Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 23
2.
Cell Rep ; 42(12): 113488, 2023 12 26.
Article En | MEDLINE | ID: mdl-37995189

Response to threatening environmental stimuli requires detection and encoding of important environmental features that dictate threat. Aversive events are highly salient, which promotes associative learning about stimuli that signal this threat. The nucleus accumbens is uniquely positioned to process this salient, aversive information and promote motivated output, through plasticity on the major projection neurons in the brain area. We describe a nucleus accumbens core local circuit whereby excitatory plasticity facilitates learning and recall of discrete aversive cues. We demonstrate that putative nucleus accumbens substance P release and long-term excitatory plasticity on dopamine 2 receptor-expressing projection neurons are required for cue-dependent fear learning. Additionally, we find that fear learning and recall is dependent on distinct projection neuron subtypes. Our work demonstrates a critical role for nucleus accumbens substance P in cue-dependent aversive learning.


Cues , Nucleus Accumbens , Nucleus Accumbens/physiology , Avoidance Learning , Substance P , Receptors, Dopamine
3.
Front Mol Neurosci ; 16: 1188184, 2023.
Article En | MEDLINE | ID: mdl-37441675

Circadian rhythms are self-sustained oscillations of biological systems that allow an organism to anticipate periodic changes in the environment and optimally align feeding, sleep, wakefulness, and the physiological and biochemical processes that support them within the 24 h cycle. These rhythms are generated at a cellular level by a set of genes, known as clock genes, which code for proteins that inhibit their own transcription in a negative feedback loop and can be perturbed by stress, a risk factor for the development of mood and anxiety disorders. A role for circadian clocks in mood and anxiety has been suggested for decades on the basis of clinical observations, and the dysregulation of circadian rhythms is a prominent clinical feature of stress-related disorders. Despite our understanding of central clock structure and function, the effect of circadian dysregulation in different neuronal subtypes in the suprachiasmatic nucleus (SCN), the master pacemaker region, as well as other brain systems regulating mood, including mesolimbic and limbic circuits, is just beginning to be elucidated. In the brain, circadian clocks regulate neuronal physiological functions, including neuronal activity, synaptic plasticity, protein expression, and neurotransmitter release which in turn affect mood-related behaviors via cell-type specific mechanisms. Both animal and human studies have revealed an association between circadian misalignment and mood disorders and suggest that internal temporal desynchrony might be part of the etiology of psychiatric disorders. To date, little work has been conducted associating mood-related phenotypes to cell-specific effects of the circadian clock disruptions. In this review, we discuss existing literature on how clock-driven changes in specific neuronal cell types might disrupt phase relationships among cellular communication, leading to neuronal circuit dysfunction and changes in mood-related behavior. In addition, we examine cell-type specific circuitry underlying mood dysfunction and discuss how this circuitry could affect circadian clock. We provide a focus for future research in this area and a perspective on chronotherapies for mood and anxiety disorders.

5.
bioRxiv ; 2023 Oct 09.
Article En | MEDLINE | ID: mdl-36798245

Response to threatening environmental stimuli requires detection and encoding of important environmental features that dictate threat. Aversive events are highly salient which promotes associative learning about stimuli that signal this threat. The nucleus accumbens is uniquely positioned to process this salient, aversive information and promote motivated output, through plasticity on the major projection neurons in the brain area. We uncovered a nucleus accumbens core local circuit whereby excitatory plasticity facilitates learning and recall of discrete aversive cues. We demonstrate that putative nucleus accumbens substance P release and long-term excitatory plasticity on dopamine 2 receptor expressing projection neurons is required for cue-dependent fear learning. Additionally, we found fear learning and recall were dependent on distinct projection-neuron subtypes. Our work demonstrates a critical role for Nucleus Accumbens substance P in cue-dependent aversive learning.

