Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Neurosci ; 39(37): 7394-7407, 2019 09 11.
Article in English | MEDLINE | ID: mdl-31331999

ABSTRACT

Recent studies suggest that the ventral medial prefrontal cortex (vmPFC) encodes both operant drug self-administration and extinction memories. Here, we examined whether these opposing memories are encoded by distinct neuronal ensembles within the vmPFC with different outputs to the nucleus accumbens (NAc) in male and female rats. Using cocaine self-administration (3 h/d for 14 d) and extinction procedures, we demonstrated that vmPFC was similarly activated (indexed by Fos) during cocaine-seeking tests after 0 (no-extinction) or 7 extinction sessions. Selective Daun02 lesioning of the self-administration ensemble (no-extinction) decreased cocaine seeking, whereas Daun02 lesioning of the extinction ensemble increased cocaine seeking. Retrograde tracing with fluorescent cholera toxin subunit B injected into NAc combined with Fos colabeling in vmPFC indicated that vmPFC self-administration ensembles project to NAc core while extinction ensembles project to NAc shell. Functional disconnection experiments (Daun02 lesioning of vmPFC and acute dopamine D1-receptor blockade with SCH39166 in NAc core or shell) confirm that vmPFC ensembles interact with NAc core versus shell to play dissociable roles in cocaine self-administration versus extinction, respectively. Our results demonstrate that neuronal ensembles mediating cocaine self-administration and extinction comingle in vmPFC but have distinct outputs to the NAc core and shell that promote or inhibit cocaine seeking.SIGNIFICANCE STATEMENT Neuronal ensembles within the vmPFC have recently been shown to play a role in self-administration and extinction of food seeking. Here, we used the Daun02 chemogenetic inactivation procedure, which allows selective inhibition of neuronal ensembles identified by the activity marker Fos, to demonstrate that different ensembles for cocaine self-administration and extinction memories coexist in the ventral mPFC and interact with distinct subregions of the nucleus accumbens.


Subject(s)
Cocaine/administration & dosage , Drug-Seeking Behavior/physiology , Extinction, Psychological/physiology , Prefrontal Cortex/drug effects , Prefrontal Cortex/physiology , Animals , Conditioning, Operant/drug effects , Conditioning, Operant/physiology , Dopamine Uptake Inhibitors/administration & dosage , Drug-Seeking Behavior/drug effects , Extinction, Psychological/drug effects , Male , Nerve Net/chemistry , Nerve Net/drug effects , Nerve Net/physiology , Nucleus Accumbens/chemistry , Nucleus Accumbens/drug effects , Nucleus Accumbens/physiology , Prefrontal Cortex/chemistry , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Rats, Transgenic , Self Administration
2.
Neuron ; 102(1): 105-119.e8, 2019 04 03.
Article in English | MEDLINE | ID: mdl-30792150

ABSTRACT

Historically, the rat has been the preferred animal model for behavioral studies. Limitations in genome modification have, however, caused a lag in their use compared to the bevy of available transgenic mice. Here, we have developed several transgenic tools, including viral vectors and transgenic rats, for targeted genome modification in specific adult rat neurons using CRISPR-Cas9 technology. Starting from wild-type rats, knockout of tyrosine hydroxylase was achieved with adeno-associated viral (AAV) vectors expressing Cas9 or guide RNAs (gRNAs). We subsequently created an AAV vector for Cre-dependent gRNA expression as well as three new transgenic rat lines to specifically target CRISPR-Cas9 components to dopaminergic neurons. One rat represents the first knockin rat model made by germline gene targeting in spermatogonial stem cells. The rats described herein serve as a versatile platform for making cell-specific and sequence-specific genome modifications in the adult brain and potentially other Cre-expressing tissues of the rat.


