Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.664
Filter
1.
Prog Neurobiol ; 239: 102630, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38834131

ABSTRACT

Dopamine critically influences reward processing, sensory perception, and motor control. Yet, the modulation of dopaminergic signaling by sensory experiences is not fully delineated. Here, by manipulating sensory experience using bilateral single-row whisker deprivation, we demonstrated that gene transcription in the dopaminergic signaling pathway (DSP) undergoes experience-dependent plasticity in both granular and supragranular layers of the primary somatosensory (barrel) cortex (S1). Sensory experience and deprivation compete for the regulation of DSP transcription across neighboring cortical columns, and sensory deprivation-induced changes in DSP are topographically constrained. These changes in DSP extend beyond cortical map plasticity and influence neuronal information processing. Pharmacological regulation of D2 receptors, a key component of DSP, revealed that D2 receptor activation suppresses excitatory neuronal excitability, hyperpolarizes the action potential threshold, and reduces the instantaneous firing rate. These findings suggest that the dopaminergic drive originating from midbrain dopaminergic neurons, targeting the sensory cortex, is subject to experience-dependent regulation and might create a regulatory feedback loop for modulating sensory processing. Finally, using topological gene network analysis and mutual information, we identify the molecular hubs of experience-dependent plasticity of DSP. These findings provide new insights into the mechanisms by which sensory experience shapes dopaminergic signaling in the brain and might help unravel the sensory deficits observed after dopamine depletion.


Subject(s)
Dopamine , Neuronal Plasticity , Signal Transduction , Somatosensory Cortex , Somatosensory Cortex/metabolism , Somatosensory Cortex/physiology , Animals , Signal Transduction/physiology , Dopamine/metabolism , Neuronal Plasticity/physiology , Dopaminergic Neurons/physiology , Dopaminergic Neurons/metabolism , Vibrissae/physiology , Receptors, Dopamine D2/metabolism , Sensory Deprivation/physiology , Mice , Male
2.
Nature ; 630(8017): 677-685, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38839962

ABSTRACT

All drugs of abuse induce long-lasting changes in synaptic transmission and neural circuit function that underlie substance-use disorders1,2. Another recently appreciated mechanism of neural circuit plasticity is mediated through activity-regulated changes in myelin that can tune circuit function and influence cognitive behaviour3-7. Here we explore the role of myelin plasticity in dopaminergic circuitry and reward learning. We demonstrate that dopaminergic neuronal activity-regulated myelin plasticity is a key modulator of dopaminergic circuit function and opioid reward. Oligodendroglial lineage cells respond to dopaminergic neuronal activity evoked by optogenetic stimulation of dopaminergic neurons, optogenetic inhibition of GABAergic neurons, or administration of morphine. These oligodendroglial changes are evident selectively within the ventral tegmental area but not along the axonal projections in the medial forebrain bundle nor within the target nucleus accumbens. Genetic blockade of oligodendrogenesis dampens dopamine release dynamics in nucleus accumbens and impairs behavioural conditioning to morphine. Taken together, these findings underscore a critical role for oligodendrogenesis in reward learning and identify dopaminergic neuronal activity-regulated myelin plasticity as an important circuit modification that is required for opioid reward.


Subject(s)
Dopaminergic Neurons , GABAergic Neurons , Morphine , Myelin Sheath , Neuronal Plasticity , Nucleus Accumbens , Oligodendroglia , Optogenetics , Reward , Ventral Tegmental Area , Ventral Tegmental Area/physiology , Ventral Tegmental Area/cytology , Ventral Tegmental Area/drug effects , Animals , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/physiology , Mice , Myelin Sheath/metabolism , Morphine/pharmacology , Male , Nucleus Accumbens/cytology , Nucleus Accumbens/metabolism , Nucleus Accumbens/physiology , Nucleus Accumbens/drug effects , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Oligodendroglia/metabolism , Oligodendroglia/cytology , Oligodendroglia/drug effects , GABAergic Neurons/metabolism , GABAergic Neurons/drug effects , Analgesics, Opioid/pharmacology , Dopamine/metabolism , Female , Mice, Inbred C57BL
3.
Behav Neurosci ; 138(3): 164-177, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38934920

