Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters











Publication year range
1.
Neuropharmacology ; 196: 108695, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34233202

ABSTRACT

Modifications in brain regions that govern reward-seeking are thought to contribute to persistent behaviors that are heavily associated with alcohol-use disorder (AUD) including binge ethanol drinking. The bed nucleus of the stria terminalis (BNST) is a critical node linked to both alcohol consumption and the onset, maintenance and progression of adaptive anxiety and stress-related disorders. Differences in anatomy, connectivity and receptor subpopulations, make the BNST a sexually dimorphic region. Previous work indicates that the ventral BNST (vBNST) receives input from the insular cortex (IC), a brain region involved in processing the body's internal state. This IC-vBNST projection has also been implicated in emotional and reward-seeking processes. Therefore, we examined the functional properties of vBNST-projecting, IC neurons in male and female mice that have undergone short-term ethanol exposure and abstinence using a voluntary Drinking in the Dark paradigm (DID) paired with whole-cell slice electrophysiology. First we show that IC neurons projected predominantly to the vBNST. Next, our data show that short-term ethanol exposure and abstinence enhanced excitatory synaptic strength onto vBNST-projecting, IC neurons in both sexes. However, we observed diametrically opposing modifications in excitability across sexes. In particular, short-term ethanol exposure resulted in increased intrinsic excitability of vBNST-projecting, IC neurons in females but not in males. Furthermore, in females, abstinence decreased the excitability of these same neurons. Taken together these findings show that short-term ethanol exposure, as well as the abstinence cause sex-related adaptations in BNST-projecting, IC neurons.


Subject(s)
Binge Drinking/metabolism , Insular Cortex/metabolism , Neuronal Plasticity/physiology , Neurons/metabolism , Septal Nuclei/metabolism , Alcohol Abstinence , Animals , Binge Drinking/physiopathology , Central Nervous System Depressants/administration & dosage , Central Nervous System Depressants/pharmacology , Ethanol/administration & dosage , Ethanol/pharmacology , Female , Insular Cortex/physiopathology , Male , Mice , Neural Pathways , Neurons/physiology , Patch-Clamp Techniques , Septal Nuclei/physiopathology , Sex Characteristics , Sex Factors
2.
Neuropharmacology ; 189: 108527, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33741403

ABSTRACT

Binge ethanol drinking is an increasingly problematic component of alcohol use disorder costing the United States approximately over $150 billion every year and causes progressive neuroplasticity alterations in numerous brain regions. However, the precise nature or machinery that underlies binge drinking has not yet been elucidated. Corticotropin releasing factor (CRF) neurons in the central amygdala (CeA) are thought to modulate binge drinking, but the specific circuit mechanisms remain poorly understood. Here, we combined optogenetics with in vivo electrophysiology to identify and record from CeA CRF neurons in mice during a repeated binge ethanol drinking task. First, we found that CeA CRF neurons were more active than CeA non-CRF cells during our binge drinking paradigm. We also observed that CeA CRF neurons displayed a heterogeneous spectrum of responses to a lick of ethanol including, pre-lick activated, lick-excited, lick-inhibited, and no response. Interestingly, pre-lick activated CeA CRF neurons exhibited higher frequency and burst firing during binge drinking sessions. Moreover, their overall tonic and phasic electrical activity enhances over repeated binge drinking sessions. Remarkably, CeA CRF units and pre-lick activated CeA CRF neurons did not show higher firing rate or bursting activity during water and sucrose consumption, suggesting that ethanol may "hijack" or plastically alter their intrinsic excitability. This article is part of the special issue on 'Neurocircuitry Modulating Drug and Alcohol Abuse'.


Subject(s)
Action Potentials/physiology , Binge Drinking/metabolism , Central Amygdaloid Nucleus/metabolism , Corticotropin-Releasing Hormone/metabolism , Ethanol/toxicity , Neurons/metabolism , Action Potentials/drug effects , Alcohol Drinking/adverse effects , Alcohol Drinking/physiopathology , Animals , Binge Drinking/physiopathology , Central Amygdaloid Nucleus/drug effects , Central Amygdaloid Nucleus/physiopathology , Ethanol/administration & dosage , Female , Male , Mice , Mice, Transgenic , Microelectrodes , Neurons/drug effects
3.
Addict Biol ; 26(3): e12961, 2021 05.
Article in English | MEDLINE | ID: mdl-32820590