Subject(s)
Adult Germline Stem Cells/metabolism , Brain/metabolism , CRISPR-Cas Systems , Dopaminergic Neurons/metabolism , Gene Editing/methods , Gene Targeting/methods , Animals , CRISPR-Associated Protein 9/genetics , Deoxyribonuclease I/genetics , Dependovirus , Disease Models, Animal , Dopamine Plasma Membrane Transport Proteins/genetics , Gene Knock-In Techniques/methods , Gene Knockout Techniques , Genetic Vectors , Integrases , Luminescent Proteins/genetics , Neurons/metabolism , Promoter Regions, Genetic , RNA, Guide, Kinetoplastida , Rats , Rats, Transgenic , Tyrosine 3-Monooxygenase/genetics , Red Fluorescent Protein
3.
Elife ; 72018 10 03.
Article in English | MEDLINE | ID: mdl-30281020

ABSTRACT

How are decisions made between different goods? One theory spanning several fields of neuroscience proposes that their values are distilled to a single common neural currency, the calculation of which allows for rational decisions. The orbitofrontal cortex (OFC) is thought to play a critical role in this process, based on the presence of neural correlates of economic value in lateral OFC in monkeys and medial OFC in humans. We previously inactivated lateral OFC in rats without affecting economic choice behavior. Here we inactivated medial OFC in the same task, again without effect. Behavior in the same rats was disrupted by inactivation during progressive ratio responding previously shown to depend on medial OFC, demonstrating the efficacy of the inactivation. These results indicate that medial OFC is not necessary for economic choice, bolstering the proposal that classic economic choice is likely mediated by multiple, overlapping neural circuits.


Subject(s)
Choice Behavior/physiology , Neurons/physiology , Prefrontal Cortex/physiology , Psychomotor Performance/physiology , Animals , Cues , Male , Models, Neurological , Optogenetics , Rats, Long-Evans
4.
J Neurosci ; 38(41): 8822-8830, 2018 10 10.
Article in English | MEDLINE | ID: mdl-30181136

ABSTRACT

Prediction errors are critical for associative learning. In the brain, these errors are thought to be signaled, in part, by midbrain dopamine neurons. However, although there is substantial direct evidence that brief increases in the firing of these neurons can mimic positive prediction errors, there is less evidence that brief pauses mimic negative errors. Whereas pauses in the firing of midbrain dopamine neurons can substitute for missing negative prediction errors to drive extinction, it has been suggested that this effect might be attributable to changes in salience rather than the operation of this signal as a negative prediction error. Here we address this concern by showing that the same pattern of inhibition will create a cue able to meet the classic definition of a conditioned inhibitor by showing suppression of responding in a summation test and slower learning in a retardation test. Importantly, these classic criteria were designed to rule out explanations founded on attention or salience; thus the results cannot be explained in this manner. We also show that this pattern of behavior is not produced by a single, prolonged, ramped period of inhibition, suggesting that it is precisely timed, sudden change and not duration that conveys the teaching signal.SIGNIFICANCE STATEMENT Here we show that brief pauses in the firing of midbrain dopamine neurons are sufficient to produce a cue that meets the classic criteria defining a conditioned inhibitor, or a cue that predicts the omission of a reward. These criteria were developed to distinguish actual learning from salience or attentional effects; thus these results formally show that brief pauses in the firing of dopamine neurons can serve as key teaching signals in the brain. Interestingly, this was not true for gradual prolonged pauses, suggesting it is the dynamic change in firing that serves as the teaching signal.


Subject(s)
Conditioning, Classical/physiology , Dopaminergic Neurons/physiology , Reward , Ventral Tegmental Area/physiology , Action Potentials , Animals , Attention/physiology , Behavior, Animal , Female , Male , Rats, Transgenic
5.
Learn Mem ; 25(9): 455-460, 2018 09.
Article in English | MEDLINE | ID: mdl-30115767

ABSTRACT

Given that addiction has been characterized as a disorder of maladaptive learning and memory, one critical question is whether there are unique physical adaptations within neuronal ensembles that support addiction-related learned behavior. The search for the physical mechanisms of encoding these and other memories in the brain, often called the engram as a whole, continues despite decades of research. As we develop new technologies and tools that allow us to study cue- and behavior-activated Fos-expressing neuronal ensembles, the possibility of identifying the engrams of learning and memory is moving into the realm of reality rather than speculation. It has become clear from recent studies that there are specific functional, electrophysiological alterations unique to Fos-expressing ensemble neurons that may participate in encoding memories. The ultimate goal is to identify the addicted engram and reverse the physical changes that support this maladaptive form of learning.