ABSTRACT

A growing body of literature indicates that mediated learning techniques have specific utility for tapping into reality testing in animal models of neuropsychiatric illness. In particular, recent work has shown that animal models that recapitulate various endophenotypes of schizophrenia are particularly vulnerable to impairments in reality testing when undergoing mediated learning. Multiple studies have indicated that these effects are dopamine receptor 2-dependent and correlated with aberrant insular cortex (IC) activity. However, until now, the connection between dopamine and the IC had not been investigated. Here, we utilized a novel intersectional approach to label mesencephalic dopamine cells that specifically project to the insular cortex in both wild-type controls and transgenic mice expressing the dominant-negative form of the Disrupted-in-Schizophrenia-1 (DISC-1) gene. Using these techniques, we identified a population of cells that project from the ventral tegmental area (VTA) to the IC. Afterward, we conducted multiple studies to test the necessity of this circuit in behaviors ranging from gustatory detection to the maintenance of effort and, finally, mediated performance. Our results indicate that perturbations of the DISC-1 genetic locus lead to a reduction in the number of cells in the VTA → IC circuit. Behaviorally, VTA → IC circuitry does not influence gustatory detection or motivation to acquire sucrose reward; however, inactivation of this circuit differentially suppresses Pavlovian approach behavior in wild-type and DISC-1 transgenic mice during mediated performance testing. Moreover, under these testing conditions, inactivation of this circuit predisposes wild-type (but not DISC-1) mice to display impaired reality testing. (PsycInfo Database Record (c) 2024 APA, all rights reserved).


Subject(s)
Dopaminergic Neurons , Insular Cortex , Mice, Transgenic , Animals , Dopaminergic Neurons/physiology , Dopaminergic Neurons/metabolism , Mice , Insular Cortex/physiology , Male , Ventral Tegmental Area/physiology , Ventral Tegmental Area/metabolism , Mice, Inbred C57BL , Neural Pathways/physiology , Reward , Disease Models, Animal , Dopamine/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Mesencephalon/metabolism , Mesencephalon/physiology , Schizophrenia/physiopathology
4.
Learn Mem ; 31(5)2024 May.
Article in English | MEDLINE | ID: mdl-38862171

ABSTRACT

Across animal species, dopamine-operated memory systems comprise anatomically segregated, functionally diverse subsystems. Although individual subsystems could operate independently to support distinct types of memory, the logical interplay between subsystems is expected to enable more complex memory processing by allowing existing memory to influence future learning. Recent comprehensive ultrastructural analysis of the Drosophila mushroom body revealed intricate networks interconnecting the dopamine subsystems-the mushroom body compartments. Here, we review the functions of some of these connections that are beginning to be understood. Memory consolidation is mediated by two different forms of network: A recurrent feedback loop within a compartment maintains sustained dopamine activity required for consolidation, whereas feed-forward connections across compartments allow short-term memory formation in one compartment to open the gate for long-term memory formation in another compartment. Extinction and reversal of aversive memory rely on a similar feed-forward circuit motif that signals omission of punishment as a reward, which triggers plasticity that counteracts the original aversive memory trace. Finally, indirect feed-forward connections from a long-term memory compartment to short-term memory compartments mediate higher-order conditioning. Collectively, these emerging studies indicate that feedback control and hierarchical connectivity allow the dopamine subsystems to work cooperatively to support diverse and complex forms of learning.


Subject(s)
Dopamine , Mushroom Bodies , Animals , Dopamine/metabolism , Dopamine/physiology , Mushroom Bodies/physiology , Mushroom Bodies/metabolism , Drosophila/physiology , Feedback, Physiological/physiology , Memory Consolidation/physiology , Nerve Net/physiology , Nerve Net/metabolism , Dopaminergic Neurons/physiology , Dopaminergic Neurons/metabolism , Neural Pathways/physiology
5.
Learn Mem ; 31(5)2024 May.
Article in English | MEDLINE | ID: mdl-38862177