ABSTRACT

Individuals suffering from substance use disorder often experience relapse events that are attributed to drug craving. Insular cortex (IC) function is implicated in processing drug-predictive cues and is thought to be a critical substrate for drug craving, but the downstream neural circuit effectors of the IC that mediate reward processing are poorly described. Here, we uncover the functional connectivity of an IC projection to the ventral bed nucleus of the stria terminalis (vBNST), a portion of the extended amygdala that has been previously shown to modulate dopaminergic activity within the ventral tegmental area (VTA), and investigate the role of this pathway in reward-related behaviors. We utilized ex vivo slice electrophysiology and in vivo optogenetics to examine the functional connectivity of the IC-vBNST projection and bidirectionally control IC-vBNST terminals in various reward-related behavioral paradigms. We hypothesized that the IC recruits mesolimbic dopamine signaling by activating VTA-projecting, vBNST neurons. Using slice electrophysiology, we found that the IC sends a glutamatergic projection onto vBNST-VTA neurons. Photoactivation of IC-vBNST terminals was sufficient to reinforce behavior in a dopamine-dependent manner. Moreover, silencing the IC-vBNST projection was aversive and resulted in anxiety-like behavior without affecting food consumption. This work provides a potential mechanism by which the IC processes exteroceptive triggers that are predictive of reward.


Subject(s)
Behavior, Animal/physiology , Cerebral Cortex/physiology , Dopamine/metabolism , Septal Nuclei/physiology , Amygdala/physiology , Animals , Anxiety/physiopathology , Female , Humans , Male , Mice, Inbred C57BL , Reward , Ventral Tegmental Area/physiology
4.
ACS Chem Neurosci ; 8(2): 243-251, 2017 02 15.
Article in English | MEDLINE | ID: mdl-27984692

ABSTRACT

The study of neuronal ensembles in awake and behaving animals is a critical question in contemporary neuroscience research. Through the examination of calcium fluctuations, which are correlated with neuronal activity, we are able to better understand complex neural circuits. Recently, the development of technologies including two-photon microscopy, miniature microscopes, and fiber photometry has allowed us to examine calcium activity in behaving subjects over time. Visualizing changes in intracellular calcium in vivo has been accomplished utilizing GCaMP, a genetically encoded calcium indicator. GCaMP allows researchers to tag cell-type specific neurons with engineered fluorescent proteins that alter their levels of fluorescence in response to changes in intracellular calcium concentration. Even with the evolution of GCaMP, in vivo calcium imaging had yet to overcome the limitation of light scattering, which occurs when imaging from neural tissue in deep brain regions. Currently, researchers have created in vivo methods to bypass this problem; this Review will delve into three of these state of the art techniques: (1) two-photon calcium imaging, (2) single photon calcium imaging, and (3) fiber photometry. Here we discuss the advantages and disadvantages of the three techniques. Continued advances in these imaging techniques will provide researchers with unparalleled access to the inner workings of the brain.


Subject(s)
Brain/anatomy & histology , Brain/metabolism , Calcium/metabolism , Nerve Net/metabolism , Animals , Humans , Microscopy, Fluorescence, Multiphoton , Neurons/metabolism , Wakefulness
5.
J Neurosci ; 36(42): 10831-10842, 2016 10 19.
Article in English | MEDLINE | ID: mdl-27798138

ABSTRACT

The neural circuitry underlying mammalian reward behaviors involves several distinct nuclei throughout the brain. It is widely accepted that the midbrain dopamine (DA) neurons are critical for the reward-related behaviors. Recent studies have shown that the centromedial nucleus of the amygdala (CeMA) has a distinct role in regulating reward-related behaviors. However, the CeMA and ventromedial PFC (vmPFC) interaction in reward regulation remains poorly understood. Here, we identify and dissect a GABAergic projection that originates in the CeMA and terminates in the vmPFC (VGat-CreCeMA-vmPFC) using viral-vector-mediated, cell-type-specific optogenetic techniques in mice. Pathway-specific optogenetic activation of the VGat-CreCeMA-vmPFC circuit in awake, behaving animals produced a positive, reward-like phenotype in real-time place preference and increased locomotor activity in open-field testing. In sucrose operant conditioning, the photoactivation of these terminals increased nose-poking effort with no effect on licking behavior and robustly facilitated the extinction of operant behavior. However, photoactivation of these terminals did not induce self-stimulation in the absence of an external reward. The results described here suggest that the VGat-CreCeMA-vmPFC projection acts to modulate existing reward-related behaviors. SIGNIFICANCE STATEMENT: Many studies have shown that the interactions between the centromedial nucleus of the amygdala (CeMA) and ventromedial PFC (vmPFC) have critical roles for emotional regulation. However, most studies have associated this circuit with fear and anxiety behaviors and emphasized top-down processing from vmPFC to CeMA. Here, we provide new evidence for bottom-up CeMA to vmPFC influence on reward-related behaviors. Although previous work implicated the CeMA in incentive salience, our results isolate the investigation to a specific CeMA GABAergic projection to the vmPFC. This long-range GABAergic interaction between amygdala and frontal cortex adds a new dimension to the complex regulation of reward-related behaviors.