Subject(s)
Learning/physiology , Neurons/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Substance-Related Disorders/metabolism , Animals , Humans
7.
Neuron ; 96(2): 414-427.e8, 2017 Oct 11.
Article in English | MEDLINE | ID: mdl-29024664

ABSTRACT

Despite decades of research on neurobiological mechanisms of psychostimulant addiction, the only effective treatment for many addicts is contingency management, a behavioral treatment that uses alternative non-drug reward to maintain abstinence. However, when contingency management is discontinued, most addicts relapse to drug use. The brain mechanisms underlying relapse after cessation of contingency management are largely unknown, and, until recently, an animal model of this human condition did not exist. Here we used a novel rat model, in which the availability of a mutually exclusive palatable food maintains prolonged voluntary abstinence from intravenous methamphetamine self-administration, to demonstrate that the activation of monosynaptic glutamatergic projections from anterior insular cortex to central amygdala is critical to relapse after the cessation of contingency management. We identified the anterior insular cortex-to-central amygdala projection as a new addiction- and motivation-related projection and a potential target for relapse prevention.


Subject(s)
Behavior, Addictive/psychology , Central Amygdaloid Nucleus/physiology , Cerebral Cortex/physiology , Eating/physiology , Eating/psychology , Methamphetamine/administration & dosage , Animals , Central Amygdaloid Nucleus/drug effects , Cerebral Cortex/drug effects , Eating/drug effects , Injections, Intravenous , Male , Neural Pathways/drug effects , Neural Pathways/physiology , Rats , Rats, Sprague-Dawley , Recurrence , Self Administration
8.
J Neurosci ; 37(36): 8845-8856, 2017 09 06.
Article in English | MEDLINE | ID: mdl-28779019

ABSTRACT

Learned associations between environmental stimuli and rewards drive goal-directed learning and motivated behavior. These memories are thought to be encoded by alterations within specific patterns of sparsely distributed neurons called neuronal ensembles that are activated selectively by reward-predictive stimuli. Here, we use the Fos promoter to identify strongly activated neuronal ensembles in rat prelimbic cortex (PLC) and assess altered intrinsic excitability after 10 d of operant food self-administration training (1 h/d). First, we used the Daun02 inactivation procedure in male FosLacZ-transgenic rats to ablate selectively Fos-expressing PLC neurons that were active during operant food self-administration. Selective ablation of these neurons decreased food seeking. We then used male FosGFP-transgenic rats to assess selective alterations of intrinsic excitability in Fos-expressing neuronal ensembles (FosGFP+) that were activated during food self-administration and compared these with alterations in less activated non-ensemble neurons (FosGFP-). Using whole-cell recordings of layer V pyramidal neurons in an ex vivo brain slice preparation, we found that operant self-administration increased excitability of FosGFP+ neurons and decreased excitability of FosGFP- neurons. Increased excitability of FosGFP+ neurons was driven by increased steady-state input resistance. Decreased excitability of FosGFP- neurons was driven by increased contribution of small-conductance calcium-activated potassium (SK) channels. Injections of the specific SK channel antagonist apamin into PLC increased Fos expression but had no effect on food seeking. Overall, operant learning increased intrinsic excitability of PLC Fos-expressing neuronal ensembles that play a role in food seeking but decreased intrinsic excitability of Fos- non-ensembles.SIGNIFICANCE STATEMENT Prefrontal cortex activity plays a critical role in operant learning, but the underlying cellular mechanisms are unknown. Using the chemogenetic Daun02 inactivation procedure, we found that a small number of strongly activated Fos-expressing neuronal ensembles in rat PLC play an important role in learned operant food seeking. Using GFP expression to identify Fos-expressing layer V pyramidal neurons in prelimbic cortex (PLC) of FosGFP-transgenic rats, we found that operant food self-administration led to increased intrinsic excitability in the behaviorally relevant Fos-expressing neuronal ensembles, but decreased intrinsic excitability in Fos- neurons using distinct cellular mechanisms.