6.
SLAS Discov ; 27(8): 448-459, 2022 12.
Article En | MEDLINE | ID: mdl-36210051

BACKGROUND: Stress responses are believed to involve corticotropin releasing factor (CRF), its two cognate receptors (CRF1 and CRF2), and the CRF-binding protein (CRFBP). Whereas decades of research has focused on CRF1, the role of CRF2 in the central nervous system (CNS) has not been thoroughly investigated. We have previously reported that CRF2, interacting with a C terminal fragment of CRFBP, CRFBP(10kD), may have a role in the modulation of neuronal activity. However, the mechanism by which CRF interacts with CRFBP(10kD) and CRF2 has not been fully elucidated due to the lack of useful chemical tools to probe CRFBP. METHODS: We miniaturized a cell-based assay, where CRFBP(10kD) is fused as a chimera with CRF2, and performed a high-throughput screen (HTS) of 350,000 small molecules to find negative allosteric modulators (NAMs) of the CRFBP(10kD)-CRF2 complex. Hits were confirmed by evaluating activity toward parental HEK293 cells, toward CRF2 in the absence of CRFBP(10kD), and toward CRF1 in vitro. Hits were further characterized in ex vivo electrophysiology assays that target: 1) the CRF1+ neurons in the central nucleus of the amygdala (CeA) of CRF1:GFP mice that express GFP under the CRF1 promoter, and 2) the CRF-induced potentiation of N-methyl-D-aspartic acid receptor (NMDAR)-mediated synaptic transmission in dopamine neurons in the ventral tegmental area (VTA). RESULTS: We found that CRFBP(10kD) potentiates CRF-intracellular Ca2+ release specifically via CRF2, indicating that CRFBP may possess excitatory roles in addition to the inhibitory role established by the N-terminal fragment of CRFBP, CRFBP(27kD). We identified novel small molecule CRFBP-CRF2 NAMs that do not alter the CRF1-mediated effects of exogenous CRF but blunt CRF-induced potentiation of NMDAR-mediated synaptic transmission in dopamine neurons in the VTA, an effect mediated by CRF2 and CRFBP. CONCLUSION: These results provide the first evidence of specific roles for CRF2 and CRFBP(10kD) in the modulation of neuronal activity and suggest that CRFBP(10kD)-CRF2 NAMs can be further developed for the treatment of stress-related disorders including alcohol and substance use disorders.


Corticotropin-Releasing Hormone , Research Design , Humans , Animals , Mice , HEK293 Cells
7.
Mol Psychiatry ; 26(6): 1846-1859, 2021 06.
Article En | MEDLINE | ID: mdl-32366954

Motor stereotypies occurring in early-onset neuropsychiatric diseases are associated with dysregulated basal ganglia direct-pathway activity. Disruptions in network connectivity through impaired neuronal structure have been implicated in both rodents and humans. However, the neurobiological mechanisms leading to direct-pathway neuron disconnectivity in stereotypy remain poorly understood. We have a mouse line with Tropomyosin receptor kinase B (TrkB) receptor deletion from D1-expressing cells (D1-Cre-flTrkB) in which a subset of animals shows repetitive rotations and head tics with juvenile onset. Here we demonstrate these behaviors may be associated with abnormal direct-pathway activity by reducing rotations using chemogenetic inhibition of dorsal striatum D1-medium spiny neurons (D1-MSNs) in both juvenile and young-adult mice. Taking advantage of phenotypical differences in animals with similar genotypes, we then interrogated the D1-MSN specific translatome associated with repetitive behavior by using RNA sequencing of ribosome-associated mRNA. Detailed translatome analysis followed by multiplexed gene expression assessment revealed profound alterations in neuronal projection and synaptic structure related genes in stereotypy mice. Examination of neuronal morphology demonstrated dendritic atrophy and dendritic spine loss in dorsal striatum D1-MSNs from mice with repetitive behavior. Together, our results uncover phenotype-specific molecular alterations in D1-MSNs that relate to morphological adaptations in mice displaying stereotypy behavior.


