Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Curr Opin Neurobiol ; 85: 102839, 2024 04.
Article in English | MEDLINE | ID: mdl-38309106

ABSTRACT

Striatal dopamine governs a wide range of behavioral functions, yet local dopamine concentrations can be dissociated from somatic activity. Here, we discuss how dopamine's diverse roles in behavior may be driven by local circuit mechanisms shaping dopamine release. We first look at historical and recent work demonstrating that striatal circuits interact with dopaminergic terminals to either initiate the release of dopamine or modulate the release of dopamine initiated by spiking in midbrain dopamine neurons, with particular attention to GABAergic and cholinergic local circuit mechanisms. Then we discuss some of the first in vivo studies of acetylcholine-dopamine interactions in striatum and broadly discuss necessary future work in understanding the roles of midbrain versus striatal dopamine regulation.


Subject(s)
Corpus Striatum , Dopamine , Dopamine/physiology , Corpus Striatum/physiology , Acetylcholine , Dopaminergic Neurons/physiology
2.
Nat Commun ; 14(1): 1920, 2023 04 06.
Article in English | MEDLINE | ID: mdl-37024449

ABSTRACT

Fronto-striatal circuits have been implicated in cognitive control of behavioral output for social and appetitive rewards. The functional diversity of prefrontal cortical populations is strongly dependent on their synaptic targets, with control of motor output mediated by connectivity to dorsal striatum. Despite evidence for functional diversity along the anterior-posterior striatal axis, it is unclear how distinct fronto-striatal sub-circuits support value-based choice. Here we found segregated prefrontal populations defined by anterior/posterior dorsomedial striatal target. During a feedback-based 2-alternative choice task, single-photon imaging revealed circuit-specific representations of task-relevant information with prelimbic neurons targeting anterior DMS (PL::A-DMS) robustly modulated during choices and negative outcomes, while prelimbic neurons targeting posterior DMS (PL::P-DMS) encoded internal representations of value and positive outcomes contingent on prior choice. Consistent with this distributed coding, optogenetic inhibition of PL::A-DMS circuits strongly impacted choice monitoring and responses to negative outcomes while inhibition of PL::P-DMS impaired task engagement and strategies following positive outcomes. Together our data uncover PL populations engaged in distributed processing for value-based choice.


Subject(s)
Corpus Striatum , Neostriatum , Mice , Male , Animals , Corpus Striatum/physiology , Prefrontal Cortex/physiology , Inhibition, Psychological
3.
Trends Neurosci ; 45(9): 649-650, 2022 09.
Article in English | MEDLINE | ID: mdl-35690496

ABSTRACT

In a recent study, Bolkan, Stone, and colleagues demonstrated that direct and indirect striatal pathways in mice exert opponent control over choice behavior in a task- and state-dependent manner. This work highlights the need for rigorously controlled behavioral experiments and novel behavioral modeling in investigations of the neural mechanisms of decision making.


Subject(s)
Cognition , Corpus Striatum , Animals , Choice Behavior , Decision Making , Mice , Neural Pathways
4.
Curr Biol ; 31(18): 4139-4147.e6, 2021 09 27.
Article in English | MEDLINE | ID: mdl-34302742

ABSTRACT

The dorsomedial striatum (DMS) is a central hub supporting goal-directed learning and motor performance. Recent evidence has revealed unexpected roles for local inhibitory GABAergic networks in modulating striatal output and behavior.1 The sparse low-threshold spiking interneuron subtype (LTSI), which exhibits robust reward-circumscribed population activity, is a bidirectional regulator of initial goal-directed learning.2 Striatal dopamine signaling is a central reward-related neuromodulatory system mediating goal-directed action and performance, serving as a teaching signal,3 facilitating synaptic plasticity,4 and invigorating motor behaviors.5 Given the dynamic modulation of LTSIs during goal-directed behavior, we hypothesized that they could provide a novel GABAergic mechanism of local striatal dopaminergic regulation to shape early learning. We provide anatomical evidence for close proximation of LTSI terminals and dopaminergic processes in striatum, suggesting that LTSIs directly control dopaminergic axon activity. Using in vitro fast scan cyclic voltammetry, we demonstrate that LTSIs directly attenuate optogenetically evoked dopamine via GABAB receptor signaling. In vivo, GRABDA dopamine sensor imaging shows that LTSIs strongly modulate striatal dopamine dynamics during operant learning, while pharmacological stabilization of dopamine via intra-striatal aripiprazole microinjection suppresses the effects of LTSI inhibition on learning. Together, these results uncover an unexpected function for LTSIs in gating striatal dopamine to facilitate goal-directed learning.