Subject(s)
Amygdala/physiology , Behavior, Animal/physiology , Prefrontal Cortex/physiology , Reward , gamma-Aminobutyric Acid/physiology , Animals , Conditioning, Operant/drug effects , Extinction, Psychological , Male , Mice , Motor Activity , Neural Pathways/physiology , Optogenetics , Self Stimulation , Sucrose/pharmacology , Vesicular Inhibitory Amino Acid Transport Proteins/genetics , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism
6.
Neuron ; 83(3): 513-5, 2014 Aug 06.
Article in English | MEDLINE | ID: mdl-25102556

ABSTRACT

Serotonin is an essential neuromodulator, but the precise circuit connectivity that regulates serotonergic neurons has not been well defined. Using rabies virus tracing strategies Weissbourd et al. (2014) and Pollak Dorocic et al. (2014) in this issue of Neuron and Ogawa et al. (2014) in Cell Reports provide a comprehensive map of the inputs to serotonergic neurons, highlighting the complexity and diversity of potential upstream cellular regulators.

7.
Front Behav Neurosci ; 8: 129, 2014.
Article in English | MEDLINE | ID: mdl-24834031

ABSTRACT

The development of excessive fear and/or stress responses to environmental cues such as contexts associated with a traumatic event is a hallmark of post-traumatic stress disorder (PTSD). The basolateral amygdala (BLA) has been implicated as a key structure mediating contextual fear conditioning. In addition, the hippocampus has an integral role in the encoding and processing of contexts associated with strong, salient stimuli such as fear. Given that both the BLA and hippocampus play an important role in the regulation of contextual fear conditioning, examining the functional connectivity between these two structures may elucidate a role for this pathway in the development of PTSD. Here, we used optogenetic strategies to demonstrate that the BLA sends a strong glutamatergic projection to the hippocampal formation through the entorhinal cortex (EC). Next, we photoinhibited glutamatergic fibers from the BLA terminating in the EC during the acquisition or expression of contextual fear conditioning. In mice that received optical inhibition of the BLA-to-EC pathway during the acquisition session, we observed a significant decrease in freezing behavior in a context re-exposure session. In contrast, we observed no differences in freezing behavior in mice that were only photoinhibited during the context re-exposure session. These data demonstrate an important role for the BLA-to-EC glutamatergic pathway in the acquisition of contextual fear conditioning.

8.
J Neurosci ; 34(10): 3699-705, 2014 Mar 05.
Article in English | MEDLINE | ID: mdl-24599468

ABSTRACT

Forming and breaking associations between emotionally salient environmental stimuli and rewarding or aversive outcomes is an essential component of learned adaptive behavior. Importantly, when cue-reward contingencies degrade, animals must exhibit behavioral flexibility to extinguish prior learned associations. Understanding the specific neural circuit mechanisms that operate during the formation and extinction of conditioned behaviors is critical because dysregulation of these neural processes is hypothesized to underlie many of the maladaptive and pathological behaviors observed in various neuropsychiatric disorders in humans. The medial prefrontal cortex (mPFC) participates in the behavioral adaptations seen in both appetitive and aversive-cue-mediated responding, but the precise cell types and circuit mechanisms sufficient for driving these complex behavioral states remain largely unspecified. Here, we recorded and manipulated the activity of parvalbumin-positive fast spiking interneurons (PV+ FSIs) in the prelimbic area (PrL) of the mPFC in mice. In vivo photostimulation of PV+ FSIs resulted in a net inhibition of PrL neurons, providing a circuit blueprint for behavioral manipulations. Photostimulation of mPFC PV+ cells did not alter anticipatory or consummatory licking behavior during reinforced training sessions. However, optical activation of these inhibitory interneurons to cues associated with reward significantly accelerated the extinction of behavior during non-reinforced test sessions. These data suggest that suppression of excitatory mPFC networks via increased activity of PV+ FSIs may enhance reward-related behavioral flexibility.


Subject(s)
Extinction, Psychological/physiology , Interneurons/metabolism , Parvalbumins/metabolism , Prefrontal Cortex/metabolism , Reward , Animals , Interneurons/cytology , Male , Mice , Mice, 129 Strain , Mice, Transgenic , Organ Culture Techniques , Photic Stimulation/methods , Prefrontal Cortex/cytology
9.
Neuropharmacology ; 76 Pt B: 320-8, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23752096

ABSTRACT

Complex motivated behavioral processes, such as those that can go awry following substance abuse and other neuropsychiatric disorders, are mediated by a distributive network of neurons that reside throughout the brain. Neural circuits within the amygdala regions, such as the basolateral amygdala (BLA), and downstream targets such as the bed nucleus of the stria terminalis (BNST), are critical neuroanatomical structures for orchestrating emotional behavioral responses that may influence motivated actions such as the reinstatement of drug seeking behavior. Here, we review the functional neurocircuitry of the BLA and the BNST, and discuss how these circuits may guide maladaptive behavioral processes such as those seen in addiction. Thus, further study of the functional connectivity within these brain regions and others may provide insight for the development of new treatment strategies for substance use disorders. This article is part of a Special Issue entitled 'NIDA 40th Anniversary Issue'.