ABSTRACT

Associative learning enables the adaptive adjustment of behavioral decisions based on acquired, predicted outcomes. The valence of what is learned is influenced not only by the learned stimuli and their temporal relations, but also by prior experiences and internal states. In this study, we used the fruit fly Drosophila melanogaster to demonstrate that neuronal circuits involved in associative olfactory learning undergo restructuring during extended periods of low-caloric food intake. Specifically, we observed a decrease in the connections between specific dopaminergic neurons (DANs) and Kenyon cells at distinct compartments of the mushroom body. This structural synaptic plasticity was contingent upon the presence of allatostatin A receptors in specific DANs and could be mimicked optogenetically by expressing a light-activated adenylate cyclase in exactly these DANs. Importantly, we found that this rearrangement in synaptic connections influenced aversive, punishment-induced olfactory learning but did not impact appetitive, reward-based learning. Whether induced by prolonged low-caloric conditions or optogenetic manipulation of cAMP levels, this synaptic rearrangement resulted in a reduction of aversive associative learning. Consequently, the balance between positive and negative reinforcing signals shifted, diminishing the ability to learn to avoid odor cues signaling negative outcomes. These results exemplify how a neuronal circuit required for learning and memory undergoes structural plasticity dependent on prior experiences of the nutritional value of food.


Subject(s)
Drosophila melanogaster , Mushroom Bodies , Neuronal Plasticity , Animals , Mushroom Bodies/physiology , Mushroom Bodies/metabolism , Drosophila melanogaster/physiology , Neuronal Plasticity/physiology , Dopaminergic Neurons/physiology , Dopaminergic Neurons/metabolism , Eating/physiology , Optogenetics , Association Learning/physiology , Smell/physiology , Olfactory Perception/physiology , Reward , Animals, Genetically Modified
6.
Proc Natl Acad Sci U S A ; 121(24): e2401929121, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38843183

ABSTRACT

Punishment such as electric shock or physical discipline employs a mixture of physical pain and emotional distress to induce behavior modification. However, a neural circuit that produces behavior modification by selectively focusing the emotional component, while bypassing the pain typically induced by peripheral nociceptor activation, is not well studied. Here, we show that genetically silencing the activity of neurons expressing calcitonin gene-related peptide (CGRP) in the parabrachial nucleus blocks the suppression of addictive-like behavior induced by footshock. Furthermore, activating CGRP neurons suppresses not only addictive behavior induced by self-stimulating dopamine neurons but also behavior resulting from self-administering cocaine, without eliciting nocifensive reactions. Moreover, among multiple downstream targets of CGRP neurons, terminal activation of CGRP in the central amygdala is effective, mimicking the results of cell body stimulation. Our results indicate that unlike conventional electric footshock, stimulation of CGRP neurons does not activate peripheral nociceptors but effectively curb addictive behavior.


Subject(s)
Behavior, Addictive , Calcitonin Gene-Related Peptide , Neurons , Parabrachial Nucleus , Animals , Parabrachial Nucleus/metabolism , Parabrachial Nucleus/physiology , Calcitonin Gene-Related Peptide/metabolism , Mice , Neurons/metabolism , Neurons/physiology , Behavior, Addictive/metabolism , Male , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/physiology , Cocaine/pharmacology , Behavior, Animal/physiology
7.
Elife ; 122024 May 15.
Article in English | MEDLINE | ID: mdl-38747563

ABSTRACT

Midbrain dopamine neurons impact neural processing in the prefrontal cortex (PFC) through mesocortical projections. However, the signals conveyed by dopamine projections to the PFC remain unclear, particularly at the single-axon level. Here, we investigated dopaminergic axonal activity in the medial PFC (mPFC) during reward and aversive processing. By optimizing microprism-mediated two-photon calcium imaging of dopamine axon terminals, we found diverse activity in dopamine axons responsive to both reward and aversive stimuli. Some axons exhibited a preference for reward, while others favored aversive stimuli, and there was a strong bias for the latter at the population level. Long-term longitudinal imaging revealed that the preference was maintained in reward- and aversive-preferring axons throughout classical conditioning in which rewarding and aversive stimuli were paired with preceding auditory cues. However, as mice learned to discriminate reward or aversive cues, a cue activity preference gradually developed only in aversive-preferring axons. We inferred the trial-by-trial cue discrimination based on machine learning using anticipatory licking or facial expressions, and found that successful discrimination was accompanied by sharper selectivity for the aversive cue in aversive-preferring axons. Our findings indicate that a group of mesocortical dopamine axons encodes aversive-related signals, which are modulated by both classical conditioning across days and trial-by-trial discrimination within a day.