Subject(s)
Corpus Striatum , Dopamine , Corpus Striatum/physiology , Dopamine/physiology , Interneurons/physiology , Learning/physiology , Reward
5.
Neuron ; 103(1): 92-101.e6, 2019 07 03.
Article in English | MEDLINE | ID: mdl-31097361

ABSTRACT

The dorsomedial striatum (DMS) is critically involved in motor control and reward processing, but the specific neural circuit mediators are poorly understood. Recent evidence highlights the extensive connectivity of low-threshold spiking interneurons (LTSIs) within local striatal circuitry; however, the in vivo function of LTSIs remains largely unexplored. We employed fiber photometry to assess LTSI calcium activity in a range of DMS-mediated behaviors, uncovering specific reward-related activity that is down-modulated during goal-directed learning. Using two mechanistically distinct manipulations, we demonstrated that this down-modulation of LTSI activity is critical for acquisition of novel contingencies, but not for their modification. In contrast, continued LTSI activation slowed instrumental learning. Similar manipulations of fast-spiking interneurons did not reproduce these effects, implying a specific function of LTSIs. Finally, we revealed a role for the γ-aminobutyric acid (GABA)ergic functions of LTSIs in learning. Together, our data provide new insights into this striatal interneuron subclass as important gatekeepers of goal-directed learning.


Subject(s)
Corpus Striatum/physiology , Goals , Interneurons/physiology , Learning/physiology , Animals , Appetite , Calcium Signaling/physiology , Conditioning, Operant/physiology , Electrophysiological Phenomena/physiology , Mice , Mice, Knockout , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels, Inwardly Rectifying/physiology , Reward , Vesicular Inhibitory Amino Acid Transport Proteins/genetics , Vesicular Inhibitory Amino Acid Transport Proteins/physiology , gamma-Aminobutyric Acid/physiology
6.
Eur J Neurosci ; 49(5): 623-636, 2019 03.
Article in English | MEDLINE | ID: mdl-29359830

ABSTRACT

The dorsomedial striatum, a key site of reward-sensitive motor output, receives extensive afferent input from cortex, thalamus and midbrain. These projections are integrated by striatal microcircuits containing both spiny projection neurons and local circuit interneurons. To explore target cell specificity of these projections, we compared inputs onto D1-dopamine receptor-positive spiny neurons, parvalbumin-positive fast-spiking interneurons and somatostatin-positive low-threshold-spiking interneurons, using cell type-specific rabies virus tracing and optogenetic-mediated projection neuron recruitment in mice. While the relative proportion of retrogradely labelled projection neurons was similar between target cell types, the convergence of inputs was systematically higher for projections onto fast-spiking interneurons. Rabies virus is frequently used to assess cell-specific anatomical connectivity but it is unclear how this correlates to synaptic connectivity and efficacy. To test this, we compared tracing data with target cell-specific measures of synaptic efficacy for anterior cingulate cortex and parafascicular thalamic projections using novel quantitative optogenetic measures. We found that target-specific patterns of convergence were extensively modified according to region of projection neuron origin and postsynaptic cell type. Furthermore, we observed significant divergence between cell type-specific anatomical connectivity and measures of excitatory synaptic strength, particularly for low-threshold-spiking interneurons. Taken together, this suggests a basic uniform connectivity map for striatal afferent inputs upon which presynaptic-postsynaptic interactions impose substantial diversity of physiological connectivity.


Subject(s)
Gyrus Cinguli/physiology , Interneurons/physiology , Neostriatum/physiology , Nerve Net/physiology , Neurons, Afferent/physiology , Thalamus/physiology , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Optogenetics
7.
Psychopharmacology (Berl) ; 233(17): 3173-86, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27376946