Subject(s)
Amygdala/pathology , Behavior, Addictive/pathology , Septal Nuclei/pathology , Animals , Humans , Neural Pathways/physiology
10.
J Comp Neurol ; 522(5): 1171-90, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24264880

ABSTRACT

Mutations in phosphatase and tensin homolog deleted on chromosome 10 (PTEN) are implicated in neuropsychiatric disorders including autism. Previous studies report that PTEN knockdown in neurons in vivo leads to increased spine density and synaptic activity. To better characterize synaptic changes in neurons lacking PTEN, we examined the effects of shRNA knockdown of PTEN in basolateral amygdala neurons on synaptic spine density and morphology by using fluorescent dye confocal imaging. Contrary to previous studies in the dentate gyrus, we find that knockdown of PTEN in basolateral amygdala leads to a significant decrease in total spine density in distal dendrites. Curiously, this decreased spine density is associated with increased miniature excitatory postsynaptic current frequency and amplitude, suggesting an increase in number and function of mature spines. These seemingly contradictory findings were reconciled by spine morphology analysis demonstrating increased mushroom spine density and size with correspondingly decreased thin protrusion density at more distal segments. The same analysis of PTEN conditional deletion in the dentate gyrus demonstrated that loss of PTEN does not significantly alter total density of dendritic protrusions in the dentate gyrus, but does decrease thin protrusion density and increases density of more mature mushroom spines. These findings suggest that, contrary to previous reports, PTEN knockdown may not induce de novo spinogenesis, but instead may increase synaptic activity by inducing morphological and functional maturation of spines. Furthermore, behavioral analysis of basolateral amygdala PTEN knockdown suggests that these changes limited only to the basolateral amygdala complex may not be sufficient to induce increased anxiety-related behaviors.


Subject(s)
Amygdala/cytology , Dendritic Spines/physiology , Hippocampus/cytology , Mutation/genetics , Neurons/ultrastructure , PTEN Phosphohydrolase/genetics , Animals , Anxiety/genetics , Behavior, Animal/physiology , Conditioning, Classical/physiology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , In Vitro Techniques , Locomotion/genetics , Mice , Mice, Transgenic , Miniature Postsynaptic Potentials/genetics , Oncogene Protein v-akt/metabolism , Phosphopyruvate Hydratase/metabolism , Reflex, Startle/genetics , Transduction, Genetic
11.
Alcohol Clin Exp Res ; 37(10): 1680-7, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23763790

ABSTRACT

BACKGROUND: Corticotropin releasing factor (CRF) and urocortin play an important role in many stress responses and also can regulate ethanol (EtOH) intake. Adaptations in CRF signaling in the central amygdala promote EtOH consumption after long-term EtOH intake in dependent animals and also after brief periods of binge EtOH intake. Thus, even brief episodes of EtOH consumption can alter the function of the CRF system, allowing CRF to regulate EtOH intake. Here, we examined whether brief binge EtOH consumption leads to CRF receptor adaptations within the ventral tegmental area (VTA), a structure involved in signaling rewarding and aversive events and important in the development and expression of drug and alcohol addiction. METHODS: We utilized a mouse model of binge drinking known as drinking in the dark (DID), where C57BL/6J mice drink approximately 6 g/kg in 4 hours and achieve blood EtOH concentrations of approximately 100 mg/dl, which is equivalent to binge drinking in humans. We used ex vivo whole-cell recordings from putative VTA dopamine (DA) neurons to examine CRF regulation of NMDA receptor (NMDAR) currents. We also examined the impact of CRF receptor antagonist injection in the VTA on binge EtOH intake. RESULTS: Ex vivo whole-cell recordings from putative VTA DA neurons showed enhanced CRF-mediated potentiation of NMDAR currents in juvenile mice that consumed EtOH in the DID procedure. CRF-induced potentiation of NMDAR currents in EtOH-drinking mice was blocked by administration of CP-154,526 (3 µM), a selective CRF1 receptor antagonist. Furthermore, intra-VTA infusion of CP-154,526 (1 µg) significantly reduced binge EtOH consumption in adult mice. These results were not due to alterations of VTA NMDAR number or function, suggesting that binge drinking may enhance signaling through VTA CRF1 receptors onto NMDARs. CONCLUSIONS: Altered CRF1 receptor-mediated signaling in the VTA promotes binge-like EtOH consumption in mice, which supports the idea that CRF1 receptors may therefore be a promising pharmacological target for reducing binge drinking in humans.


