Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
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.
J Neurosci ; 43(5): 736-748, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36549906

ABSTRACT

The estrous cycle is a potent modulator of neuron physiology. In rodents, in vivo ventral tegmental area (VTA) dopamine (DA) activity has been shown to fluctuate across the estrous cycle. Although the behavioral effect of fluctuating sex steroids on the reward circuit is well studied in response to drugs of abuse, few studies have focused on the molecular adaptations in the context of stress and motivated social behaviors. We hypothesized that estradiol fluctuations across the estrous cycle acts on the dopaminergic activity of the VTA to alter excitability and stress response. We used whole-cell slice electrophysiology of VTA DA neurons in naturally cycling, adult female C57BL/6J mice to characterize the effects of the estrous cycle and the role of 17ß-estradiol on neuronal activity. We show that the estrous phase alters the effect of 17ß-estradiol on excitability in the VTA. Behaviorally, the estrous phase during a series of acute variable social stressors modulates subsequent reward-related behaviors. Pharmacological inhibition of estrogen receptors in the VTA before stress during diestrus mimics the stress susceptibility found during estrus, whereas increased potassium channel activity in the VTA before stress reverses stress susceptibility found during estrus as assessed by social interaction behavior. This study identifies one possible potassium channel mechanism underlying the increased DA activity during estrus and reveals estrogen-dependent changes in neuronal function. Our findings demonstrate that the estrous cycle and estrogen signaling changes the physiology of DA neurons resulting in behavioral differences when the reward circuit is challenged with stress.SIGNIFICANCE STATEMENT The activity of the ventral tegmental area encodes signals of stress and reward. Dopaminergic activity has been found to be regulated by both local synaptic inputs as well as inputs from other brain regions. Here, we provide evidence that cycling sex steroids also play a role in modulating stress sensitivity of dopaminergic reward behavior. Specifically, we reveal a correlation of ionic activity with estrous phase, which influences the behavioral response to stress. These findings shed new light on how estrous cycle may influence dopaminergic activity primarily during times of stress perturbation.


Subject(s)
Dopaminergic Neurons , Estrous Cycle , Mice , Animals , Female , Mice, Inbred C57BL , Dopaminergic Neurons/physiology , Estrous Cycle/physiology , Estrogens/pharmacology , Estradiol/pharmacology , Social Behavior , Mesencephalon , Potassium Channels , Ventral Tegmental Area
3.
Nat Commun ; 13(1): 6384, 2022 10 26.
Article in English | MEDLINE | ID: mdl-36289231

ABSTRACT

With an incidence of ~1 in 800 births, Down syndrome (DS) is the most common chromosomal condition linked to intellectual disability worldwide. While the genetic basis of DS has been identified as a triplication of chromosome 21 (HSA21), the genes encoded from HSA21 that directly contribute to cognitive deficits remain incompletely understood. Here, we found that the HSA21-encoded chromatin effector, BRWD1, was upregulated in neurons derived from iPS cells from an individual with Down syndrome and brain of trisomic mice. We showed that selective copy number restoration of Brwd1 in trisomic animals rescued deficits in hippocampal LTP, cognition and gene expression. We demonstrated that Brwd1 tightly binds the BAF chromatin remodeling complex, and that increased Brwd1 expression promotes BAF genomic mistargeting. Importantly, Brwd1 renormalization rescued aberrant BAF localization, along with associated changes in chromatin accessibility and gene expression. These findings establish BRWD1 as a key epigenomic mediator of normal neurodevelopment and an important contributor to DS-related phenotypes.


Subject(s)
Cognition Disorders , Down Syndrome , Mice , Animals , Down Syndrome/genetics , Down Syndrome/metabolism , DNA Copy Number Variations/genetics , Disease Models, Animal , Cognition Disorders/genetics , Chromatin/genetics , Mice, Transgenic
4.
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
5.
Front Neural Circuits ; 16: 1081099, 2022.
Article in English | MEDLINE | ID: mdl-36698552

ABSTRACT

The bed nucleus of the stria terminalis (BNST) is a highly heterogeneous limbic forebrain structure that serves as a relay connecting autonomic, neuroendocrine and behavioral function. It can be divided into over 16 individual subregions with distinct neuronal subpopulations based on receptors, transmitters, and neuropeptides. Specifically, the BNST projection to the ventral tegmental area (VTA), the dopamine hub of the brain, has been shown to have a crucial role in the stress response. However, in mice there is a lack of unbiased data on the functional diversity of this sub-population which serves as an upstream input to the VTA. The dopaminergic neurons in the VTA modify their ion channel activity and intrinsic membrane properties to adapt to stress in part from inputs from BNST projections. Therefore, we aimed to perform a multi-component characterization of the functional diversity of the BNST-VTA pathway. We studied the passive and active electrophysiological properties of virally identified population of BNST neurons that project to the VTA. We used a comprehensive series of in vitro recordings of electrophysiological variables and performed hierarchical clustering to determine the functional diversity of the projection neurons in the BNST-VTA pathway. Our study revealed four subpopulations in the BNST-VTA pathway, all of which differ in their activation profiles and likely have distinct inputs and function in the VTA. Our results will help resolve the discord in interpretation of the various roles of this electrophysiologically diverse projection and builds a foundation for understanding how the different neuronal types integrate signals.


Subject(s)
Septal Nuclei , Ventral Tegmental Area , Mice , Animals , Ventral Tegmental Area/physiology , Septal Nuclei/physiology , Dopaminergic Neurons/metabolism , Dopamine/metabolism , Interneurons
7.
J Neurosci ; 40(32): 6228-6233, 2020 08 05.
Article in English | MEDLINE | ID: mdl-32561672

ABSTRACT

Chronic stress in both humans and rodents induces a robust downregulation of neuroligin-2, a key component of the inhibitory synapse, in the NAc that modifies behavioral coping mechanisms and stress resiliency in mice. Here we extend this observation by examining the role of two other inhibitory synapse constituents, vesicular GABA transporter (vGAT) and gephyrin, in the NAc of male mice that underwent chronic social defeat stress (CSDS) and in patients with major depressive disorder (MDD). We first performed transcriptional profiling of vGAT and gephyrin in postmortem NAc samples from a cohort of healthy controls, medicated, and nonmedicated MDD patients. In parallel, we conducted whole-cell electrophysiology recordings in the NAc of stress-susceptible and stress-resilient male mice following 10 d of CSDS. Finally, we used immunohistochemistry to analyze protein levels of vGAT and gephyrin in the NAc of mice after CSDS. We found that decreased vGAT and gephyrin mRNA in the NAc of nonmedicated MDD patients is paralleled by decreased inhibitory synapse markers and decreased frequency of mini inhibitory postsynaptic currents (mIPSC) in the NAc of susceptible mice, indicating a reduction in the number of NAc inhibitory synapses that is correlated with depression-like behavior. Overall, these findings suggest a common state of reduced inhibitory tone in the NAc in depression and stress susceptibility.SIGNIFICANCE STATEMENT Existing studies focus on excitatory synaptic changes after social stress, although little is known about stress-induced inhibitory synaptic plasticity and its relevance for neuropsychiatric disease. These results extend our previous findings on the critical role of impaired inhibitory tone in the NAc following stress and provide new neuropathological evidence for reduced levels of inhibitory synaptic markers in human NAc from nonmedicated major depressive disorder patients. This finding is corroborated in stress-susceptible male mice that have undergone chronic social defeat stress, a mouse model of depression, at both the level of synaptic function and protein expression. These data support the hypothesis that reduced inhibitory synaptic transmission within the NAc plays a critical role in the stress response.


Subject(s)
Depression/metabolism , Inhibitory Postsynaptic Potentials , Nucleus Accumbens/physiopathology , Social Defeat , Stress, Psychological/metabolism , Adult , Aged , Animals , Depression/physiopathology , Female , Humans , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Middle Aged , Nucleus Accumbens/metabolism , Stress, Psychological/physiopathology , Vesicular Inhibitory Amino Acid Transport Proteins/genetics , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism
8.
Mol Psychiatry ; 25(6): 1323-1333, 2020 06.
Article in English | MEDLINE | ID: mdl-30385872

ABSTRACT

Major depressive disorder (MDD) is a leading cause of disability worldwide, yet current treatment strategies remain limited in their mechanistic diversity. Recent evidence has highlighted a promising novel pharmaceutical target-the KCNQ-type potassium channel-for the treatment of depressive disorders, which may exert a therapeutic effect via functional changes within the brain reward system, including the ventral striatum. The current study assessed the effects of the KCNQ channel opener ezogabine (also known as retigabine) on reward circuitry and clinical symptoms in patients with MDD. Eighteen medication-free individuals with MDD currently in a major depressive episode were enrolled in an open-label study and received ezogabine up to 900 mg/day orally over the course of 10 weeks. Resting-state functional magnetic resonance imaging data were collected at baseline and posttreatment to examine brain reward circuitry. Reward learning was measured using a computerized probabilistic reward task. After treatment with ezogabine, subjects exhibited a significant reduction of depressive symptoms (Montgomery-Asberg Depression Rating Scale score change: -13.7 ± 9.7, p < 0.001, d = 2.08) and anhedonic symptoms (Snaith-Hamilton Pleasure Scale score change: -6.1 ± 5.3, p < 0.001, d = 1.00), which remained significant even after controlling for overall depression severity. Improvement in depression was associated with decreased functional connectivity between the ventral caudate and clusters within the mid-cingulate cortex and posterior cingulate cortex (n = 14, voxel-wise p < 0.005). In addition, a subgroup of patients tested with a probabilistic reward task (n = 9) showed increased reward learning following treatment. These findings highlight the KCNQ-type potassium channel as a promising target for future drug discovery efforts in mood disorders.


Subject(s)
Carbamates/pharmacology , Carbamates/therapeutic use , Depressive Disorder, Major/drug therapy , Ion Channel Gating/drug effects , Phenylenediamines/pharmacology , Phenylenediamines/therapeutic use , Ventral Striatum/drug effects , Depressive Disorder, Major/metabolism , Female , Humans , KCNQ Potassium Channels/agonists , KCNQ Potassium Channels/metabolism , Magnetic Resonance Imaging , Male , Middle Aged , Reward , Ventral Striatum/metabolism
9.
Curr Biol ; 29(3): 435-448.e8, 2019 02 04.
Article in English | MEDLINE | ID: mdl-30661803

ABSTRACT

Age-related declines in cognitive fitness are associated with a reduction in autophagy, an intracellular lysosomal catabolic process that regulates protein homeostasis and organelle turnover. However, the functional significance of autophagy in regulating cognitive function and its decline during aging remains largely elusive. Here, we show that stimulating memory upregulates autophagy in the hippocampus. Using hippocampal injections of genetic and pharmacological modulators of autophagy, we find that inducing autophagy in hippocampal neurons is required to form novel memory by promoting activity-dependent structural and functional synaptic plasticity, including dendritic spine formation, neuronal facilitation, and long-term potentiation. We show that hippocampal autophagy activity is reduced during aging and that restoring its levels is sufficient to reverse age-related memory deficits. Moreover, we demonstrate that systemic administration of young plasma into aged mice rejuvenates memory in an autophagy-dependent manner, suggesting a prominent role for autophagy to favor the communication between systemic factors and neurons in fostering cognition. Among these youthful factors, we identify osteocalcin, a bone-derived molecule, as a direct hormonal inducer of hippocampal autophagy. Our results reveal that inducing autophagy in hippocampal neurons is a necessary mechanism to enhance the integration of novel stimulations of memory and to promote the influence of systemic factors on cognitive fitness. We also demonstrate the potential therapeutic benefits of modulating autophagy in the aged brain to counteract age-related cognitive impairments.