ABSTRACT

RATIONALE: Intermittent social defeat stress engenders persistent neuroadaptations and can result in later increased cocaine taking and seeking. However, there are individual differences in stress-escalated cocaine self-administration behavior, which may be a direct result of individual differences in the manner in which rats experience social defeat stress. OBJECTIVE: The present study dissected the discrete behavioral phases of social defeat and analyzed which behavioral characteristics may be predictive of subsequent cocaine self-administration. METHODS: Male Long-Evans rats underwent nine intermittent social defeat episodes over 21 days in a three-compartment apparatus permitting approach to and escape from a confrontation with an aggressive resident rat. Rats then self-administered intravenous cocaine, which culminated in a 24-h unlimited access "binge." Behaviors during social defeat and cocaine self-administration were evaluated by principal component analysis (PCA). RESULTS: PCA revealed that the latency to enter the threatening environment was highly predictive of later cocaine self-administration during the 24-h binge. This behavior was not associated with other cocaine-predictive traits, such as reactivity to novelty in an open field, saccharin preference, and motor impulsivity. Additionally, there was no effect of latency to enter a threatening environment on physiological measures of stress, including plasma corticosterone and corticotropin releasing factor (CRF) in the extended amygdala. However, latency to enter the threatening environment was negatively correlated with brain-derived neurotropic factor (BDNF) and its receptor, tyrosine kinase B (TrkB) in the hippocampus. CONCLUSION: These data suggest that latency to enter a threatening environment is a novel behavioral characteristic predictive of later cocaine self-administration.


Subject(s)
Behavior, Animal , Choice Behavior , Cocaine/administration & dosage , Dopamine Uptake Inhibitors/administration & dosage , Social Behavior , Stress, Psychological/psychology , Aggression , Animals , Cocaine-Related Disorders , Corticosterone/metabolism , Corticotropin-Releasing Hormone/metabolism , Individuality , Male , Principal Component Analysis , Rats , Rats, Long-Evans , Saccharin/administration & dosage , Self Administration , Stress, Psychological/metabolism , Sweetening Agents/administration & dosage
8.
J Neurosci ; 36(14): 4093-105, 2016 Apr 06.
Article in English | MEDLINE | ID: mdl-27053215

ABSTRACT

Intermittent social defeat stress escalates later cocaine self-administration. Reward and stress both activate ventral tegmental area (VTA) dopamine neurons, increasing downstream extracellular dopamine concentration in the medial prefrontal cortex and nucleus accumbens. The stress neuropeptide corticotropin releasing factor (CRF) and its receptors (CRF-R1, CRF-R2) are located in the VTA and influence dopaminergic activity. These experiments explore how CRF release and the activation of its receptors within the VTA both during and after stress influence later cocaine self-administration in rats.In vivo microdialysis of CRF in the VTA demonstrated that CRF is phasically released in the posterior VTA (pVTA) during acute defeat, but, with repeated defeat, CRF is recruited into the anterior VTA (aVTA) and CRF tone is increased in both subregions. Intra-VTA antagonism of CRF-R1 in the pVTA and CRF-R2 in the aVTA during each social defeat prevented escalated cocaine self-administration in a 24 h "binge." VTA CRF continues to influence cocaine seeking in stressed animals long after social defeat exposure. Unlike nonstressed controls, previously stressed rats show significant cocaine seeking after 15 d of forced abstinence. Previously stressed rats continue to express elevated CRF tone within the VTA and antagonism of pVTA CRF-R1 or aVTA CRF-R2 reverses cocaine seeking. In conclusion, these experiments demonstrate neuroadaptive changes in tonic and phasic CRF with repeated stress, that CRF release during stress may contribute to later escalated cocaine taking, and that persistently elevated CRF tone in the VTA may drive later cocaine seeking through increased activation of pVTA CRF-R1 and aVTA CRF-R2. SIGNIFICANCE STATEMENT: Corticotropin releasing factor (CRF) within the ventral tegmental area (VTA) has emerged as a likely candidate molecule underlying the fundamental link between stress history and escalated drug self-administration. However, the nature of CRF release in the VTA during acute and repeated stress, as well as its role in enduring neuroadaptations driving later drug taking and seeking, are poorly understood. These experiments explore how CRF is released and interacts with its receptors in specific regions of the VTA both during and after stress to fuel later escalated cocaine taking and seeking behavior. Understanding these acute and persistent changes to the VTA CRF system may lead to better therapeutic interventions for addiction.