Subject(s)
Binge Drinking/metabolism , Darkness , Receptors, Corticotropin-Releasing Hormone/metabolism , Ventral Tegmental Area/metabolism , Animals , Male , Mice , Mice, Inbred C57BL , Organ Culture Techniques
12.
Behav Brain Res ; 255: 19-25, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23684554

ABSTRACT

Optogenetic techniques have given researchers unprecedented access to the function of discrete neural circuit elements and have been instrumental in the identification of novel brain pathways that become dysregulated in neuropsychiatric diseases. For example, stress is integrally linked to the manifestation and pathophysiology of neuropsychiatric illness, including anxiety, addiction and depression. Due to the heterogeneous populations of genetically and neurochemically distinct neurons in areas such as the bed nucleus of the stria terminalis (BNST), as well as their substantial number of projections, our understanding of how neural circuits become disturbed after stress has been limited. Using optogenetic tools, we are now able to selectively isolate distinct neural circuits that contribute to these disorders and perturb these circuits in vivo, which in turn may lead to the normalization of maladaptive behavior. This review will focus on current optogenetic strategies to identify, manipulate, and record from discrete neural circuit elements in vivo as well as highlight recent optogenetic studies that have been utilized to parcel out BNST function.


Subject(s)
Brain Mapping , Optogenetics/methods , Septal Nuclei/physiology , Stress, Psychological/physiopathology , Animals , Neural Pathways/physiology
13.
Nature ; 496(7444): 224-8, 2013 Apr 11.
Article in English | MEDLINE | ID: mdl-23515155

ABSTRACT

The co-morbidity of anxiety and dysfunctional reward processing in illnesses such as addiction and depression suggests that common neural circuitry contributes to these disparate neuropsychiatric symptoms. The extended amygdala, including the bed nucleus of the stria terminalis (BNST), modulates fear and anxiety, but also projects to the ventral tegmental area (VTA), a region implicated in reward and aversion, thus providing a candidate neural substrate for integrating diverse emotional states. However, the precise functional connectivity between distinct BNST projection neurons and their postsynaptic targets in the VTA, as well as the role of this circuit in controlling motivational states, have not been described. Here we record and manipulate the activity of genetically and neurochemically identified VTA-projecting BNST neurons in freely behaving mice. Collectively, aversive stimuli exposure produced heterogeneous firing patterns in VTA-projecting BNST neurons. By contrast, in vivo optically identified glutamatergic projection neurons displayed a net enhancement of activity to aversive stimuli, whereas the firing rate of identified GABAergic (γ-aminobutyric acid-containing) projection neurons was suppressed. Channelrhodopsin-2-assisted circuit mapping revealed that both BNST glutamatergic and GABAergic projections preferentially innervate postsynaptic non-dopaminergic VTA neurons, thus providing a mechanistic framework for in vivo circuit perturbations. In vivo photostimulation of BNST glutamatergic projections resulted in aversive and anxiogenic behavioural phenotypes. Conversely, activation of BNST GABAergic projections produced rewarding and anxiolytic phenotypes, which were also recapitulated by direct inhibition of VTA GABAergic neurons. These data demonstrate that functionally opposing BNST to VTA circuits regulate rewarding and aversive motivational states, and may serve as a crucial circuit node for bidirectionally normalizing maladaptive behaviours.


Subject(s)
Amygdala/physiology , Motivation/physiology , Animals , Anxiety/physiopathology , Avoidance Learning , Behavior, Animal/physiology , Channelrhodopsins , Cues , Electroshock , GABAergic Neurons/metabolism , Glutamine/metabolism , Male , Mice , Mice, Inbred C57BL , Optogenetics , Phenotype , Reward , Septal Nuclei/physiology , Ventral Tegmental Area/physiology
14.
Nat Protoc ; 7(1): 12-23, 2011 Dec 08.
Article in English | MEDLINE | ID: mdl-22157972

ABSTRACT

In vivo optogenetic strategies have redefined our ability to assay how neural circuits govern behavior. Although acutely implanted optical fibers have previously been used in such studies, long-term control over neuronal activity has been largely unachievable. Here we describe a method to construct implantable optical fibers to readily manipulate neural circuit elements with minimal tissue damage or change in light output over time (weeks to months). Implanted optical fibers readily interface with in vivo electrophysiological arrays or electrochemical detection electrodes. The procedure described here, from implant construction to the start of behavioral experimentation, can be completed in approximately 2-6 weeks. Successful use of implantable optical fibers will allow for long-term control of mammalian neural circuits in vivo, which is integral to the study of the neurobiology of behavior.