Subject(s)
Axons , Conditioning, Classical , Dopaminergic Neurons , Prefrontal Cortex , Animals , Prefrontal Cortex/physiology , Mice , Axons/physiology , Conditioning, Classical/physiology , Dopaminergic Neurons/physiology , Male , Reward , Dopamine/metabolism , Mice, Inbred C57BL , Cues
8.
Nat Commun ; 15(1): 4233, 2024 May 18.
Article in English | MEDLINE | ID: mdl-38762463

ABSTRACT

The ventral pallidum (VP) contains GABA and glutamate neurons projecting to ventral tegmental area (VTA) whose stimulation drives approach and avoidance, respectively. Yet little is known about the mechanisms by which VP cell types shape VTA activity and drive behavior. Here, we found that both VP GABA and glutamate neurons were activated during approach to reward or by delivery of an aversive stimulus. Stimulation of VP GABA neurons inhibited VTA GABA, but activated dopamine and glutamate neurons. Remarkably, stimulation-evoked activation was behavior-contingent such that VTA recruitment was inhibited when evoked by the subject's own action. Conversely, VP glutamate neurons activated VTA GABA, as well as dopamine and glutamate neurons, despite driving aversion. However, VP glutamate neurons evoked dopamine in aversion-associated ventromedial nucleus accumbens (NAc), but reduced dopamine release in reward-associated dorsomedial NAc. These findings show how heterogeneous VP projections to VTA can be engaged to shape approach and avoidance behaviors.


Subject(s)
Avoidance Learning , Basal Forebrain , GABAergic Neurons , Glutamic Acid , Reward , Ventral Tegmental Area , Ventral Tegmental Area/physiology , Ventral Tegmental Area/metabolism , Ventral Tegmental Area/cytology , Animals , Glutamic Acid/metabolism , Basal Forebrain/metabolism , Basal Forebrain/physiology , Male , GABAergic Neurons/metabolism , GABAergic Neurons/physiology , Avoidance Learning/physiology , Mice , Dopamine/metabolism , Nucleus Accumbens/metabolism , Nucleus Accumbens/cytology , Nucleus Accumbens/physiology , Neurons/metabolism , Neurons/physiology , gamma-Aminobutyric Acid/metabolism , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/physiology , Mice, Inbred C57BL , Behavior, Animal/physiology
9.
Neurobiol Learn Mem ; 212: 107937, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38735637

ABSTRACT

Systemic manipulations that enhance dopamine (DA) transmission around the time of fear extinction can strengthen fear extinction and reduce conditioned fear relapse. Prior studies investigating the brain regions where DA augments fear extinction focus on targets of mesolimbic and mesocortical DA systems originating in the ventral tegmental area, given the role of these DA neurons in prediction error. The dorsal striatum (DS), a primary target of the nigrostriatal DA system originating in the substantia nigra (SN), is implicated in behaviors beyond its canonical role in movement, such as reward and punishment, goal-directed action, and stimulus-response associations, but whether DS DA contributes to fear extinction is unknown. We have observed that chemogenetic stimulation of SN DA neurons during fear extinction prevents the return of fear in contexts different from the extinction context, a form of relapse called renewal. This effect of SN DA stimulation is mimicked by a DA D1 receptor (D1R) agonist injected into the DS, thus implicating DS DA in fear extinction. Different DS subregions subserve unique functions of the DS, but it is unclear where in the DS D1R agonist acts during fear extinction to reduce renewal. Furthermore, although fear extinction increases neural activity in DS subregions, whether neural activity in DS subregions is causally involved in fear extinction is unknown. To explore the role of DS subregions in fear extinction, adult, male Long-Evans rats received microinjections of either the D1R agonist SKF38393 or a cocktail consisting of GABAA/GABAB receptor agonists muscimol/baclofen selectively into either dorsomedial (DMS) or dorsolateral (DLS) DS subregions immediately prior to fear extinction, and extinction retention and renewal were subsequently assessed drug-free. While increasing D1R signaling in the DMS during fear extinction did not impact fear extinction retention or renewal, DMS inactivation reduced later renewal. In contrast, DLS inactivation had no effect on fear extinction retention or renewal but increasing D1R signaling in the DLS during extinction reduced fear renewal. These data suggest that DMS and DLS activity during fear extinction can have opposing effects on later fear renewal, with the DMS promoting renewal and the DLS opposing renewal. Mechanisms through which the DS could influence the contextual gating of fear extinction are discussed.