Subject(s)
Cocaine-Related Disorders/metabolism , Cocaine-Related Disorders/psychology , Corticotropin-Releasing Hormone/metabolism , Social Environment , Stress, Psychological/metabolism , Stress, Psychological/psychology , Ventral Tegmental Area/metabolism , Animals , Drug-Seeking Behavior , Male , Microdialysis , Rats , Rats, Long-Evans , Receptors, Corticotropin-Releasing Hormone/metabolism , Self Administration , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/psychology
9.
Psychopharmacology (Berl) ; 233(4): 681-90, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26576941

ABSTRACT

RATIONALE: Excessive alcohol (EtOH) drinking is difficult to model in animals despite the extensive human literature demonstrating that stress increases EtOH consumption. OBJECTIVE: The current experiments show escalations in voluntary EtOH drinking caused by a history of social defeat stress and intermittent access to EtOH in C57BL/6J mice compared to non-stressed mice given intermittent EtOH or continuous EtOH. To explore a mechanistic link between stress and drinking, we studied the role of corticotropin-releasing factor type-1 receptors (CRF-R1) in the dopamine-rich ventral tegmental area (VTA). RESULTS: Intra-VTA infusions of a CRF-R1 antagonist, CP376395, infused into the VTA dose-dependently and selectively reduced intermittent EtOH intake in stressed and non-stressed mice, but not in mice given continuous EtOH. In contrast, intra-VTA infusions of the CRF-R2 antagonist astressin2B non-specifically suppressed both EtOH and H2O drinking in the stressed group without effects in the non-stressed mice. Using in vivo microdialysis in the nucleus accumbens (NAc) shell, we observed that stressed mice drinking EtOH intermittently had elevated levels of tonic dopamine concentrations compared to non-stressed drinking mice. Also, VTA CP376395 potentiated dopamine output to the NAc only in the stressed group causing further elevations of dopamine post-infusion. CONCLUSIONS: These findings illustrate a role for extrahypothalamic CRF-R1 as especially important for stress-escalated EtOH drinking beyond schedule-escalated EtOH drinking. CRF-R1 may be a mechanism for balancing the dysregulation of stress and reward in alcohol use disorders.


Subject(s)
Alcohol Drinking/metabolism , Ethanol/administration & dosage , Nucleus Accumbens/metabolism , Receptors, Corticotropin-Releasing Hormone/physiology , Stress, Psychological/metabolism , Ventral Tegmental Area/metabolism , Alcohol Drinking/psychology , Animals , Corticotropin-Releasing Hormone/administration & dosage , Dopamine/metabolism , Interpersonal Relations , Male , Mice , Mice, Inbred C57BL , Microdialysis , Microinjections , Nucleus Accumbens/drug effects , Peptide Fragments/administration & dosage , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Reward , Stress, Psychological/psychology , Ventral Tegmental Area/drug effects
10.
Psychopharmacology (Berl) ; 233(2): 163-86, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26676983

ABSTRACT

Aversive events rapidly and potently excite certain dopamine neurons in the ventral tegmental area (VTA), promoting phasic increases in the medial prefrontal cortex and nucleus accumbens. This is in apparent contradiction to a wealth of literature demonstrating that most VTA dopamine neurons are strongly activated by reward and reward-predictive cues while inhibited by aversive stimuli. How can these divergent processes both be mediated by VTA dopamine neurons? The answer may lie within the functional and anatomical heterogeneity of the VTA. We focus on VTA heterogeneity in anatomy, neurochemistry, electrophysiology, and afferent/efferent connectivity. Second, recent evidence for a critical role of VTA dopamine neurons in response to both acute and repeated stress will be discussed. Understanding which dopamine neurons are activated by stress, the neural mechanisms driving the activation, and where these neurons project will provide valuable insight into how stress can promote psychiatric disorders associated with the dopamine system, such as addiction and depression.


Subject(s)
Dopamine/metabolism , Dopaminergic Neurons , Stress, Psychological/metabolism , Stress, Psychological/physiopathology , Ventral Tegmental Area/metabolism , Ventral Tegmental Area/physiopathology , Animals , Humans
12.
Psychopharmacology (Berl) ; 232(24): 4469-79, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26403083