Receptors, Dopamine D1 , Receptors, Dopamine D2 , Animals , Corpus Striatum/metabolism , Individuality , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism
8.
Mol Psychiatry ; 25(5): 1022-1034, 2020 05.
Article En | MEDLINE | ID: mdl-30120419

Depression alters the structure and function of brain reward circuitry. Preclinical evidence suggests that medium spiny neurons (MSNs) in the nucleus accumbens (NAc) undergo structural plasticity; however, the molecular mechanism and behavioral significance is poorly understood. Here we report that atrophy of D1, but not D2 receptor containing MSNs is strongly associated with social avoidance in mice subject to social defeat stress. D1-MSN atrophy is caused by cell-type specific upregulation of the GTPase RhoA and its effector Rho-kinase. Pharmacologic and genetic reduction of activated RhoA prevents depressive outcomes to stress by preventing loss of D1-MSN dendritic arbor. Pharmacologic and genetic promotion of activated RhoA enhances depressive outcomes by reducing D1-MSN dendritic arbor and is sufficient to promote depressive-like behaviors in the absence of stress. Chronic treatment with Rho-kinase inhibitor Y-27632 after chronic social defeat stress reverses depression-like behaviors by restoring D1-MSN dendritic complexity. Taken together, our data indicate functional roles for RhoA and Rho-kinase in mediating depression-like behaviors via dendritic remodeling of NAc D1-MSNs and may prove a useful target for new depression therapeutics.


Dendrites/enzymology , Dendrites/pathology , Depression/pathology , Depression/psychology , Neuronal Plasticity , Receptors, Dopamine D1/metabolism , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Depression/enzymology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nucleus Accumbens/metabolism , Nucleus Accumbens/pathology , Receptors, Dopamine D2/metabolism
9.
Neuron ; 103(3): 432-444.e3, 2019 08 07.
Article En | MEDLINE | ID: mdl-31221559

Subtypes of nucleus accumbens medium spiny neurons (MSNs) promote dichotomous outcomes in motivated behaviors. However, recent reports indicate enhancing activity of either nucleus accumbens (NAc) core MSN subtype augments reward, suggesting coincident MSN activity may underlie this outcome. Here, we report a collateral excitation mechanism in which high-frequency, NAc core dopamine 1 (D1)-MSN activation causes long-lasting potentiation of excitatory transmission (LLP) on dopamine receptor 2 (D2)-MSNs. Our mechanistic investigation demonstrates that this form of plasticity requires release of the excitatory peptide substance P from D1-MSNs and robust cholinergic interneuron activation through neurokinin receptor stimulation. We also reveal that D2-MSN LLP requires muscarinic 1 receptor activation, intracellular calcium signaling, and GluR2-lacking AMPAR insertion. This study uncovers a mechanism for shaping NAc core activity through the transfer of excitatory information from D1-MSNs to D2-MSNs and may provide a means for altering goal-directed behavior through coordinated MSN activity.


Dopaminergic Neurons/physiology , Long-Term Potentiation/physiology , Nucleus Accumbens/physiology , Substance P/metabolism , Action Potentials/physiology , Animals , Aprepitant/pharmacology , Calcium Signaling/physiology , Cholinergic Neurons/physiology , Dopaminergic Neurons/radiation effects , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Interneurons/physiology , Mice , Mice, Inbred C57BL , Motivation , Neurokinin-1 Receptor Antagonists/pharmacology , Nucleus Accumbens/cytology , Photic Stimulation , Piperidines/pharmacology , Receptor, Muscarinic M1/physiology , Receptors, AMPA/physiology , Receptors, Dopamine D1/analysis , Receptors, Dopamine D2/analysis , Receptors, Neurokinin-1/physiology
10.
Neuropsychopharmacology ; 44(11): 1876-1885, 2019 10.
Article En | MEDLINE | ID: mdl-31132785