Subject(s)
Action Potentials/physiology , Association Learning/physiology , Conditioning, Operant/physiology , Nerve Net/physiology , Neuronal Plasticity/physiology , Prefrontal Cortex/physiology , Animals , Male , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Rats, Transgenic
9.
Curr Biol ; 27(14): 2089-2100.e5, 2017 Jul 24.
Article in English | MEDLINE | ID: mdl-28690111

ABSTRACT

Eating is a learned process. Our desires for specific foods arise through experience. Both electrical stimulation and optogenetic studies have shown that increased activity in the lateral hypothalamus (LH) promotes feeding. Current dogma is that these effects reflect a role for LH neurons in the control of the core motivation to feed, and their activity comes under control of forebrain regions to elicit learned food-motivated behaviors. However, these effects could also reflect the storage of associative information about the cues leading to food in LH itself. Here, we present data from several studies that are consistent with a role for LH in learning. In the first experiment, we use a novel GAD-Cre rat to show that optogenetic inhibition of LH γ-aminobutyric acid (GABA) neurons restricted to cue presentation disrupts the rats' ability to learn that a cue predicts food without affecting subsequent food consumption. In the second experiment, we show that this manipulation also disrupts the ability of a cue to promote food seeking after learning. Finally, we show that inhibition of the terminals of the LH GABA neurons in ventral-tegmental area (VTA) facilitates learning about reward-paired cues. These results suggest that the LH GABA neurons are critical for storing and later disseminating information about reward-predictive cues.


Subject(s)
Feeding Behavior/physiology , GABAergic Neurons/physiology , Hypothalamic Area, Lateral/physiology , Learning/physiology , Motivation/physiology , Reward , Ventral Tegmental Area/physiology , Animals , Cues , Female , Male , Optogenetics , Rats , Rats, Long-Evans
10.
J Neurosci Methods ; 284: 1-14, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28380331

ABSTRACT

BACKGROUND: The use of genetically-encoded fluorescent reporters is essential for the identification and observation of cells that express transgenic modulatory proteins. Near-infrared (NIR) fluorescent proteins have superior light penetration through biological tissue, but are not yet widely adopted. NEW METHOD: Using the near-infrared fluorescent protein, iRFP713, improves the imaging resolution in thick tissue sections or the intact brain due to the reduced light-scattering at the longer, NIR wavelengths used to image the protein. Additionally, iRFP713 can be used to identify transgenic cells without photobleaching other fluorescent reporters or affecting opsin function. We have generated a set of adeno-associated vectors in which iRFP713 has been fused to optogenetic channels, and can be expressed constitutively or Cre-dependently. RESULTS: iRFP713 is detectable when expressed in neurons both in vitro and in vivo without exogenously supplied chromophore biliverdin. Neuronally-expressed iRFP713 has similar properties to GFP-like fluorescent proteins, including the ability to be translationally fused to channelrhodopsin or halorhodopsin, however, it shows superior photostability compared to EYFP. Furthermore, electrophysiological recordings from iRFP713-labeled cells compared to cells labeled with mCherry suggest that iRFP713 cells are healthier and therefore more stable and reliable in an ex vivo preparation. Lastly, we have generated a transgenic rat that expresses iRFP713 in a Cre-dependent manner. CONCLUSIONS: Overall, we have demonstrated that iRFP713 can be used as a reporter in neurons without the use of exogenous biliverdin, with minimal impact on viability and function thereby making it feasible to extend the capabilities for imaging genetically-tagged neurons in slices and in vivo.