Subject(s)
Electrophysiology/methods , Implants, Experimental , Nerve Net/physiology , Optical Fibers , Fiber Optic Technology/instrumentation , Fiber Optic Technology/methods
15.
Nature ; 475(7356): 377-80, 2011 Jun 29.
Article in English | MEDLINE | ID: mdl-21716290

ABSTRACT

The basolateral amygdala (BLA) has a crucial role in emotional learning irrespective of valence. The BLA projection to the nucleus accumbens (NAc) is thought to modulate cue-triggered motivated behaviours, but our understanding of the interaction between these two brain regions has been limited by the inability to manipulate neural-circuit elements of this pathway selectively during behaviour. To circumvent this limitation, we used in vivo optogenetic stimulation or inhibition of glutamatergic fibres from the BLA to the NAc, coupled with intracranial pharmacology and ex vivo electrophysiology. Here we show that optical stimulation of the pathway from the BLA to the NAc in mice reinforces behavioural responding to earn additional optical stimulation of these synaptic inputs. Optical stimulation of these glutamatergic fibres required intra-NAc dopamine D1-type receptor signalling, but not D2-type receptor signalling. Brief optical inhibition of fibres from the BLA to the NAc reduced cue-evoked intake of sucrose, demonstrating an important role of this specific pathway in controlling naturally occurring reward-related behaviour. Moreover, although optical stimulation of glutamatergic fibres from the medial prefrontal cortex to the NAc also elicited reliable excitatory synaptic responses, optical self-stimulation behaviour was not observed by activation of this pathway. These data indicate that whereas the BLA is important for processing both positive and negative affect, the glutamatergic pathway from the BLA to the NAc, in conjunction with dopamine signalling in the NAc, promotes motivated behavioural responding. Thus, optogenetic manipulation of anatomically distinct synaptic inputs to the NAc reveals functionally distinct properties of these inputs in controlling reward-seeking behaviours.


Subject(s)
Amygdala/physiology , Excitatory Postsynaptic Potentials/physiology , Neural Pathways/physiology , Nucleus Accumbens/physiology , Reward , Amygdala/cytology , Animals , Behavior, Addictive/physiopathology , Channelrhodopsins , Cues , Dopamine/metabolism , Drinking , Glutamic Acid/metabolism , Light , Male , Mice , Mice, Inbred C57BL , Nerve Fibers/physiology , Neurons/metabolism , Nucleus Accumbens/cytology , Patch-Clamp Techniques , Photic Stimulation , Receptors, Dopamine D1/antagonists & inhibitors , Receptors, Dopamine D1/metabolism , Sucrose/metabolism , Sucrose/pharmacology
16.
ILAR J ; 52(3): 239-50, 2011.
Article in English | MEDLINE | ID: mdl-23382142

ABSTRACT

Much has been written about the interaction of stressors (physical, social, and psychological) and alcohol addiction based on studies in humans and preclinical models. We begin by considering the significance and complexity of alcoholism and the options for effectively modeling it in animals, particularly rodents. We then focus on the following aspects of stress-alcohol interactions: (1) compulsive alcohol consumption, characterized by continued intake despite the presence of stressful or aversive consequences; (2) the possible relationship between acute stress and increased alcohol intake; (3) an apparent cross sensitization of stress and alcohol exposure, which increases both future reactivity to stress and the risk of developing alcohol addiction; and (4) efforts to target stress in therapeutic interventions for alcoholism. We also describe possible neuroadaptations and genetic factors that may interact with stress to increase susceptibility to alcoholism. Throughout, we describe the challenges and inconsistencies inherent in both human and animal studies of alcoholism, its etiology, and its impacts. We believe the relationship between preclinical and human studies is of paramount importance to understand addiction-related behavior in humans and to direct, improve, and expand animal models. It is our hope that a full understanding of the mechanistic bases of pathological alcohol intake will have translational benefits for the development of behavioral and pharmacological therapies.


Subject(s)
Alcohol Drinking , Alcoholism , Alcoholism/genetics , Animals , Behavior, Addictive , Disease Models, Animal , Ethanol , Humans , Models, Animal , Stress, Psychological
17.
Alcohol Clin Exp Res ; 34(9): 1565-73, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20586757