Enkephalins, endogenous ligands for delta opioid receptors (DORs), are highly enriched in the nucleus accumbens (NAc). They are implicated in depression but their role in the NAc, a critical brain region for motivated behavior, is not fully investigated. To provide insight into enkephalin function we used a chronic social defeat stress paradigm, where animals are either categorized as susceptible or resilient to stress based on their performance in a social interaction test. Compared to controls, susceptible animals showed reduced enkephalin levels in the NAc. Such decrease in enkephalin levels is not due to a change in mRNA of its precursor protein, proenkephalin, in susceptible mice but is consistent with increased mRNA levels of enkephalinases in the NAc of susceptible animals. Systemic administration of enkephalinase inhibitors or NAc infusion of the DOR agonist, SNC80, caused a resilient outcome to CSDS. Both treatments increased phosphorylation of ERK, which was downregulated by social defeat stress. To further validate these results, we also used Q175 knock-in mice, an animal model of Huntington's disease in which enkephalin levels are reduced in striatum and comorbidity with mood disorders is common. Consistent with data in wild-type mice, Q175 animals showed reduced enkephalin levels in the NAc and enhanced susceptibility to a social defeat stress. Overall, our data implicate that depression-like behavior induced by social defeat stress arises from disrupted DOR signaling resulting from lowered levels of enkephalins, which is partly mediated through elevated expression of enkephalinases.


Behavior, Animal/physiology , Dominance-Subordination , Enkephalins/metabolism , Nucleus Accumbens/metabolism , Stress, Psychological/metabolism , Analgesics, Opioid/pharmacology , Animals , Behavior, Animal/drug effects , Benzamides/pharmacology , Depression/metabolism , Disease Models, Animal , Mice , Mice, Inbred C57BL , Nucleus Accumbens/drug effects , Piperazines/pharmacology , Receptors, Opioid, delta/agonists
11.
Biol Psychiatry ; 85(12): 1001-1010, 2019 06 15.
Article En | MEDLINE | ID: mdl-30955841

BACKGROUND: Nucleus accumbens dopamine 1 receptor medium spiny neurons (D1-MSNs) play a critical role in the development of depression-like behavior in mice. Social defeat stress causes dendritic morphological changes on this MSN subtype through expression and activation of early growth response 3 (EGR3) and the Rho guanosine triphosphatase RhoA. However, it is unknown how RhoA inhibition affects electrophysiological properties underlying stress-induced susceptibility. METHODS: A novel RhoA-specific inhibitor, Rhosin, was used to inhibit RhoA activity following chronic social defeat stress. Whole-cell electrophysiological recordings of D1-MSNs were performed to assess synaptic and intrinsic consequences of Rhosin treatment on stressed mice. Additionally, recorded cells were filled and analyzed for their morphological properties. RESULTS: We found that RhoA inhibition prevents both D1-MSN hyperexcitability and reduced excitatory input to D1-MSNs caused by social defeat stress. Nucleus accumbens-specific RhoA inhibition is capable of blocking susceptibility caused by D1-MSN EGR3 expression. Lastly, we found that Rhosin enhances spine density, which correlates with D1-MSN excitability, without affecting overall dendritic branching. CONCLUSIONS: These findings demonstrate that pharmacological inhibition of RhoA during stress drives an enhancement of total spine number in a subset of nucleus accumbens neurons that prevents stress-related electrophysiological deficits and promotes stress resiliency.


Neuronal Plasticity , Neurons/physiology , Nucleus Accumbens/physiology , Organic Chemicals/administration & dosage , Receptors, Dopamine D1/physiology , Resilience, Psychological , Stress, Psychological/physiopathology , rhoA GTP-Binding Protein/physiology , Animals , Early Growth Response Protein 3/metabolism , Male , Mice, Inbred C57BL , Neuronal Plasticity/drug effects , Neurons/cytology , Neurons/drug effects , Nucleus Accumbens/cytology , Nucleus Accumbens/drug effects , Resilience, Psychological/drug effects , rhoA GTP-Binding Protein/antagonists & inhibitors
12.
Neuropharmacology ; 150: 38-45, 2019 05 15.
Article En | MEDLINE | ID: mdl-30851310

This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/our-business/policies/article-withdrawal). This article has been retracted at the request of the Authors. After publication, Scott M. Thompson found significant concerns about the data and duly notified The University of Maryland. The University of Maryland conducted an internal investigation which confirmed that the article was compromised. Namely in Figure 2B, the Investigation Committee determined that the western blots used to create the figure were not the ones used for the quantification and concluded that the figure was falsified to fit the hypothesis. In Figure 2C and 2D, the Investigation Committee determined that the densitometry data (pCaMKII, pS831, CamKII and GluA1) used to create the histogram were falsified to fit the hypothesis.


Excitatory Postsynaptic Potentials/drug effects , Fluoxetine/pharmacology , Hippocampus/drug effects , Selective Serotonin Reuptake Inhibitors/pharmacology , Synaptic Transmission/drug effects , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Excitatory Postsynaptic Potentials/physiology , Hippocampus/metabolism , Male , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Synapses/drug effects , Synapses/metabolism , Synaptic Transmission/physiology
13.
Nature ; 564(7735): 258-262, 2018 12.
Article En | MEDLINE | ID: mdl-30478293

Reward drives motivated behaviours and is essential for survival, and therefore there is strong evolutionary pressure to retain contextual information about rewarding stimuli. This drive may be abnormally strong, such as in addiction, or weak, such as in depression, in which anhedonia (loss of pleasure in response to rewarding stimuli) is a prominent symptom. Hippocampal input to the shell of the nucleus accumbens (NAc) is important for driving NAc activity1,2 and activity-dependent modulation of the strength of this input may contribute to the proper regulation of goal-directed behaviours. However, there have been few robust descriptions of the mechanisms that underlie the induction or expression of long-term potentiation (LTP) at these synapses, and there is, to our knowledge, no evidence about whether such plasticity contributes to reward-related behaviour. Here we show that high-frequency activity induces LTP at hippocampus-NAc synapses in mice via canonical, but dopamine-independent, mechanisms. The induction of LTP at this synapse in vivo drives conditioned place preference, and activity at this synapse is required for conditioned place preference in response to a natural reward. Conversely, chronic stress, which induces anhedonia, decreases the strength of this synapse and impairs LTP, whereas antidepressant treatment is accompanied by a reversal of these stress-induced changes. We conclude that hippocampus-NAc synapses show activity-dependent plasticity and suggest that their strength may be critical for contextual reward behaviour.


Anhedonia/physiology , Hippocampus/cytology , Hippocampus/physiology , Neuronal Plasticity , Nucleus Accumbens/cytology , Nucleus Accumbens/physiology , Reward , Synapses/metabolism , Animals , Chronic Disease , Conditioning, Psychological/physiology , Dopamine , Female , Goals , Long-Term Potentiation , Male , Mice , Stress, Psychological/physiopathology
14.
Neuron ; 96(6): 1327-1341.e6, 2017 12 20.
Article En | MEDLINE | ID: mdl-29268097

Altered brain energy homeostasis is a key adaptation occurring in the cocaine-addicted brain, but the effect of cocaine on the fundamental source of energy, mitochondria, is unknown. We demonstrate an increase of dynamin-related protein-1 (Drp1), the mitochondrial fission mediator, in nucleus accumbens (NAc) after repeated cocaine exposure and in cocaine-dependent individuals. Mdivi-1, a demonstrated fission inhibitor, blunts cocaine seeking and locomotor sensitization, while blocking c-Fos induction and excitatory input onto dopamine receptor-1 (D1) containing NAc medium spiny neurons (MSNs). Drp1 and fission promoting Drp1 are increased in D1-MSNs, consistent with increased smaller mitochondria in D1-MSN dendrites after repeated cocaine. Knockdown of Drp1 in D1-MSNs blocks drug seeking after cocaine self-administration, while enhancing the fission promoting Drp1 enhances seeking after long-term abstinence from cocaine. We demonstrate a role for altered mitochondrial fission in the NAc, during early cocaine abstinence, suggesting potential therapeutic treatment of disrupting mitochondrial fission in cocaine addiction.


Cocaine/pharmacology , Dopaminergic Neurons/drug effects , Dynamins/metabolism , Locomotion/drug effects , Mitochondria/metabolism , Receptors, Dopamine D1/metabolism , Animals , Cocaine/administration & dosage , Conditioning, Operant/drug effects , Conditioning, Operant/physiology , Dopamine Uptake Inhibitors/administration & dosage , Dopamine Uptake Inhibitors/pharmacology , Dopaminergic Neurons/ultrastructure , Dynamins/genetics , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Locomotion/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/drug effects , Neuronal Plasticity/drug effects , Neuronal Plasticity/genetics , Nucleus Accumbens/cytology , Quinazolinones/pharmacology , Receptors, Dopamine D1/genetics , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/metabolism , Self Administration
15.
J Neurosci ; 37(27): 6527-6538, 2017 07 05.
Article En | MEDLINE | ID: mdl-28576941

Previous research demonstrates that Slc6a15, a neutral amino acid transporter, is associated with depression susceptibility. However, no study examined Slc6a15 in the ventral striatum [nucleus accumbens (NAc)] in depression. Given our previous characterization of Slc6a15 as a striatal dopamine receptor 2 (D2)-neuron-enriched gene, we examined the role of Slc6a15 in NAc D2-neurons in mediating susceptibility to stress in male mice. First, we showed that Slc6a15 mRNA was reduced in NAc of mice susceptible to chronic social defeat stress (CSDS), a paradigm that produces behavioral and molecular adaptations that resemble clinical depression. Consistent with our preclinical data, we observed Slc6a15 mRNA reduction in NAc of individuals with major depressive disorder (MDD). The Slc6a15 reduction in NAc occurred selectively in D2-neurons. Next, we used Cre-inducible viruses combined with D2-Cre mice to reduce or overexpress Slc6a15 in NAc D2-neurons. Slc6a15 reduction in D2-neurons caused enhanced susceptibility to a subthreshold social defeat stress (SSDS) as observed by reduced social interaction, while a reduction in social interaction following CSDS was not observed when Slc6a15 expression in D2-neurons was restored. Finally, since both D2-medium spiny neurons (MSNs) and D2-expressing choline acetyltransferase (ChAT) interneurons express Slc6a15, we examined Slc6a15 protein in these interneurons after CSDS. Slc6a15 protein was unaltered in ChAT interneurons. Consistent with this, reducing Slc5a15 selectively in NAc D2-MSNs, using A2A-Cre mice that express Cre selectively in D2-MSNs, caused enhanced susceptibility to SSDS. Collectively, our data demonstrate that reduced Slc6a15 in NAc occurs in MDD individuals and that Slc6a15 reduction in NAc D2-neurons underlies stress susceptibility.SIGNIFICANCE STATEMENT Our study demonstrates a role for reduced Slc6a15, a neutral amino acid transporter, in nucleus accumbens (NAc) in depression and stress susceptibility. The reduction of Slc6a15 occurs selectively in the NAc D2-neurons. Genetic reduction of Slc6a15 induces susceptibility to a subthreshold stress, while genetic overexpression in D2-neurons prevents social avoidance after chronic social defeat stress.


Amino Acid Transport Systems, Neutral/metabolism , Dominance-Subordination , Dopaminergic Neurons/metabolism , Nucleus Accumbens/physiopathology , Receptors, Dopamine D2/metabolism , Stress, Psychological/physiopathology , Animals , Behavior, Animal , Disease Susceptibility/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Social Behavior
16.
Dev Cell ; 40(6): 583-594.e6, 2017 03 27.
Article En | MEDLINE | ID: mdl-28350990

Mitochondrial fission mediated by the GTPase dynamin-related protein 1 (Drp1) is an attractive drug target in numerous maladies that range from heart disease to neurodegenerative disorders. The compound mdivi-1 is widely reported to inhibit Drp1-dependent fission, elongate mitochondria, and mitigate brain injury. Here, we show that mdivi-1 reversibly inhibits mitochondrial complex I-dependent O2 consumption and reverse electron transfer-mediated reactive oxygen species (ROS) production at concentrations (e.g., 50 µM) used to target mitochondrial fission. Respiratory inhibition is rescued by bypassing complex I using yeast NADH dehydrogenase Ndi1. Unexpectedly, respiratory impairment by mdivi-1 occurs without mitochondrial elongation, is not mimicked by Drp1 deletion, and is observed in Drp1-deficient fibroblasts. In addition, mdivi-1 poorly inhibits recombinant Drp1 GTPase activity (Ki > 1.2 mM). Overall, these results suggest that mdivi-1 is not a specific Drp1 inhibitor. The ability of mdivi-1 to reversibly inhibit complex I and modify mitochondrial ROS production may contribute to effects observed in disease models.


Dynamins/antagonists & inhibitors , Electron Transport Complex I/antagonists & inhibitors , GTP Phosphohydrolases/antagonists & inhibitors , Microtubule-Associated Proteins/antagonists & inhibitors , Mitochondria/metabolism , Mitochondrial Proteins/antagonists & inhibitors , Quinazolinones/pharmacology , Reactive Oxygen Species/metabolism , Animals , COS Cells , Cell Respiration/drug effects , Chlorocebus aethiops , Dynamins/metabolism , Electron Transport Complex I/metabolism , Fibroblasts/metabolism , Fibroblasts/ultrastructure , GTP Phosphohydrolases/metabolism , Humans , Mice , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Mitochondria/drug effects , Mitochondrial Proteins/metabolism , NAD/metabolism , Neurons/metabolism , Oxidation-Reduction/drug effects , Oxygen Consumption/drug effects , Rats, Sprague-Dawley , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
17.
Int J Neuropsychopharmacol ; 20(5): 428-433, 2017 05 01.
Article En | MEDLINE | ID: mdl-28165117

Background: Polymorphisms in the CACNA1C gene are associated with human mood disorders. The rodent social defeat model of stress/mood-disorder susceptibility results in maladaptive consequences mediated by altered function of mesolimbic circuits. Methods: mRNA levels of Cacna1c in the nucleus accumbens of mice exposed to social defeat were assessed. Cacna1c was selectively deleted in the nucleus accumbens of floxed Cacna1c mice using viral Cre-recombinase to examine Cacna1c in social defeat susceptibility. Results: Reduced expression of Cacan1c in the nucleus accumbens is associated with increased susceptibility to social defeat stress, and a knockdown of Cacna1c in the nucleus accumbens significantly increases susceptibility measured by social interaction and female urine preference. Conclusions: Cacna1c reduction causally predisposes to the maladaptive outcomes of social stress. Normal Cacna1c function in the nucleus accumbens is crucial for resiliency to social stressors. Variations in expression of CACNA1C in the nucleus accumbens may mediate human risk for developing mood disorders and be a target for therapeutic intervention.


Calcium Channels, L-Type/metabolism , Genetic Predisposition to Disease/genetics , Nucleus Accumbens/metabolism , Stress, Psychological/genetics , Stress, Psychological/pathology , Animals , Anxiety/genetics , Calcium Channels, L-Type/genetics , Disease Models, Animal , Dominance-Subordination , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Smell/genetics , Transduction, Genetic
18.
Biol Psychiatry ; 81(7): 564-572, 2017 04 01.
Article En | MEDLINE | ID: mdl-27939396

BACKGROUND: Molecules critically involved in cocaine behavioral plasticity are known to regulate and interact with peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α). In addition, the PGC-1α promoter has binding sites for early growth response 3 (Egr3), which plays a dynamic role in cocaine action in nucleus accumbens (NAc) medium spiny neuron (MSN) subtypes, those enriched in dopamine receptor D1 (D1-MSN) versus D2 (D2-MSN). However, the role of PGC-1α in NAc in cocaine action is unknown. METHODS: PGC-1α messenger RNA and protein were examined in NAc after repeated cocaine exposure. Binding of Egr3 to and histone methylation at the PGC-1α promoter was examined in NAc using chromatin immunoprecipitation after repeated cocaine. PGC-1α ribosome-associated messenger RNA in MSN subtypes was assessed after repeated cocaine using D1-Cre-RiboTag and D2-Cre-RiboTag lines. Finally, PGC-1α was expressed in NAc D1-MSNs versus D2-MSNs using a Cre-inducible adeno-associated virus and Cre lines during cocaine conditioned place preference and cocaine-induced locomotion. RESULTS: Repeated cocaine increased PGC-1α levels and increased Egr3 binding and H3K4me3 at the PGC-1α promoter in NAc. Increased PGC-1α occurred in D1-MSNs, while D2-MSNs showed reduced levels. Viral-mediated expression of PGC-1α in D1-MSNs enhanced behavioral responses to cocaine, while expression in D2-MSNs blunted these behaviors. CONCLUSIONS: We demonstrate a novel role for PGC-1α in NAc in cocaine action. PGC-1α is enhanced in NAc D1-MSNs, specifically after cocaine exposure. These data are consistent with increased active methylation and Egr3 binding at the PGC-1α promoter. Finally, we demonstrate a bidirectional role for PGC-1α in mediating behavioral plasticity to cocaine through D1-MSNs versus D2-MSNs.


Cocaine/administration & dosage , Neurons/drug effects , Neurons/metabolism , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Animals , Early Growth Response Protein 3/metabolism , Histones/metabolism , Male , Methylation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic , RNA, Messenger/metabolism , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism
19.
Biol Psychiatry ; 81(8): 645-653, 2017 04 15.
Article En | MEDLINE | ID: mdl-27871668

The ventral striatum (nucleus accumbens) and its role in mood, reward, and motivation has been the focus of significant research. Despite this interest, little work has addressed cell type-specific distinctions in medium spiny neurons (MSNs), the main projection neurons in the nucleus accumbens and dorsal striatum, and their function in relation to stress and depression. Previous work has shown opposing roles for D1 and D2 receptor MSN subtypes in depression-like outcomes to stress, particularly in regard to repeated neuronal stimulation and excitatory transmission. Yet the mechanisms of action are still unknown. We discuss potential mechanisms by which MSN subtype function promotes dichotomous behavioral outcomes caused by differences in cellular plasticity, subcellular signaling pathways, and genetic expression. This review aims to address our current understanding about the role of nucleus accumbens MSN subtypes in stress-related depression behavior and speculates on how currently understood mechanisms contribute to factors that control the activity of MSNs.


Depressive Disorder/physiopathology , Neurons/physiology , Nucleus Accumbens/physiopathology , Stress, Psychological/physiopathology , Animals , Brain/physiopathology , Depression/physiopathology , Depressive Disorder/complications , Excitatory Postsynaptic Potentials , Female , Humans , Male , Mice , Mice, Inbred C57BL , Neural Pathways/physiopathology , Receptors, Dopamine D1/physiology , Receptors, Dopamine D2/physiology , Social Behavior , Stress, Psychological/complications
20.
J Neurosci ; 35(23): 8948-58, 2015 Jun 10.
Article En | MEDLINE | ID: mdl-26063926

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.


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
...