Subject(s)
Aging/physiology , Autophagy/physiology , Hippocampus/physiology , Memory Disorders , Memory/physiology , Animals , Autophagy/drug effects , Autophagy/genetics , Disease Models, Animal , Male , Memory/drug effects , Memory Disorders/physiopathology , Mice , Mice, Inbred C57BL
10.
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
11.
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.

12.
J Neurosci ; 38(3): 575-585, 2018 01 17.
Article in English | MEDLINE | ID: mdl-29196318

ABSTRACT

Cocaine self-administration increases expression of GluA1 subunits in ventral tegmental area (VTA) dopamine neurons, which subsequently enhance the motivation for cocaine. This increase in GluA1 may be dependent on concomitant NMDA receptor (NMDAR) activation during self-administration, similar to cocaine-induced long-term potentiation in the VTA. In this study, we used viral-mediated expression of a dominant-negative GluN1 subunit (HSV-dnGluN1) in VTA neurons to study the effect of transient NMDAR inactivation on the GluA1 increases induced by chronic cocaine self-administration in male rats. We found that dnGluN1 expression in the VTA limited to the 3 weeks of cocaine self-administration prevents the subsequent increase in tissue GluA1 levels when compared with control infusions of HSV-LacZ. Surprisingly, dnGluN1 expression led to an enhancement in the motivation to self-administer cocaine as measured using a progressive ratio reinforcement schedule and to enhanced cocaine seeking measured in extinction/reinstatement tests following an extended 3 week withdrawal period. Despite blocking tissue GluA1 increases in cocaine self-administering animals, the HSV-dnGluN1 treatment resulted in increased membrane levels of GluA1 and GluN2B, along with markedly higher locomotor responses to intra-VTA infusions of AMPA, suggesting a paradoxical increase in VTA AMPA receptor responsiveness. Together, these data suggest that NMDARs mediate cocaine-induced increases in VTA GluA1 expression, but such transient NMDAR inactivation also leads to compensatory scaling of synaptic AMPA receptors that enhance the motivational for cocaine.SIGNIFICANCE STATEMENT Dopamine neurons in the ventral tegmental area (VTA) are critical substrates of drug rewards. Animal models indicate that chronic cocaine use enhances excitatory glutamatergic input to these neurons, making them more susceptible to environmental stimuli that trigger drug craving and relapse. We previously found that self-administration of cocaine increases AMPA glutamate receptors in the VTA, and this effect enhances motivation for cocaine. Here we report that the mechanism for this upregulation involves NMDA receptor activity during cocaine use. While interference with NMDA receptor function blocks AMPA receptor upregulation, it also produces a paradoxical enhancement in membrane AMPA receptor subunits, AMPA responsiveness, and the motivation for cocaine. Thus, pharmacotherapy targeting NMDA receptors may inadvertently produce substantial adverse consequences for cocaine addiction.


Subject(s)
Cocaine-Related Disorders/metabolism , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Ventral Tegmental Area/metabolism , Animals , Cocaine/pharmacology , Cocaine-Related Disorders/physiopathology , Dopamine Uptake Inhibitors/pharmacology , Drug-Seeking Behavior/physiology , Male , Rats , Rats, Sprague-Dawley , Self Administration , Up-Regulation , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/physiopathology
13.
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
14.
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
15.
Biol Psychiatry ; 80(6): 469-478, 2016 09 15.
Article in English | MEDLINE | ID: mdl-26858215

ABSTRACT

BACKGROUND: Previous work has shown that chronic social defeat stress (CSDS) induces increased phasic firing of ventral tegmental area (VTA) dopamine (DA) neurons that project to the nucleus accumbens (NAc) selectively in mice that are susceptible to the deleterious effects of the stress. In addition, acute optogenetic phasic stimulation of these neurons promotes susceptibility in animals exposed to acute defeat stress. These findings are paradoxical, as increased DA signaling in NAc normally promotes motivation and reward, and the influence of chronic phasic VTA firing in the face of chronic stress is unknown. METHODS: We used CSDS with repeated optogenetic activation and pharmacologic manipulations of the mesolimbic VTA-NAc pathway to examine the role of brain-derived neurotrophic factor (BDNF) and DA signaling in depressive-like behaviors. We measured BDNF protein expression and DA release in this model. RESULTS: Pharmacologic blockade of BDNF-tyrosine receptor kinase B (TrkB) signaling, but not DA signaling, in NAc prevented CSDS-induced behavioral abnormalities. Chronic optogenetic phasic stimulation of the VTA-NAc circuit during CSDS exacerbated the defeat-induced behavioral symptoms, and these aggravated symptoms were also normalized by BDNF-TrkB blockade in NAc. The aggravated behavioral deficits induced by phasic stimulation of the VTA-NAc pathway were blocked as well by local knockdown of BDNF in VTA. CONCLUSIONS: These findings show that BDNF-TrkB signaling, rather than DA signaling, in the VTA-NAc circuit is crucial for facilitating depressive-like outcomes after CSDS and they establish BDNF-TrkB signaling as a pathologic mechanism during periods of chronic stress.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Depression/physiopathology , Depression/psychology , Nucleus Accumbens/physiology , Social Behavior , Stress, Psychological/physiopathology , Ventral Tegmental Area/physiology , Animals , Azepines/administration & dosage , Azepines/pharmacology , Benzamides/administration & dosage , Benzamides/pharmacology , Benzazepines/administration & dosage , Benzazepines/pharmacology , Brain-Derived Neurotrophic Factor/biosynthesis , Brain-Derived Neurotrophic Factor/genetics , Cocaine/pharmacology , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Dopamine/metabolism , Gene Knockdown Techniques , Male , Mice , Mice, Transgenic , Microinjections , Neural Pathways/physiology , Nucleus Accumbens/drug effects , Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor, trkB , Salicylamides/administration & dosage , Salicylamides/pharmacology
16.
Curr Protoc Neurosci ; 72: 4.36.1-4.36.7, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26131662

ABSTRACT

Herpes simplex virus (HSV) can be used for a wide range of genetic manipulations in ex vivo slices of central nervous system tissue from both young and adult rodents. The fast expression of the HSV viral-mediated gene transfer, which can be engineered to produce cell-type specificity, can be utilized in slice cultures for a variety of purposes over a 1- to 4-day period with spatial and temporal specificity. This protocol exploits the rapid expression of HSV viral vectors by utilizing slice culture for electrophysiological recordings, avoiding the need to do intracranial viral injections. Brain slice cultures maintain many aspects of in vivo biology, including functional local synaptic circuitry with preserved brain architecture, while allowing good experimental access and precise control of the extracellular environment, making them ideal platforms for quick access to evaluate expression effects of HSV viral-mediated gene transfer on the molecular and cellular properties of specific neurons. This protocol provides an easy way to study neuronal function following viral expression of a gene of interest.


Subject(s)
Brain/cytology , Genetic Vectors/physiology , Herpesvirus 1, Human/genetics , Neurons/metabolism , Organ Culture Techniques , Action Potentials/genetics , Action Potentials/physiology , Age Factors , Animals , Gene Transfer Techniques , In Vitro Techniques
17.
Nat Neurosci ; 18(7): 962-4, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26030846

ABSTRACT

Postsynaptic remodeling of glutamatergic synapses on ventral striatum (vSTR) medium spiny neurons (MSNs) is critical for shaping stress responses. However, it is unclear which presynaptic inputs are involved. Susceptible mice exhibited increased synaptic strength at intralaminar thalamus (ILT), but not prefrontal cortex (PFC), inputs to vSTR MSNs following chronic social stress. Modulation of ILT-vSTR versus PFC-vSTR neuronal activity differentially regulated dendritic spine plasticity and social avoidance.


Subject(s)
Dendritic Spines/physiology , Excitatory Postsynaptic Potentials/physiology , Neuronal Plasticity/physiology , Prefrontal Cortex/physiology , Stress, Psychological/physiopathology , Thalamus/physiology , Ventral Striatum/physiology , Animals , Behavior, Animal/physiology , Disease Susceptibility , Male , Mice , Mice, Inbred C57BL , Reward , Social Behavior , Ventral Striatum/cytology
18.
J Neurosci ; 35(16): 6326-34, 2015 Apr 22.
Article in English | MEDLINE | ID: mdl-25904786

ABSTRACT

We investigate stimulus specificity of repetition priming in a tractable model system; the feeding network of Aplysia. Previous studies primarily focused on an aspect of behavior that is altered during ingestive priming, radula opening. Priming of radula opening occurs when two modulatory peptides [feeding circuit activating peptide (FCAP) and cerebral peptide-2 (CP-2)] are released from the cholinergic command-like neuron cerebral buccal interneuron 2. Effects of FCAP/CP-2 on radula opening motor neurons are cAMP mediated. The present experiments sought to determine whether FCAP/CP-2 and cAMP are also involved in the priming of radula opening during an incompatible activity, i.e., during egestive motor programs. Egestive priming is induced when motor programs are triggered by afferents with processes in the esophageal nerve. We demonstrate that egestive priming is not FCAP/CP-2 mediated. Instead, it is induced by an unrelated peptide (small cardioactive peptide), which exerts PKC-mediated effects. Our data, therefore, suggest that different feeding motor programs are primed via actions of different sets of intercellular and intracellular substances. We suggest that this accounts for the stimulus specificity that can be characteristic of repetition priming. Different stimuli activate different central pattern generator inputs. These inputs release different modulators, which induce functionally distinct motor programs.


Subject(s)
Neuropeptides/physiology , Repetition Priming/physiology , Synaptic Transmission/physiology , Animals , Aplysia , Feeding Behavior/physiology
19.
Curr Opin Neurobiol ; 29: 33-8, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25261622

ABSTRACT

It is becoming apparent that the activity of many neural networks is shaped by effects of endogenous neuromodulators. Modulators exert second messenger-mediated actions that persist. We consider how this may impact network function and its potential role in the induction of repetition priming (increased performance when behavior is repeated). When effects of modulators persist and modulatory substances are repeatedly released, their effects will accumulate (summate) and become more pronounced. If this enhances the ability of a network to generate a particular output, performance will improve. We review data that support this model, and consider its implications for task switching. This model predicts that priming of one type of network activity will negatively impact the rapid transition to an incompatible type.


Subject(s)
Learning , Nerve Net/physiology , Neuronal Plasticity/physiology , Neurons/physiology , Neurotransmitter Agents/physiology , Animals , Humans , Models, Neurological
20.
Nat Neurosci ; 17(8): 1055-63, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24952642

ABSTRACT

Ion channels are essential for the regulation of neuronal functions. The significance of plasma membrane, mitochondrial, endoplasmic reticulum and lysosomal ion channels in the regulation of Ca(2+) is well established. In contrast, surprisingly little is known about the function of ion channels on the nuclear envelope (NE). Here we demonstrate the presence of functional large-conductance, calcium-activated potassium channels (BK channels) on the NE of rodent hippocampal neurons. Functionally, blockade of nuclear BK channels (nBK channels) induces NE-derived Ca(2+) release, nucleoplasmic Ca(2+) elevation and cyclic AMP response element binding protein (CREB)-dependent transcription. More importantly, blockade of nBK channels regulates nuclear Ca(2+)-sensitive gene expression and promotes dendritic arborization in a nuclear Ca(2+)-dependent manner. These results suggest that the nBK channel functions as a molecular link between neuronal activity and nuclear Ca(2+) to convey signals from synapse to nucleus and is a new modulator, operating at the NE, of synaptic activity-dependent neuronal functions.


Subject(s)
Calcium Signaling/genetics , Gene Expression Regulation , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channels/physiology , Nuclear Envelope/genetics , Nuclear Envelope/metabolism , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/deficiency , Large-Conductance Calcium-Activated Potassium Channels/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/cytology , Neurons/metabolism , Nuclear Envelope/ultrastructure , Phosphorylation/genetics , Rats , Rats, Sprague-Dawley , Synaptic Transmission/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...