Subject(s)
Genes, Reporter/genetics , Green Fluorescent Proteins/metabolism , Microscopy, Fluorescence/methods , Neurons/metabolism , Optogenetics/methods , Spectroscopy, Near-Infrared/methods , Voltage-Sensitive Dye Imaging/methods , Animals , Cells, Cultured , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , Luminescent Proteins , Molecular Imaging/methods , Neurons/cytology , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Reproducibility of Results , Sensitivity and Specificity
11.
J Neurosci ; 36(25): 6691-703, 2016 06 22.
Article in English | MEDLINE | ID: mdl-27335401

ABSTRACT

UNLABELLED: In operant learning, initial reward-associated memories are thought to be distinct from subsequent extinction-associated memories. Memories formed during operant learning are thought to be stored in "neuronal ensembles." Thus, we hypothesize that different neuronal ensembles encode reward- and extinction-associated memories. Here, we examined prefrontal cortex neuronal ensembles involved in the recall of reward and extinction memories of food self-administration. We first trained rats to lever press for palatable food pellets for 7 d (1 h/d) and then exposed them to 0, 2, or 7 daily extinction sessions in which lever presses were not reinforced. Twenty-four hours after the last training or extinction session, we exposed the rats to either a short 15 min extinction test session or left them in their homecage (a control condition). We found maximal Fos (a neuronal activity marker) immunoreactivity in the ventral medial prefrontal cortex of rats that previously received 2 extinction sessions, suggesting that neuronal ensembles in this area encode extinction memories. We then used the Daun02 inactivation procedure to selectively disrupt ventral medial prefrontal cortex neuronal ensembles that were activated during the 15 min extinction session following 0 (no extinction) or 2 prior extinction sessions to determine the effects of inactivating the putative food reward and extinction ensembles, respectively, on subsequent nonreinforced food seeking 2 d later. Inactivation of the food reward ensembles decreased food seeking, whereas inactivation of the extinction ensembles increased food seeking. Our results indicate that distinct neuronal ensembles encoding operant reward and extinction memories intermingle within the same cortical area. SIGNIFICANCE STATEMENT: A current popular hypothesis is that neuronal ensembles in different prefrontal cortex areas control reward-associated versus extinction-associated memories: the dorsal medial prefrontal cortex (mPFC) promotes reward seeking, whereas the ventral mPFC inhibits reward seeking. In this paper, we use the Daun02 chemogenetic inactivation procedure to demonstrate that Fos-expressing neuronal ensembles mediating both food reward and extinction memories intermingle within the same ventral mPFC area.


Subject(s)
Extinction, Psychological/physiology , Neurons/metabolism , Oncogene Proteins v-fos/metabolism , Prefrontal Cortex/cytology , Prefrontal Cortex/physiology , Reward , Animals , Conditioning, Operant/drug effects , Conditioning, Operant/physiology , Daunorubicin/analogs & derivatives , Daunorubicin/pharmacology , Enzyme Inhibitors/pharmacology , Extinction, Psychological/drug effects , GABA Agents/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Male , Mental Recall/drug effects , Neurons/drug effects , Phosphopyruvate Hydratase/metabolism , Prefrontal Cortex/drug effects , Rats , Rats, Long-Evans , Self Administration , Time Factors , Vesicular Glutamate Transport Protein 1/genetics , Vesicular Glutamate Transport Protein 1/metabolism , Vesicular Inhibitory Amino Acid Transport Proteins/genetics , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism
12.
J Neurosci ; 36(11): 3281-94, 2016 Mar 16.
Article in English | MEDLINE | ID: mdl-26985037

ABSTRACT

In many human alcoholics, abstinence is self-imposed because of the negative consequences of excessive alcohol use, and relapse is often triggered by exposure to environmental contexts associated with prior alcohol drinking. We recently developed a rat model of this human condition in which we train alcohol-preferring P rats to self-administer alcohol in one context (A), punish the alcohol-reinforced responding in a different context (B), and then test for relapse to alcohol seeking in Contexts A and B without alcohol or shock. Here, we studied the role of projections to nucleus accumbens (NAc) shell from ventral subiculum (vSub), basolateral amygdala, paraventricular thalamus, and ventral medial prefrontal cortex in context-induced relapse after punishment-imposed abstinence. First, we measured double-labeling of the neuronal activity marker Fos with the retrograde tracer cholera toxin subunit B (injected in NAc shell) and demonstrated that context-induced relapse is associated with selective activation of the vSub→NAc shell projection. Next, we reversibly inactivated the vSub with GABA receptor agonists (muscimol+baclofen) before the context-induced relapse tests and provided evidence for a causal role of vSub in this relapse. Finally, we used a dual-virus approach to restrict expression of the inhibitory κ opioid-receptor based DREADD (KORD) in vSub→NAc shell projection neurons. We found that systemic injections of the KORD agonist salvinorin B, which selectively inhibits KORD-expressing neurons, decreased context-induced relapse to alcohol seeking. Our results demonstrate a critical role of vSub in context-induced relapse after punishment-imposed abstinence and further suggest a role of the vSub→NAc projection in this relapse. SIGNIFICANCE STATEMENT: In many human alcoholics, abstinence is self-imposed because of the negative consequences of excessive use, and relapse is often triggered by exposure to environmental contexts associated with prior alcohol use. Until recently, an animal model of this human condition did not exist. We developed a rat model of this human condition in which we train alcohol-preferring P rats to self-administer alcohol in one context (A), punish the alcohol-reinforced responding in a different context (B), and test for relapse to alcohol seeking in Contexts A and B. Here, we used neuroanatomical, neuropharmacological, and chemogenetic methods to demonstrate a role of ventral subiculum and potentially its projections to nucleus accumbens in context-induced relapse after punishment-imposed abstinence.


Subject(s)
Alcohol Abstinence/psychology , Alcohol Drinking/psychology , Conditioning, Operant/physiology , Extinction, Psychological/physiology , Nucleus Accumbens/physiopathology , Punishment , Alcohol Drinking/physiopathology , Animals , Cholera Toxin/metabolism , Conditioning, Operant/drug effects , Diterpenes/pharmacology , Diterpenes, Clerodane , Ethanol/administration & dosage , Extinction, Psychological/drug effects , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Neurons/metabolism , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Nucleus Accumbens/pathology , Oncogene Proteins v-fos/genetics , Oncogene Proteins v-fos/metabolism , Rats , Receptors, Opioid, kappa/metabolism , Recurrence , Reinforcement, Psychology , Self Administration , Transduction, Genetic
13.
Biol Psychiatry ; 80(3): 246-56, 2016 08 01.
Article in English | MEDLINE | ID: mdl-26386479

ABSTRACT

BACKGROUND: Learned associations between environmental stimuli and rewards play a critical role in addiction. Associative learning requires alterations in sparsely distributed populations of strongly activated neurons, or neuronal ensembles. Until recently, assessment of functional alterations underlying learned behavior was restricted to global neuroadaptations in a particular brain area or cell type, rendering it impossible to identify neuronal ensembles critically involved in learned behavior. METHODS: We used Fos-GFP transgenic mice that contained a transgene with a Fos promoter driving expression of green fluorescent protein (GFP) to detect neurons that were strongly activated during associative learning, in this case, context-independent and context-specific cocaine-induced locomotor sensitization. Whole-cell electrophysiological recordings were used to assess synaptic alterations in specifically activated GFP-positive (GFP+) neurons compared with surrounding nonactivated GFP-negative (GFP-) neurons 90 min after the sensitized locomotor response. RESULTS: After context-independent cocaine sensitization, cocaine-induced locomotion was equally sensitized by repeated cocaine injections in two different sensitization contexts. Correspondingly, silent synapses in these mice were induced in GFP+ neurons, but not GFP- neurons, after sensitization in both of these contexts. After context-specific cocaine sensitization, cocaine-induced locomotion was sensitized exclusively in mice trained and tested in the same context (paired group), but not in mice that were trained in one context and then tested in a different context (unpaired group). Silent synapses increased in GFP+ neurons, but not in GFP- neurons from mice in the paired group, but not from mice in the unpaired group. CONCLUSIONS: Our results indicate that silent synapses are formed only in neuronal ensembles of the nucleus accumbens shell that are related to associative learning.


Subject(s)
Association Learning/physiology , Neurons/metabolism , Nucleus Accumbens/cytology , Synapses/metabolism , Animals , Central Nervous System Sensitization/drug effects , Central Nervous System Sensitization/physiology , Cocaine/pharmacology , Locomotion/drug effects , Locomotion/physiology , Male , Mice , Mice, Transgenic , Nucleus Accumbens/physiology , Proto-Oncogene Proteins c-fos/metabolism
14.
Neuropsychopharmacology ; 41(2): 402-9, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26019014

ABSTRACT

In the past decade, novel methods using engineered receptors have enabled researchers to manipulate neuronal activity with increased spatial and temporal specificity. One widely used chemogenetic method in mice and rats is the DREADD (designer receptors exclusively activated by designer drugs) system in which a mutated muscarinic G protein-coupled receptor is activated by an otherwise inert synthetic ligand, clozapine-N-oxide (CNO). Recently, the Roth laboratory developed a novel inhibitory DREADD in which a mutated kappa-opioid receptor (KORD) is activated by the pharmacologically inert drug salvinorin B (SalB; Vardy et al, 2015). They demonstrated the feasibility of using KORD to study brain circuits involved in motivated behavior in mice. Here, we used behavioral, electrophysiological, and neuroanatomical methods to demonstrate the feasibility of using the novel KORD to study brain circuits involved in motivated behavior in rats. In Exp. 1, we show that SalB dose-dependently decreased spontaneous and cocaine-induced locomotor activity in rats expressing KORD to midbrain (ventral tegmental area/substantia nigra). In Exp. 2, we show that SalB completely inhibited tonic firing in KORD-expressing putative dopamine neurons in midbrain. In Exp. 3, we used a 'retro-DREADD' dual-virus approach to restrict expression of KORD in ventral subiculum neurons that project to nucleus accumbens shell. We show that KORD activation selectively decreased novel context-induced Fos expression in this projection. Our results indicate that the novel KORD is a promising tool to selectively inactivate brain areas and neural circuits in rat studies of motivated behavior.


Subject(s)
Gene Transfer Techniques , Mesencephalon/physiology , Receptors, Opioid, kappa/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Animals , Central Nervous System Agents/pharmacology , Cocaine/pharmacology , Dependovirus/genetics , Diterpenes/pharmacology , Diterpenes, Clerodane , Dopamine Uptake Inhibitors/pharmacology , Feasibility Studies , Genetic Engineering , Genetic Vectors , Male , Mesencephalon/cytology , Mesencephalon/drug effects , Motor Activity/drug effects , Motor Activity/physiology , Mutation , Neurons/cytology , Neurons/drug effects , Neurons/physiology , Rats, Sprague-Dawley , Receptors, Opioid, kappa/genetics
15.
PLoS One ; 10(10): e0139273, 2015.
Article in English | MEDLINE | ID: mdl-26451944

ABSTRACT

Endoplasmic reticulum calcium homeostasis is critical for cellular functions and is disrupted in diverse pathologies including neurodegeneration and cardiovascular disease. Owing to the high concentration of calcium within the ER, studying this subcellular compartment requires tools that are optimized for these conditions. To develop a single-fluorophore genetically encoded calcium indicator for this organelle, we targeted a low affinity variant of GCaMP3 to the ER lumen (GCaMPer (10.19)). A set of viral vectors was constructed to express GCaMPer in human neuroblastoma cells, rat primary cortical neurons, and human induced pluripotent stem cell-derived cardiomyocytes. We observed dynamic changes in GCaMPer (10.19) fluorescence in response to pharmacologic manipulations of the ER calcium store. Additionally, periodic calcium efflux from the ER was observed during spontaneous beating of cardiomyocytes. GCaMPer (10.19) has utility in imaging ER calcium in living cells and providing insight into luminal calcium dynamics under physiologic and pathologic states.


