopathology , Cholecystokinin/physiology , Depressive Disorder/physiopathology , Prefrontal Cortex/physiology , Proto-Oncogene Proteins c-fos/physiology , Receptor, Cholecystokinin B/physiology , Animals , Anti-Anxiety Agents/pharmacology , Anxiety Disorders/pathology , Brain Mapping , Chronic Disease , Depressive Disorder/pathology , Indoles/pharmacology , Limbic System/cytology , Limbic System/drug effects , Limbic System/physiology , Male , Meglumine/analogs & derivatives , Meglumine/pharmacology , Mice , Mice, Inbred C57BL , Neural Pathways/physiology , Prefrontal Cortex/cytology , Prefrontal Cortex/drug effects , Proto-Oncogene Proteins c-fos/genetics , Receptor, Cholecystokinin B/antagonists & inhibitors , Receptor, Cholecystokinin B/genetics , Social Dominance , Stress, Psychological/pathology , Stress, Psychological/physiopathology
14.
Nat Neurosci ; 17(1): 27-9, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24270188

ABSTRACT

Mechanisms controlling release of brain-derived neurotrophic factor (BDNF) in the mesolimbic dopamine reward pathway remain unknown. We report that phasic optogenetic activation of this pathway increases BDNF amounts in the nucleus accumbens (NAc) of socially stressed mice but not of stress-naive mice. This stress gating of BDNF signaling is mediated by corticotrophin-releasing factor (CRF) acting in the NAc. These results unravel a stress context-detecting function of the brain's mesolimbic circuit.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Corticotropin-Releasing Hormone/metabolism , Neurons/metabolism , Nucleus Accumbens/metabolism , Stress, Psychological/metabolism , Ventral Tegmental Area/metabolism , Animals , Azepines/pharmacology , Bacterial Proteins/genetics , Benzamides/pharmacology , Brain-Derived Neurotrophic Factor/genetics , Channelrhodopsins , Corticotropin-Releasing Hormone/pharmacology , Disease Models, Animal , Dopamine/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Interpersonal Relations , Luminescent Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microinjections , Nucleus Accumbens/cytology , Optogenetics , Peptide Fragments/pharmacology , Photic Stimulation , Statistics, Nonparametric , Stress, Psychological/physiopathology , Time Factors , Tyrosine 3-Monooxygenase/genetics , Ventral Tegmental Area/cytology
15.
Nature ; 493(7433): 532-6, 2013 Jan 24.
Article in English | MEDLINE | ID: mdl-23235832

ABSTRACT

Ventral tegmental area (VTA) dopamine neurons in the brain's reward circuit have a crucial role in mediating stress responses, including determining susceptibility versus resilience to social-stress-induced behavioural abnormalities. VTA dopamine neurons show two in vivo patterns of firing: low frequency tonic firing and high frequency phasic firing. Phasic firing of the neurons, which is well known to encode reward signals, is upregulated by repeated social-defeat stress, a highly validated mouse model of depression. Surprisingly, this pathophysiological effect is seen in susceptible mice only, with no apparent change in firing rate in resilient individuals. However, direct evidence--in real time--linking dopamine neuron phasic firing in promoting the susceptible (depression-like) phenotype is lacking. Here we took advantage of the temporal precision and cell-type and projection-pathway specificity of optogenetics to show that enhanced phasic firing of these neurons mediates susceptibility to social-defeat stress in freely behaving mice. We show that optogenetic induction of phasic, but not tonic, firing in VTA dopamine neurons of mice undergoing a subthreshold social-defeat paradigm rapidly induced a susceptible phenotype as measured by social avoidance and decreased sucrose preference. Optogenetic phasic stimulation of these neurons also quickly induced a susceptible phenotype in previously resilient mice that had been subjected to repeated social-defeat stress. Furthermore, we show differences in projection-pathway specificity in promoting stress susceptibility: phasic activation of VTA neurons projecting to the nucleus accumbens (NAc), but not to the medial prefrontal cortex (mPFC), induced susceptibility to social-defeat stress. Conversely, optogenetic inhibition of the VTA-NAc projection induced resilience, whereas inhibition of the VTA-mPFC projection promoted susceptibility. Overall, these studies reveal novel firing-pattern- and neural-circuit-specific mechanisms of depression.


Subject(s)
Depression/physiopathology , Dopaminergic Neurons/metabolism , Mesencephalon/cytology , Social Behavior , Stress, Psychological/physiopathology , Animals , Depression/etiology , Food Preferences , Male , Mice , Neural Pathways , Nucleus Accumbens/physiology , Optogenetics , Phenotype , Prefrontal Cortex/physiology , Stress, Psychological/complications , Sucrose/administration & dosage , Time Factors , Ventral Tegmental Area/physiology
16.
J Neurosci ; 32(22): 7577-84, 2012 May 30.
Article in English | MEDLINE | ID: mdl-22649236

ABSTRACT

The molecular mechanism underlying induction by cocaine of ΔFosB, a transcription factor important for addiction, remains unknown. Here, we demonstrate a necessary role for two transcription factors, cAMP response element binding protein (CREB) and serum response factor (SRF), in mediating this induction within the mouse nucleus accumbens (NAc), a key brain reward region. CREB and SRF are both activated in NAc by cocaine and bind to the fosB gene promoter. Using viral-mediated Cre recombinase expression in the NAc of single- or double-floxed mice, we show that deletion of both transcription factors from this brain region completely blocks cocaine induction of ΔFosB in NAc, whereas deletion of either factor alone has no effect. Furthermore, deletion of both SRF and CREB from NAc renders animals less sensitive to the rewarding effects of moderate doses of cocaine when tested in the conditioned place preference (CPP) procedure and also blocks locomotor sensitization to higher doses of cocaine. Deletion of CREB alone has the opposite effect and enhances both cocaine CPP and locomotor sensitization. In contrast to ΔFosB induction by cocaine, ΔFosB induction in NAc by chronic social stress, which we have shown previously requires activation of SRF, is unaffected by the deletion of CREB alone. These surprising findings demonstrate the involvement of distinct transcriptional mechanisms in mediating ΔFosB induction within this same brain region by cocaine versus stress. Our results also establish a complex mode of regulation of ΔFosB induction in response to cocaine, which requires the concerted activities of both SRF and CREB.


Subject(s)
CREB-Binding Protein/metabolism , Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Nucleus Accumbens/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Serum Response Factor/metabolism , Analysis of Variance , Animals , CREB-Binding Protein/deficiency , Chromatin Immunoprecipitation , Conditioning, Operant/drug effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Motor Activity/genetics , Nucleus Accumbens/metabolism , Phosphorylation/drug effects , Phosphorylation/genetics , Proto-Oncogene Proteins c-fos/genetics , RNA, Messenger/metabolism , Serum Response Factor/deficiency , Stress, Psychological/genetics , Stress, Psychological/metabolism , Stress, Psychological/physiopathology , Transduction, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...