ABSTRACT

BACKGROUND: Continued consumption of alcohol despite deleterious consequences is a hallmark of alcoholism and represents a critical challenge to therapeutic intervention. Previous rat studies showed that enduring alcohol self-administration despite pairing alcohol with normally aversive stimuli was only observed after very long-term intake (>8 months). Aversion-resistant alcohol intake has been previously interpreted to indicate pathological or compulsive motivation to consume alcohol. However, given the time required to model compulsive alcohol seeking in previous studies, there is considerable interest in developing more efficient and quantitative rodent models of aversion-resistant alcohol self-administration. METHODS: Outbred Wistar rats underwent 3 to 4 months or approximately 1.5 months of intermittent, home-cage, two-bottle access (IAA) to 20% alcohol (v/v) or water. Then, after brief operant training, the effect of the bitter-tasting quinine (0.1 g/l) on the motivation to seek alcohol was quantified via progressive ratio (PR). Motivation for quinine-adulterated 2% sucrose under PR was assayed in a separate cohort of 3 to 4 months IAA rats. The effects of quinine on home-cage alcohol consumption in IAA rats and rats with continuous access to alcohol were also examined. Finally, a dose-response for quinine taste preference in IAA and continuous-access animals was determined. RESULTS: Motivation for alcohol after 3 to 4 months IAA, measured using an operant PR procedure, was not altered by adulteration of alcohol with 0.1 g/l quinine. In contrast, after 3 to 4 months of IAA, motivation for sucrose under PR was significantly reduced by adulteration of sucrose with 0.1 g/l quinine. In addition, motivation for alcohol after only approximately 1.5 months IAA was significantly reduced by adulteration of alcohol with 0.1 g/l quinine. Furthermore, home-cage alcohol intake by IAA rats was insensitive to quinine at concentrations (0.01, 0.03 g/l) that significantly reduced alcohol drinking in animals with continuous access to alcohol. Finally, no changes in quinine taste preference after 3 to 4 months IAA or continuous access to alcohol were observed. CONCLUSIONS: We have developed a novel and technically simple hybrid operant/IAA model in which quinine-resistant motivation for alcohol is evident after an experimentally tractable period of time (3 to 4 months vs. 8 months). Quinine dramatically reduced sucrose and water intake by IAA rats, indicating that continued responding for alcohol in IAA rats despite adulteration with the normally aversive quinine might reflect maladaptive or compulsive motivation for alcohol. This model could facilitate identification of novel therapeutic interventions for pathological alcohol seeking in humans.


Subject(s)
Ethanol/administration & dosage , Motivation/drug effects , Quinine/pharmacology , Animals , Choice Behavior/drug effects , Conditioning, Operant/drug effects , Dose-Response Relationship, Drug , Drinking/drug effects , Male , Rats , Rats, Wistar , Self Administration , Sucrose/administration & dosage , Time Factors
18.
Biol Psychiatry ; 65(8): 646-53, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19118821

ABSTRACT

BACKGROUND: Genetic factors and previous alcohol experience influence alcohol consumption in both humans and rodents. Specifically, a prior experience with ethanol increases ethanol intake in both ethanol-preferring C57BL/6 (C57) and ethanol non-preferring DBA/2 (DBA) mice. Whereas the ventral tegmental area (VTA) importantly regulates dopamine levels and ethanol intake, it is unknown whether ethanol experience differentially alters synaptic properties of VTA dopamine neurons in ethanol-preferring and non-preferring mice. METHODS: The properties of excitatory and inhibitory inputs and the ability to elicit long-term potentiation (LTP) were assessed with whole-cell patch-clamp recordings in VTA dopamine neurons from C57 and DBA mice 24 hours after a single ethanol (2 g/kg, IP) or equivalent saline injection. RESULTS: Ethanol exposure increased gamma-aminobutyric acid (GABA) release onto VTA dopamine neurons in DBA mice, as previously observed in C57 mice. However, a single ethanol exposure reduced alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptor (AMPAR) and N-methyl-D-aspartate receptor (NMDAR) function and LTP in VTA dopamine neurons from DBA but not C57 mice. CONCLUSIONS: A single ethanol exposure selectively reduced glutamate receptor function in VTA dopamine neurons from the ethanol non-preferring DBA strain but enhanced GABA signaling in both C57 and DBA strains. These results support the notion that VTA dopamine neurons are a central target of ethanol-induced neural plasticity, which could contribute to ethanol consumption. Furthermore, these findings highlight the possible need for specialized therapeutic interventions for alcoholism based on individual intrinsic differences.


Subject(s)
Dopamine/metabolism , Ethanol/pharmacology , Mice, Inbred C57BL/physiology , Mice, Inbred DBA/physiology , Neuronal Plasticity/drug effects , Presynaptic Terminals/drug effects , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/physiology , Animals , In Vitro Techniques , Long-Term Potentiation/drug effects , Mice , Mice, Inbred C57BL/metabolism , Mice, Inbred DBA/metabolism , Neurons/metabolism , Presynaptic Terminals/physiology , Receptors, AMPA/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Species Specificity , Ventral Tegmental Area/metabolism , gamma-Aminobutyric Acid/metabolism
19.
Alcohol Clin Exp Res ; 33(1): 31-42, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18945225