Subject(s)
Calcium/metabolism , Endoplasmic Reticulum/metabolism , Molecular Imaging/methods , Recombinant Fusion Proteins/metabolism , Amino Acid Sequence , Animals , Cell Line, Tumor , HEK293 Cells , Humans , Models, Molecular , Molecular Sequence Data , Myocytes, Cardiac/cytology , Neurons/cytology , Protein Conformation , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics
16.
Neuron ; 77(2): 335-45, 2013 Jan 23.
Article in English | MEDLINE | ID: mdl-23352169

ABSTRACT

Drug addiction is driven, in part, by powerful drug-related memories. Deficits in social life, particularly during adolescence, increase addiction vulnerability. Social isolation in rodents has been used extensively to model the effects of deficient social experience, yet its impact on learning and memory processes underlying addiction remains elusive. Here, we show that social isolation of rats during a critical period of adolescence (postnatal days 21-42) enhances long-term potentiation of NMDA receptor (NMDAR)-mediated glutamatergic transmission in the ventral tegmental area (VTA). This enhancement, which is caused by an increase in metabotropic glutamate receptor-dependent Ca(2+) signaling, cannot be reversed by subsequent resocialization. Notably, memories of amphetamine- and ethanol-paired contextual stimuli are acquired faster and, once acquired, amphetamine-associated contextual memory is more resistant to extinction in socially isolated rats. We propose that NMDAR plasticity in the VTA may represent a neural substrate by which early life deficits in social experience increase addiction vulnerability.


Subject(s)
Dextroamphetamine/pharmacology , Learning/physiology , Neuronal Plasticity/physiology , Social Isolation/psychology , Synapses/physiology , Ventral Tegmental Area/physiology , Age Factors , Animals , Learning/drug effects , Male , Methoxyhydroxyphenylglycol/analogs & derivatives , Methoxyhydroxyphenylglycol/pharmacology , Neuronal Plasticity/drug effects , Rats , Rats, Sprague-Dawley , Synapses/drug effects , Ventral Tegmental Area/drug effects
17.
J Neurosci ; 31(14): 5205-12, 2011 Apr 06.
Article in English | MEDLINE | ID: mdl-21471355

ABSTRACT

Alcohol addiction (alcoholism) is one of the most prevalent substance abuse disorders worldwide. Addiction is thought to arise, in part, from a maladaptive learning process in which enduring memories of drug experiences are formed. However, alcohol (ethanol) generally interferes with synaptic plasticity mechanisms in the CNS and thus impairs various types of learning and memory. Therefore, it is unclear how powerful memories associated with alcohol experience are formed during the development of alcoholism. Here, using brain slice electrophysiology in mice, we show that repeated in vivo ethanol exposure (2 g/kg, i.p., three times daily for 7 d) causes increased susceptibility to the induction of long-term potentiation (LTP) of NMDA receptor (NMDAR)-mediated transmission in mesolimbic dopamine neurons, a form of synaptic plasticity that may drive the learning of stimuli associated with rewards, including drugs of abuse. Enhancement of NMDAR plasticity results from an increase in the potency of inositol 1,4,5-trisphosphate (IP(3)) in producing facilitation of action potential-evoked Ca(2+) signals, which is critical for LTP induction. This increase in IP(3) effect, which lasts for a week but not a month after ethanol withdrawal, occurs through a protein kinase A (PKA)-dependent mechanism. Corticotropin-releasing factor, a stress-related neuropeptide implicated in alcoholism and other addictions, further amplifies the PKA-mediated increase in IP(3) effect in ethanol-treated mice. Finally, we found that ethanol-treated mice display enhanced place conditioning induced by the psychostimulant cocaine. These data suggest that repeated ethanol experience may promote the formation of drug-associated memories by enhancing synaptic plasticity of NMDARs in dopamine neurons.


Subject(s)
Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Neuronal Plasticity/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Ventral Tegmental Area/drug effects , Amphibian Proteins/pharmacology , Analysis of Variance , Animals , Biophysics , Cocaine/pharmacology , Colforsin/pharmacology , Conditioning, Operant/drug effects , Conditioning, Operant/physiology , Corticotropin-Releasing Hormone/pharmacology , Dopamine Antagonists/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Interactions , Electric Stimulation , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , In Vitro Techniques , Inositol 1,4,5-Trisphosphate/pharmacology , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Male , Methoxyhydroxyphenylglycol/analogs & derivatives , Methoxyhydroxyphenylglycol/pharmacology , Mice , Mice, Inbred C57BL , Neurons/drug effects , Peptide Hormones/pharmacology , Quinoxalines/pharmacology , Salicylamides/pharmacology , Ventral Tegmental Area/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...