ABSTRACT

RATIONALE: Stress activates a subset of dopamine neurons in the ventral tegmental area (VTA), increasing extracellular dopamine in the medial prefrontal cortex (mPFC) and nucleus accumbens shell (NAcSh). The stress neuropeptide corticotropin releasing factor (CRF) and its receptors (CRF-R1 and CRF-R2) are located within the VTA and directly and indirectly influence dopaminergic activity. However, it has yet to be shown in vivo whether VTA CRF receptor activation is necessary for acute and repeated stress-induced dopamine efflux. OBJECTIVE: With intra-VTA CRF-R1 and CRF-R2 antagonism during social defeat, we assessed whether blockade of these receptors could prevent stress-induced dopamine increases in the mPFC and NAcSh using in vivo microdialysis. METHODS: Rats were microinjected with a CRF-R1 or CRF-R2 antagonist into the VTA prior to social defeat stress on days 1, 4, 7, and 10. In vivo microdialysis for dopamine in the mPFC and NAcSh was performed during stress on days 1 and 10. RESULTS: During the first social defeat, extracellular dopamine was significantly elevated in both the mPFC and NAcSh, and this increase in the NAcSh was blocked by intra-VTA CRF-R2, but not CRF-R1, antagonism. During the final social defeat, the dopaminergic increase was neither sensitized nor habituated in the mPFC and NAcSh, and intra-VTA CRF-R2, but not CRF-R1, antagonism prevented the dopamine increase in both brain regions. CONCLUSION: These findings show that CRF-R2 in the VTA is necessary for acute and repeated stress-induced dopamine efflux in the NAcSh, but is only recruited into mPFC-projecting dopamine neurons with repeated stress exposure.


Subject(s)
Dopamine/metabolism , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Social Dominance , Stress, Psychological/metabolism , Ventral Tegmental Area/metabolism , Animals , Corticotropin-Releasing Hormone/metabolism , Male , Microdialysis , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Rats , Rats, Long-Evans , Ventral Tegmental Area/drug effects
13.
Psychopharmacology (Berl) ; 232(16): 2889-902, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25899790

ABSTRACT

RATIONALE: Disrupted social behavior, including occasional aggressive outbursts, is characteristic of withdrawal from long-term alcohol (EtOH) use. Heavy EtOH use and exaggerated responses during withdrawal may be treated using glutamatergic N-methyl-D-aspartate receptor (NMDAR) antagonists. OBJECTIVES: The current experiments explore aggression and medial prefrontal cortex (mPFC) glutamate as consequences of withdrawal from intermittent access to EtOH and changes in aggression and mPFC glutamate caused by NMDAR antagonists memantine and ketamine. METHODS: Swiss male mice underwent withdrawal following 1-8 weeks of intermittent access to 20 % EtOH. Aggressive and nonaggressive behaviors with a conspecific were measured 6-8 h into EtOH withdrawal after memantine or ketamine (0-30 mg/kg, i.p.) administration. In separate mice, extracellular mPFC glutamate after memantine was measured during withdrawal using in vivo microdialysis. RESULTS: At 6-8 h withdrawal from EtOH, mice exhibited more convulsions and aggression and decreased social contact compared to age-matched water controls. Memantine, but not ketamine, increased withdrawal aggression at the 5-mg/kg dose in mice with a history of 8 weeks of EtOH but not 1 or 4 weeks of EtOH or in water drinkers. Tonic mPFC glutamate was higher during withdrawal after 8 weeks of EtOH compared to 1 week of EtOH or 8 weeks of water. Five milligrams per kilogram of memantine increased glutamate in 8-week EtOH mice, but also in 1-week EtOH and water drinkers. CONCLUSIONS: These studies reveal aggressive behavior as a novel symptom of EtOH withdrawal in outbred mice and confirm a role of NMDARs during withdrawal aggression and for disrupted social behavior.


Subject(s)
Aggression/drug effects , Behavior, Animal/drug effects , Ethanol/adverse effects , Glutamic Acid/metabolism , Prefrontal Cortex/drug effects , Substance Withdrawal Syndrome/metabolism , Aggression/physiology , Animals , Excitatory Amino Acid Antagonists/pharmacology , Ketamine/pharmacology , Male , Memantine/pharmacology , Mice , Prefrontal Cortex/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Social Behavior , Substance Withdrawal Syndrome/psychology
14.
Psychopharmacology (Berl) ; 232(4): 825-34, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25178816