ABSTRACT

BACKGROUND: The alcohol deprivation effect (ADE) is characterized by transient excessive alcohol consumption upon reinstatement of ethanol following a period of ethanol deprivation. While this phenomenon has been observed in rats using both bottle drinking (consummatory behavior) and operant self-administration (consummatory and appetitive "ethanol-seeking" behavior) procedures, ADE studies in mice have primarily relied on bottle drinking measures. Furthermore, the neurochemical pathways that modulate the ADE are not well understood. Therefore, we determined whether the ADE can be observed in C57BL/6J mice using operant self-administration procedures and if expression of the ADE is modulated by the corticotropin releasing factor-1 (CRF-1) receptor. METHODS: C57BL/6J mice were trained in a 2-hour operant self-administration paradigm to lever press for 10% ethanol or water on separate response keys. Between operant sessions, mice had access to ethanol in their homecage. Once stable responding occurred, mice were deprived of ethanol for 4 days and were then retested with ethanol in the operant paradigm for 3 consecutive days. Next, to assess the role of the CRF-1 receptor, mice were given intraperitoneal (i.p.) injection (0, 10, or 20 mg/kg) of the CRF-1 receptor antagonist CP-154,526 30 minutes before ADE testing. Additional experiments assessed (i) ADE responding in which the alternate response lever was inactive, (ii) the effects of CP-154,526 on self-administration of a 1% sucrose solution following 4 days of deprivation, and (iii) ADE responding in which mice did not received i.p. injections throughout the experiment. RESULTS: Mice exhibited a significant increase in postdeprivation lever responding for ethanol with either a water reinforced or inactive alternate lever. Interestingly, i.p. injection of a 10 mg/kg dose of CP-154,526 protected against the ADE while not affecting lever responding for a sucrose solution. Finally, baseline and deprivation-induced increases of ethanol reinforced lever responding were greater in mice not given i.p. injections. CONCLUSIONS: The ADE in C57BL/6J mice can be modeled using the operant self-administration paradigm and increased ethanol self-administration associated with the ADE is modulated by CRF-1 receptor signaling.


Subject(s)
Conditioning, Operant/physiology , Ethanol/administration & dosage , Receptors, Corticotropin-Releasing Hormone/physiology , Signal Transduction/physiology , Substance Withdrawal Syndrome/physiopathology , Alcoholism/physiopathology , Alcoholism/psychology , Animals , Conditioning, Operant/drug effects , Male , Mice , Mice, Inbred C57BL , Pyrimidines/administration & dosage , Pyrroles/administration & dosage , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Self Administration , Signal Transduction/drug effects , Substance Withdrawal Syndrome/psychology
20.
Alcohol Clin Exp Res ; 32(11): 1962-8, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18782340

ABSTRACT

BACKGROUND: Drinking in the dark (DID) procedures have recently been developed to induce high levels of ethanol drinking in C57BL/6J mice, which result in blood ethanol concentrations reaching levels that have measurable affects on physiology and/or behavior. The present study determined if increased ethanol drinking associated with DID procedures may be motivated by caloric need rather than by the postingestive pharmacological effects of ethanol. To this end, food availability was manipulated or mice were given peripheral administration of orexigenic or anorectic agents during DID procedures. METHODS: C57BL/6J had 2-hours of access to the 20% (v/v) ethanol solution beginning 3-hours into the dark cycle on days 1 to 3, and 4-hours of access to the ethanol bottle on day 4 of DID procedures. In Experiment 1, the effects of food deprivation on ethanol consumption during DID procedures was assessed. In Experiments 2 and 3, mice were given intraperitoneal (i.p.) injection of the orexigenic peptide ghrelin (0, 10 or 30 mg/kg) or the anorectic protein leptin (0 or 20 microg/g), respectively, before access to ethanol on day 4 of DID procedures. In Experiment 4, hourly consumption of food and a 0.05% saccharin solution were assessed over a period of hours that included those used with DID procedures. RESULTS: Consistent with previous research, mice achieved blood ethanol concentrations (BECs) that ranged between 100 and 150 mg% on day 4 of DID experiments. Neither food deprivation nor administration of orexigenic or anorectic compounds significantly altered ethanol drinking with DID procedures. Interestingly, mice exhibited their highest level of food and saccharin solution consumption during hours that overlapped with DID procedures. CONCLUSIONS: The present observations are inconsistent with the hypothesis that C57BL/6J mice consume large amounts of ethanol during DID procedures in order to satisfy a caloric need.


Subject(s)
Alcohol Drinking/physiopathology , Appetite Depressants/pharmacology , Appetite Stimulants/pharmacology , Food Deprivation/physiology , Alcoholism/physiopathology , Animals , Appetite Depressants/administration & dosage , Appetite Stimulants/administration & dosage , Darkness , Disease Models, Animal , Energy Intake/drug effects , Energy Intake/physiology , Ethanol/blood , Feeding Behavior/drug effects , Ghrelin/administration & dosage , Ghrelin/pharmacology , Injections, Intraperitoneal , Leptin/administration & dosage , Leptin/pharmacology , Male , Mice , Mice, Inbred C57BL , Saccharin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL