Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Biol Psychiatry ; 93(2): 197-208, 2023 01 15.
Article in English | MEDLINE | ID: mdl-35961792

ABSTRACT

BACKGROUND: Neuropeptides are contained in nearly every neuron in the central nervous system and can be released not only from nerve terminals but also from somatodendritic sites. Cholecystokinin (CCK), among the most abundant neuropeptides in the brain, is expressed in the majority of midbrain dopamine neurons. Despite this high expression, CCK function within the ventral tegmental area (VTA) is not well understood. METHODS: We confirmed CCK expression in VTA dopamine neurons through immunohistochemistry and in situ hybridization and detected optogenetically induced CCK release using an enzyme-linked immunosorbent assay. To investigate whether CCK modulates VTA circuit activity, we used whole-cell patch clamp recordings in mouse brain slices. We infused CCK locally in vivo and tested food intake and locomotion in fasted mice. We also used in vivo fiber photometry to measure Ca2+ transients in dopamine neurons during feeding. RESULTS: Here we report that VTA dopamine neurons release CCK from somatodendritic regions, where it triggers long-term potentiation of GABAergic (gamma-aminobutyric acidergic) synapses. The somatodendritic release occurs during trains of optogenetic stimuli or prolonged but modest depolarization and is dependent on synaptotagmin-7 and T-type Ca2+ channels. Depolarization-induced long-term potentiation is blocked by a CCK2 receptor antagonist and mimicked by exogenous CCK. Local infusion of CCK in vivo inhibits food consumption and decreases distance traveled in an open field test. Furthermore, intra-VTA-infused CCK reduced dopamine cell Ca2+ signals during food consumption after an overnight fast and was correlated with reduced food intake. CONCLUSIONS: Our experiments introduce somatodendritic neuropeptide release as a previously unknown feedback regulator of VTA dopamine cell excitability and dopamine-related behaviors.


Subject(s)
Dopamine , Ventral Tegmental Area , Mice , Animals , Dopamine/metabolism , Cholecystokinin/metabolism , Cholecystokinin/pharmacology , Synapses/metabolism , Dopaminergic Neurons
2.
Neuroscience ; 461: 11-22, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33689861

ABSTRACT

Despite the high prevalence of major depressive disorder (MDD), understanding of the biological underpinnings remains limited. Rodent models suggest that changes in activity and output of dopamine (DA) neurons in the ventral tegmental area (VTA) are important for depressive-like phenotypes. Additionally, brain inflammatory processes are thought to contribute to MDD pathology and inflammation in the VTA has been linked to changes in VTA DA neuronal activity. Thus, we sought to determine whether there is increased inflammatory signaling in the VTA following forms of chronic stress that induce depressive-like symptoms. First, we subjected male mice to either physical or vicarious chronic social defeat stress (CSDS), paradigms known to induce long-term depressive-like behavior and changes in VTA signaling. Second, we subjected male and female mice to subchronic variable stress (SCVS), a paradigm that induces depressive-like behavior only in female mice. We then isolated mRNA from the VTA and assessed proinflammatory gene regulation via RT-PCR. Our results show that physical, but not vicarious, CSDS increases interleukin 1ß (IL-1ß) mRNA expression and this inversely correlates with social interaction score. In contrast, IL-1ß expression was unchanged in male or female mice following SCVS. No significant increases in VTA ionized calcium binding adapter molecule 1 (Iba1) and glial fibrillary acidic protein (GFAP) immunochemistry were detected following CSDS that would be indicative of a robust inflammatory response. In conclusion, we show that chronic stressors distinctively alter expression of proinflammatory genes in the VTA and changes may depend on the severity and time-course of the stress exposure.


Subject(s)
Depressive Disorder, Major , Ventral Tegmental Area , Animals , Disease Models, Animal , Dopaminergic Neurons , Female , Male , Mice , Stress, Psychological
3.
Behav Brain Res ; 401: 113084, 2021 03 05.
Article in English | MEDLINE | ID: mdl-33358922

ABSTRACT

Women are more likely to suffer from stress-related affective disorders than men, but the underlying mechanisms of sex differences remain unclear. Previous works show that microRNA (miRNA) profiles are altered in stressed animals and patients with depression and anxiety disorders. In this study, we investigated how miRNA expression in the anterior bed nucleus of stria terminalis (BNST) was affected by social defeat stress in female and male California mice (Peromyscus californicus). We performed sequencing to identify miRNA transcripts in the whole brain and anterior BNST followed by qPCR analysis to compare miRNA expression between control and stressed animals. The results showed that social defeat stress induced sex-specific miRNA expression changes in the anterior BNST. Let-7a, let-7f and miR-181a-5p were upregulated in stressed female but not male mice. Our study provided evidence that social stress produces distinct molecular responses in the BNST of males and females.


Subject(s)
Behavior, Animal/physiology , MicroRNAs/metabolism , Septal Nuclei/metabolism , Sex Characteristics , Social Defeat , Stress, Psychological/metabolism , Animals , Female , Male , Peromyscus , Sequence Analysis, RNA , Up-Regulation
4.
Nat Commun ; 11(1): 4484, 2020 09 08.
Article in English | MEDLINE | ID: mdl-32901027

ABSTRACT

Chronic stress is a key risk factor for mood disorders like depression, but the stress-induced changes in brain circuit function and gene expression underlying depression symptoms are not completely understood, hindering development of novel treatments. Because of its projections to brain regions regulating reward and anxiety, the ventral hippocampus is uniquely poised to translate the experience of stress into altered brain function and pathological mood, though the cellular and molecular mechanisms of this process are not fully understood. Here, we use a novel method of circuit-specific gene editing to show that the transcription factor ΔFosB drives projection-specific activity of ventral hippocampus glutamatergic neurons causing behaviorally diverse responses to stress. We establish molecular, cellular, and circuit-level mechanisms for depression- and anxiety-like behavior in response to stress and use circuit-specific gene expression profiling to uncover novel downstream targets as potential sites of therapeutic intervention in depression.


Subject(s)
Avoidance Learning/physiology , Hippocampus/physiology , Proto-Oncogene Proteins c-fos/physiology , Animals , Anxiety/metabolism , Behavior, Animal/physiology , Gene Knockout Techniques , Gene Silencing , Hippocampus/anatomy & histology , Hippocampus/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Proto-Oncogene Proteins c-fos/deficiency , Proto-Oncogene Proteins c-fos/genetics , Social Behavior , Stress, Psychological
5.
Schizophr Bull ; 46(1): 54-67, 2020 01 04.
Article in English | MEDLINE | ID: mdl-31150554

ABSTRACT

Impairments in reality testing are core features of numerous neuropsychiatric conditions. However, relatively few animal models have been developed to assess this critical facet of neuropsychiatric illness, thus impeding our understanding of the underlying central systems and circuits. Using mice in which dominant-negative Disrupted-in-Schizophrenia-1 is expressed throughout central nervous system circuitry (DN-DISC1-PrP), the capacity for an auditory conditioned stimulus (CS) to evoke perceptual processing of an absent sucrose solution was examined. At test, during CS presentations, DN-DISC1-PrP mice consumed more water and displayed a licking profile that is more typically revealed while ingesting a sweet-tasting solution. DN-DISC1-PrP mice also displayed greater c-fos expression in the insular (gustatory) cortex when consuming water in the presence of the CS. This capacity for the CS to more readily substitute for the taste features of the absent sucrose solution in DN-DISC1-PrP mice was attenuated following systemic treatment with the antipsychotic haloperidol. Conversely, social isolation during adolescence promoted the manifestation of these effects. These results provide strong validation for using associative learning procedures to examine dopamine-mediated reality testing associated with insular cortex activation.


Subject(s)
Association Learning/physiology , Behavior, Animal/physiology , Cerebral Cortex/physiopathology , Delusions/physiopathology , Dopamine/physiology , Hallucinations/physiopathology , Reality Testing , Reward , Taste Perception/physiology , Animals , Antipsychotic Agents/pharmacology , Auditory Perception/physiology , Behavior, Animal/drug effects , Cerebral Cortex/drug effects , Conditioning, Classical/physiology , Delusions/drug therapy , Disease Models, Animal , Hallucinations/drug therapy , Haloperidol/pharmacology , Mice , Mice, Transgenic , Nerve Tissue Proteins , Social Isolation , Taste Perception/drug effects
6.
Biol Psychiatry ; 87(6): 492-501, 2020 03 15.
Article in English | MEDLINE | ID: mdl-31601425

ABSTRACT

BACKGROUND: Depression affects women nearly twice as often as men, but the neurobiological underpinnings of this discrepancy are unclear. Preclinical studies in male mice suggest that activity of ventral hippocampus (vHPC) neurons projecting to the nucleus accumbens (NAc) regulates mood-related behavioral responses to stress. We sought to characterize this circuit in both sexes and to investigate its role in potential sex differences in models of depression. METHODS: We used male and female adult C57BL/6J mice in the subchronic variable stress model to precipitate female-specific reduction in sucrose preference and performed gonadectomies to test the contributions of gonadal hormones to this stress response. In addition, ex vivo slice electrophysiology of transgenic Cre-inducible Rosa-eGFP-L10a mice in combination with retrograde viral tracing to identify circuits was used to test contributions of gonadal hormones to sex differences in vHPC afferents. Finally, we used an intersecting viral DREADD (designer receptor exclusively activated by designer drugs) strategy to manipulate vHPC-NAc excitability directly in awake behaving mice. RESULTS: We show a testosterone-dependent lower excitability in male versus female vHPC-NAc neurons and corresponding testosterone-dependent male resilience to reduced sucrose preference after subchronic variable stress. Importantly, we show that long-term DREADD stimulation of vHPC-NAc neurons causes decreased sucrose preference in male mice after subchronic variable stress, whereas DREADD inhibition of this circuit prevents this effect in female mice. CONCLUSIONS: We demonstrate a circuit-specific sex difference in vHPC-NAc neurons that is dependent on testosterone and causes susceptibility to stress in female mice. These data provide a substantive mechanism linking gonadal hormones to cellular excitability and anhedonia-a key feature in depressive states.


Subject(s)
Androgens , Nucleus Accumbens , Animals , Female , Hippocampus , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
7.
J Neurosci ; 39(42): 8305-8314, 2019 10 16.
Article in English | MEDLINE | ID: mdl-31477569

ABSTRACT

Drug addiction results in part from maladaptive learning, including the formation of strong associations between the drug and the circumstances of consumption. However, drug-induced changes in gene expression underlying the saliency of these associations remain understudied. Consolidation of explicit memories occurs within the hippocampus, and we have shown that spatial learning induces expression of the transcription factor ΔFosB in hippocampus and that this induction is critical for learning. Drugs of abuse also upregulate ΔFosB in hippocampus, but the mechanism of its induction by cocaine and its role in hippocampus-dependent cocaine responses is unknown. We investigated differences in mouse dorsal and ventral hippocampal ΔFosB expression in response to chronic cocaine, because these regions appear to regulate distinct cocaine-related behaviors. We found that cocaine-mediated induction of ΔFosB was subregion-specific, and that ΔFosB transcriptional activity in both the dorsal and ventral hippocampus is necessary for cocaine conditioned place preference. Further, we characterize changes in histone modifications at the FosB promoter in hippocampus in response to chronic cocaine and found that locus-specific epigenetic modification is essential for FosB induction and multiple hippocampus-dependent behaviors, including cocaine place preference. Collectively, these findings suggest that exposure to cocaine induces histone modification at the hippocampal FosB gene promoter to cause ΔFosB induction critical for cocaine-related learning.SIGNIFICANCE STATEMENT Although cocaine addiction is driven in part by the formation of indelible associations between the drug and the environment, paraphernalia, and circumstances of use, and although this type of associative learning is dependent upon changes in gene expression in a brain region called the hippocampus, the mechanisms by which cocaine alters hippocampal gene expression to drive formation of these associations is poorly understood. Here, we demonstrate that chronic cocaine engages locus-specific changes in the epigenetic profile of the FosB gene in the hippocampus, and that these alterations are required for cocaine-dependent gene expression and cocaine-environment associations. This work provides novel insight into addiction etiology and potential inroads for therapeutic intervention in cocaine addiction.


Subject(s)
Cocaine/administration & dosage , Dopamine Uptake Inhibitors/administration & dosage , Epigenesis, Genetic/drug effects , Gene Expression Regulation/drug effects , Hippocampus/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Animals , Hippocampus/drug effects , Male , Mice , Motor Activity/drug effects , Up-Regulation/drug effects
8.
Neuroscience ; 406: 225-233, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30902680

ABSTRACT

Neural proliferation in the dentate gyrus (DG) is closely linked with learning and memory, but the transcriptional programming that drives adult proliferation remains incompletely understood. Our lab previously elucidated the critical role of the transcription factor ΔFosB in the dorsal hippocampus (dHPC) in learning and memory, and the FosB gene has been suggested to play a role in neuronal proliferation. However, the subregion-specific and potentially cell-autonomous role of dHPC ΔFosB in neurogenesis-dependent learning has not been studied. Here, we crossed neurotensin receptor-2 (NtsR2) Cre mice, which express Cre within the subgranular zone (SGZ) of dHPC DG, with floxed FosB mice to show that knockout of ΔFosB in hippocampal SGZ neurons reduces antidepressant-induced neurogenesis and impedes hippocampus-dependent learning in the novel object recognition task. Taken together, these data indicate that FosB gene expression in SGZ is necessary for both hippocampal neurogenesis and memory formation.


Subject(s)
Hippocampus/metabolism , Maze Learning/physiology , Neurogenesis/physiology , Neurons/metabolism , Proto-Oncogene Proteins c-fos/biosynthesis , Animals , Female , Hippocampus/cytology , Learning/physiology , Male , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-fos/genetics
9.
Front Behav Neurosci ; 11: 77, 2017.
Article in English | MEDLINE | ID: mdl-28503137

ABSTRACT

Over the past three decades, it has become clear that aberrant function of the network of interconnected brain regions responsible for reward processing and motivated behavior underlies a variety of mood disorders, including depression and anxiety. It is also clear that stress-induced changes in reward network activity underlying both normal and pathological behavior also cause changes in gene expression. Here, we attempt to define the reward circuitry and explore the known and potential contributions of activity-dependent changes in gene expression within this circuitry to stress-induced changes in behavior related to mood disorders, and contrast some of these effects with those induced by exposure to drugs of abuse. We focus on a series of immediate early genes regulated by stress within this circuitry and their connections, both well-explored and relatively novel, to circuit function and subsequent reward-related behaviors. We conclude that IEGs play a crucial role in stress-dependent remodeling of reward circuitry, and that they may serve as inroads to the molecular, cellular, and circuit-level mechanisms of mood disorder etiology and treatment.

10.
Biol Psychiatry ; 80(5): 406-14, 2016 09 01.
Article in English | MEDLINE | ID: mdl-26620251

ABSTRACT

BACKGROUND: Oxytocin (OT) is considered to be a stress-buffering hormone, dampening the physiologic effects of stress. However, OT can also be anxiogenic. We examined acute and long-lasting effects of social defeat on OT neurons in male and female California mice. METHODS: We used immunohistochemistry for OT and c-fos cells to examine OT neuron activity immediately after defeat (n = 6-9) and 2 weeks (n = 6-9) and 10 weeks (n = 4-5) later. We quantified Oxt messenger RNA with quantitative polymerase chain reaction (n = 5-9). Intranasal OT was administered to naïve and stressed mice tested in social interaction and resident-intruder tests (n = 8-14). RESULTS: Acute exposure to a third episode of defeat increased OT/c-fos colocalizations in the paraventricular nucleus of both sexes. In the medioventral bed nucleus of the stria terminalis, defeat increased Oxt messenger RNA, total OT neurons, and OT/c-fos colocalizations in female mice but not male mice. Intranasal OT failed to reverse stress-induced social withdrawal in female mice and reduced social interaction behavior in female mice naïve to defeat. In contrast, intranasal OT increased social interaction in stressed male mice and reduced freezing in the resident-intruder test. CONCLUSIONS: Social defeat induces long-lasting increases in OT production and OT/c-fos cells in the medioventral bed nucleus of the stria terminalis of female mice but not male mice. Intranasal OT largely reversed the effects of stress on behavior in male mice, but effects were mixed in female mice. These results suggest that changes in OT-sensitive networks contribute to sex differences in behavioral responses to stress.


Subject(s)
Neurons/drug effects , Oxytocin/administration & dosage , Oxytocin/metabolism , Sex Characteristics , Stress, Psychological/drug therapy , Stress, Psychological/pathology , Administration, Intranasal , Animals , Disease Models, Animal , Female , Male , Mice , Neurons/metabolism , Oxytocin/genetics , Paraventricular Hypothalamic Nucleus/cytology , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/metabolism , Social Behavior , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...