ABSTRACT

RATIONALE: Women are twice as likely as men to develop major depressive disorder. Exposure to chronic stress can induce depression in some vulnerable individuals, while others are resistant to depressive-like symptoms after equivalent levels of chronic stress. OBJECTIVES: In female rats, individual differences in saccharin intake during chronic social defeat stress may predict subsequent cocaine self-administration, and may be attributed to alterations in mesolimbic dopamine activity. METHODS: Female rats were exposed to 21 days of chronic social defeat stress, during which they were evaluated for their anhedonia-like responses in the form of saccharin intake. After chronic social defeat stress, the rats were tested for behavioral cross-sensitization to cocaine and escalated cocaine self-administration in a 24-h "binge." A separate group of animals underwent in vivo microdialysis of the nucleus accumbens (NAc) shell to assess dopamine (DA) in response to acute cocaine challenge. RESULTS: Cluster analysis revealed two phenotypes among the stressed female rats based on their saccharin intake while being exposed to stress, termed stress-resistant (SR, 28 %) and stress-sensitive (SS, 72 %). The amount of cocaine self-administered during the 24-h "binge" was positively correlated with preceding saccharin intake. The NAc DA response to a cocaine challenge was significantly lower in SR rats than in the SS and non-stressed control rats. No other significant differences were observed in behavioral cross-sensitization or cocaine self-administration prior to the "binge." CONCLUSION: Female rats showed individual differences in their anhedonic-like response to chronic social defeat stress, and these differences were reliably associated with subsequent cocaine-taking behavior.


Subject(s)
Cocaine/administration & dosage , Depressive Disorder, Major/metabolism , Nucleus Accumbens/metabolism , Saccharin/administration & dosage , Stress, Psychological/metabolism , Animals , Cocaine-Related Disorders/metabolism , Cocaine-Related Disorders/psychology , Depressive Disorder, Major/psychology , Dopamine/metabolism , Female , Individuality , Male , Microdialysis , Nucleus Accumbens/chemistry , Nucleus Accumbens/drug effects , Random Allocation , Rats , Rats, Long-Evans , Self Administration , Sex Factors , Social Behavior , Stress, Psychological/psychology
15.
Psychopharmacology (Berl) ; 232(9): 1555-69, 2015 May.
Article in English | MEDLINE | ID: mdl-25373870

ABSTRACT

RATIONALE: Intermittent social defeat stress can induce neuroadaptations that promote compulsive drug taking. Within the mesocorticolimbic circuit, repeated cocaine administration activates extracellular signal-regulated kinase (ERK). OBJECTIVE: The present experiments examine whether changes in ERK phosphorylation are necessary for the behavioral and neural adaptations that occur as a consequence of intermittent defeat stress. MATERIALS AND METHODS: Rats were exposed to four brief intermittent defeats over the course of 10 days. Ten days after the last defeat, rats were challenged with cocaine (10 mg/kg, i.p.) or saline, and ERK activity was examined in mesocorticolimbic regions. To determine the role of ERK in defeat stress-induced behavioral sensitization, we bilaterally microinjected the MAPK/ERK kinase inhibitor U0126 (1 µg/side) or vehicle (20 % DMSO) into the ventral tegmental area (VTA) prior to each of four defeats. Ten days following the last defeat, locomotor activity was assessed for the expression of behavioral cross-sensitization to cocaine (10 mg/kg, i.p.). Thereafter, rats self-administered cocaine under fixed and progressive ratio schedules of reinforcement, including a 24-h continuous access "binge" (0.3 mg/kg/infusion). RESULTS: We found that repeated defeat stress increased ERK phosphorylation in the VTA. Inhibition of VTA ERK prior to each social defeat attenuated the development of stress-induced sensitization and prevented stress-induced enhancement of cocaine self-administration during a continuous access binge. CONCLUSIONS: These results suggest that enhanced activation of ERK in the VTA due to brief defeats is critical in the induction of sensitization and escalated cocaine taking.


Subject(s)
Cocaine/administration & dosage , Dopamine Uptake Inhibitors/administration & dosage , Extracellular Signal-Regulated MAP Kinases/metabolism , Stress, Psychological/metabolism , Ventral Tegmental Area/metabolism , Animals , Hierarchy, Social , Male , Mitogen-Activated Protein Kinases/metabolism , Rats , Rats, Long-Evans , Reinforcement, Psychology , Self Administration , Signal Transduction/drug effects , Signal Transduction/physiology , Ventral Tegmental Area/drug effects
16.
J Neurosci ; 34(19): 6659-67, 2014 May 07.
Article in English | MEDLINE | ID: mdl-24806691

ABSTRACT

The nature of neuroadaptations in the genesis of escalated cocaine taking remains a topic of considerable interest. Intermittent social defeat stress induces both locomotor and dopaminergic cross-sensitization to cocaine, as well as escalated cocaine self-administration. The current study examines the role of corticotropin releasing factor receptor subtypes 1 and 2 (CRFR1, CRFR2) within the ventral tegmental area (VTA) during social defeat stress. This study investigated whether injecting either a CRFR1 or CRFR2 antagonist directly into the VTA before each social defeat would prevent the development of later (1) locomotor sensitization, (2) dopaminergic sensitization, and (3) escalated cocaine self-administration in rats. CRFR1 antagonist CP376395 (50 or 500 ng/side), CRFR2 antagonist Astressin2-B (100 or 1000 ng/side), or vehicle (aCSF) was microinjected into the VTA 20 min before social defeat stress (or handling) on days 1, 4, 7, and 10. Ten days later, rats were injected with cocaine (10 mg/kg, i.p.) and assessed for either locomotor sensitization, measured by walking activity, or dopaminergic sensitization, measured by extracellular dopamine (DA) in the nucleus accumbens shell (NAcSh) through in vivo microdialysis. Locomotor sensitization testing was followed by intravenous cocaine self-administration. Intra-VTA antagonism of CRFR1, but not CRFR2, inhibited the induction of locomotor cross-sensitization to cocaine, whereas both prevented dopaminergic cross-sensitization and escalated cocaine self-administration during a 24 h "binge." This may suggest dissociation between locomotor sensitization and cocaine taking. These data also suggest that interactions between CRF and VTA DA neurons projecting to the NAcSh are essential for the development of dopaminergic cross-sensitization to cocaine.


Subject(s)
Cocaine-Related Disorders/psychology , Cocaine/pharmacology , Corticotropin-Releasing Hormone/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Dopamine/pharmacology , Social Environment , Stress, Psychological/psychology , Ventral Tegmental Area/drug effects , Aminopyridines/pharmacology , Animals , Behavior, Animal/drug effects , Cocaine/administration & dosage , Dopamine Uptake Inhibitors/administration & dosage , Male , Microdialysis , Microinjections , Motor Activity/drug effects , Motor Activity/physiology , Peptide Fragments/pharmacology , Rats , Rats, Long-Evans , Receptors, Corticotropin-Releasing Hormone/drug effects , Self Administration , Social Behavior , Substance Abuse, Intravenous
17.
Psychopharmacology (Berl) ; 224(1): 179-88, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22926005

ABSTRACT

RATIONALE: Episodic social defeat stress results in cross-sensitization to cocaine, characterized by augmentation of locomotor activity, dopamine (DA) levels in the nucleus accumbens (NAc), and cocaine self-administration during a 24-h "binge" in male rats. However, females are more vulnerable than males at each phase of cocaine addiction, and while these sex differences have been replicated in rats, the role of social stress in females remains largely neglected. OBJECTIVE: This study examined sex and estrous cycle differences in behavioral and dopaminergic cross-sensitization to cocaine, as well as cocaine taking in an unlimited-access self-administration "binge." METHODS: Long-Evans rats underwent episodic social defeat and were assessed 10 days later for either (1) behavioral sensitization, as determined by locomotor activity in response to acute cocaine (10 mg/kg, i.p.), (2) neural sensitization, as determined by in vivo microdialysis of DA in the NAc shell in response to acute cocaine, or (3) intravenous self-administration of cocaine (0.3 mg/kg/infusion) in an unlimited-access "binge." RESULTS: Social defeat stress resulted in behavioral and dopaminergic cross-sensitization in both sexes, but the effect was larger and longer lasting in stressed females. Furthermore, while stress engendered a longer "binge" in both sexes, females had a significantly longer "binge" duration than males. CONCLUSIONS: These data suggest that socially stressed females exhibit a larger and longer lasting behavioral and neural cross-sensitization, as well as more dysregulated cocaine taking, than males possibly due to different alterations in the dopaminergic response in the NAc. Furthermore, estrogens appear to play a facilitatory role in both behavioral and dopaminergic sensitization.


Subject(s)
Cocaine/administration & dosage , Dominance-Subordination , Estrous Cycle/physiology , Stress, Psychological/psychology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Dopamine/metabolism , Estrogens/metabolism , Female , Infusions, Intravenous , Male , Microdialysis , Motor Activity/drug effects , Motor Activity/physiology , Neurons/drug effects , Neurons/metabolism , Nucleus Accumbens/metabolism , Rats , Rats, Long-Evans , Self Administration , Sex Factors , Social Behavior , Time Factors
18.
Psychopharmacology (Berl) ; 218(1): 271-9, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21638221

ABSTRACT

RATIONALE: Women have twice the risk as men to develop depression. Approximately, 24% of major depression disorder cases have comorbid disorders with substance abuse. Several central systems, including dopaminergic and serotonergic pathways, are thought to be involved in such comorbidity. OBJECTIVES: The present study established a chronic social stress model in female rats, which produces some cardinal features of depressive-like symptoms. Further, we examined the effects of acute cocaine on dopamine (DA) and serotonin (5-HT) in the nucleus accumbens (NAc) using this model. METHODS: Female Long-Evans rats confronted a nursing dam in its home cage for 30 min twice daily for 21 days. The non-stressed control group was handled daily throughout the experiment. During the 21 days of stress, behaviors during confrontations, weight, preference for saccharin, and estrous cycles were measured. Ten days after the last confrontation, the experimental rat was challenged with 10 mg/kg of cocaine, and levels of DA and 5-HT in the NAc were measured using in vivo microdialysis. RESULTS: During the course of daily confrontations for 21 days, the experimental females significantly increased the duration of immobility, reduced weight gain and the preference for saccharin, and disrupted estrous cycles during the stress. Chronic social stress significantly attenuated cocaine-induced DA levels, and to some extent, attenuated a percent change of 5-HT compared to the non-stressed control group. CONCLUSIONS: Chronic social defeat stress for 21 days induced physiological and behavioral depression-relevant deficits and blunted response of dopaminergic and to some extent, serotonergic neurons to cocaine challenge in females.


Subject(s)
Cocaine/pharmacology , Depression/physiopathology , Dopamine/metabolism , Serotonin/metabolism , Animals , Behavior, Animal/drug effects , Body Weight , Cocaine/administration & dosage , Disease Models, Animal , Estrous Cycle , Female , Microdialysis , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Rats , Rats, Long-Evans , Saccharin/administration & dosage , Sex Factors
19.
Eur Neuropsychopharmacol ; 21(7): 526-31, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21277173

ABSTRACT

Agonists for neurotensin (NT)-1 receptors have produced antipsychotic-like effects in many animals, including reversal of prepulse inhibition deficits and psychostimulant-induced increases in spontaneous activity. The present study sought to provide a basic assessment of the putative antipsychotic effects of PD149163 in rats using a two way conditioned avoidance response task, which is highly validated for screening antipsychotic drugs, and an inclined grid assessment, which is used to assess extrapyramidal side effect liability. PD149163 (0.0625-8.0 mg/kg) significantly suppressed conditioned avoidance responding (CAR) following administration of a 1.0 or 8.0 mg/kg dose. PD149163 failed to significantly increase catalepsy scores. The typical antipsychotic drug haloperidol (0.01-1.0 mg/kg) significantly suppressed CAR at a 0.1, 0.3, and 1.0 mg/kg dose, and a significant increase in catalepsy scores was found at the 1.0 mg/kg dose. The atypical antipsychotic drug clozapine (2.5-10.0 mg/kg) also produced a significant inhibition of CAR, which occurred following administration of a 10.0 mg/kg dose. Clozapine failed to significantly increase catalepsy scores. Finally, D-amphetamine (1.0 mg/kg), serving as a negative control, failed to suppress CAR or increase catalepsy scores. These data further suggest that PD149163 may have atypical antipsychotic-like properties.


Subject(s)
Antipsychotic Agents/pharmacology , Catalepsy/chemically induced , Neurotensin/analogs & derivatives , Receptors, Neurotensin/agonists , Animals , Antipsychotic Agents/toxicity , Avoidance Learning/drug effects , Central Nervous System Stimulants/pharmacology , Central Nervous System Stimulants/toxicity , Clozapine/pharmacology , Clozapine/toxicity , Conditioning, Classical/drug effects , Dextroamphetamine/pharmacology , Dextroamphetamine/toxicity , Drug Evaluation, Preclinical , Haloperidol/adverse effects , Haloperidol/pharmacology , Haloperidol/toxicity , Male , Neurotensin/pharmacology , Neurotensin/toxicity , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...