Subject(s)
Corpus Striatum , Extinction, Psychological , Fear , Receptors, Dopamine D1 , Animals , Fear/physiology , Fear/drug effects , Extinction, Psychological/drug effects , Extinction, Psychological/physiology , Male , Rats , Corpus Striatum/drug effects , Corpus Striatum/physiology , Corpus Striatum/metabolism , Receptors, Dopamine D1/physiology , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D1/antagonists & inhibitors , Dopamine Agonists/pharmacology , Conditioning, Classical/drug effects , Conditioning, Classical/physiology , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/physiology , Substantia Nigra/drug effects , Substantia Nigra/physiology , Rats, Long-Evans , Dopamine/metabolism , Dopamine/physiology
10.
Sci Adv ; 10(22): eadn4203, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38809978

ABSTRACT

Learning causal relationships relies on understanding how often one event precedes another. To investigate how dopamine neuron activity and neurotransmitter release change when a retrospective relationship is degraded for a specific pair of events, we used outcome-selective Pavlovian contingency degradation in rats. Conditioned responding was attenuated for the cue-reward contingency that was degraded, as was dopamine neuron activity in the midbrain and dopamine release in the ventral striatum in response to the cue and subsequent reward. Contingency degradation also abolished the trial-by-trial history dependence of the dopamine responses at the time of trial outcome. This profile of changes in cue- and reward-evoked responding is not easily explained by a standard reinforcement learning model. An alternative model based on learning causal relationships was better able to capture dopamine responses during contingency degradation, as well as conditioned behavior following optogenetic manipulations of dopamine during noncontingent rewards. Our results suggest that mesostriatal dopamine encodes the contingencies between meaningful events during learning.


Subject(s)
Cues , Dopamine , Dopaminergic Neurons , Reward , Animals , Dopamine/metabolism , Rats , Male , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/physiology , Conditioning, Classical , Ventral Striatum/metabolism , Ventral Striatum/physiology , Learning/physiology , Mesencephalon/metabolism , Mesencephalon/physiology , Reinforcement, Psychology
11.
Proc Natl Acad Sci U S A ; 121(20): e2316658121, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38717856

ABSTRACT

Individual survival and evolutionary selection require biological organisms to maximize reward. Economic choice theories define the necessary and sufficient conditions, and neuronal signals of decision variables provide mechanistic explanations. Reinforcement learning (RL) formalisms use predictions, actions, and policies to maximize reward. Midbrain dopamine neurons code reward prediction errors (RPE) of subjective reward value suitable for RL. Electrical and optogenetic self-stimulation experiments demonstrate that monkeys and rodents repeat behaviors that result in dopamine excitation. Dopamine excitations reflect positive RPEs that increase reward predictions via RL; against increasing predictions, obtaining similar dopamine RPE signals again requires better rewards than before. The positive RPEs drive predictions higher again and thus advance a recursive reward-RPE-prediction iteration toward better and better rewards. Agents also avoid dopamine inhibitions that lower reward prediction via RL, which allows smaller rewards than before to elicit positive dopamine RPE signals and resume the iteration toward better rewards. In this way, dopamine RPE signals serve a causal mechanism that attracts agents via RL to the best rewards. The mechanism improves daily life and benefits evolutionary selection but may also induce restlessness and greed.


Subject(s)
Dopamine , Dopaminergic Neurons , Reward , Animals , Dopamine/metabolism , Dopaminergic Neurons/physiology , Dopaminergic Neurons/metabolism , Humans , Reinforcement, Psychology
12.
Nat Commun ; 15(1): 4100, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773091

ABSTRACT

In most models of neuronal plasticity and memory, dopamine is thought to promote the long-term maintenance of Long-Term Potentiation (LTP) underlying memory processes, but not the initiation of plasticity or new information storage. Here, we used optogenetic manipulation of midbrain dopamine neurons in male DAT::Cre mice, and discovered that stimulating the Schaffer collaterals - the glutamatergic axons connecting CA3 and CA1 regions - of the dorsal hippocampus concomitantly with midbrain dopamine terminals within a 200 millisecond time-window triggers LTP at glutamatergic synapses. Moreover, we showed that the stimulation of this dopaminergic pathway facilitates contextual learning in awake behaving mice, while its inhibition hinders it. Thus, activation of midbrain dopamine can operate as a teaching signal that triggers NeoHebbian LTP and promotes supervised learning.


Subject(s)
Dopamine , Dopaminergic Neurons , Hippocampus , Learning , Long-Term Potentiation , Optogenetics , Ventral Tegmental Area , Animals , Long-Term Potentiation/physiology , Ventral Tegmental Area/physiology , Male , Dopamine/metabolism , Mice , Dopaminergic Neurons/physiology , Dopaminergic Neurons/metabolism , Hippocampus/physiology , Hippocampus/metabolism , Learning/physiology , Mice, Transgenic , CA1 Region, Hippocampal/physiology , CA1 Region, Hippocampal/cytology , Synapses/physiology , Synapses/metabolism , Mice, Inbred C57BL , Memory/physiology
13.
PLoS Comput Biol ; 20(4): e1011516, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38626219

ABSTRACT

When facing an unfamiliar environment, animals need to explore to gain new knowledge about which actions provide reward, but also put the newly acquired knowledge to use as quickly as possible. Optimal reinforcement learning strategies should therefore assess the uncertainties of these action-reward associations and utilise them to inform decision making. We propose a novel model whereby direct and indirect striatal pathways act together to estimate both the mean and variance of reward distributions, and mesolimbic dopaminergic neurons provide transient novelty signals, facilitating effective uncertainty-driven exploration. We utilised electrophysiological recording data to verify our model of the basal ganglia, and we fitted exploration strategies derived from the neural model to data from behavioural experiments. We also compared the performance of directed exploration strategies inspired by our basal ganglia model with other exploration algorithms including classic variants of upper confidence bound (UCB) strategy in simulation. The exploration strategies inspired by the basal ganglia model can achieve overall superior performance in simulation, and we found qualitatively similar results in fitting model to behavioural data compared with the fitting of more idealised normative models with less implementation level detail. Overall, our results suggest that transient dopamine levels in the basal ganglia that encode novelty could contribute to an uncertainty representation which efficiently drives exploration in reinforcement learning.


Subject(s)
Basal Ganglia , Dopamine , Models, Neurological , Reward , Dopamine/metabolism , Dopamine/physiology , Uncertainty , Animals , Basal Ganglia/physiology , Exploratory Behavior/physiology , Reinforcement, Psychology , Dopaminergic Neurons/physiology , Computational Biology , Computer Simulation , Male , Algorithms , Decision Making/physiology , Behavior, Animal/physiology , Rats
14.
Neurosci Biobehav Rev ; 161: 105675, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38608828

ABSTRACT

Social behaviour is essential for animal survival, and the hypothalamic neuropeptide oxytocin (OXT) critically impacts bonding, parenting, and decision-making. Dopamine (DA), is released by ventral tegmental area (VTA) dopaminergic neurons, regulating social cues in the mesolimbic system. Despite extensive exploration of OXT and DA roles in social behaviour independently, limited studies investigate their interplay. This narrative review integrates insights from human and animal studies, particularly rodents, emphasising recent research on pharmacological manipulations of OXT or DA systems in social behaviour. Additionally, we review studies correlating social behaviour with blood/cerebral OXT and DA levels. Behavioural facets include sociability, cooperation, pair bonding and parental care. In addition, we provide insights into OXT-DA interplay in animal models of social stress, autism, and schizophrenia. Emphasis is placed on the complex relationship between the OXT and DA systems and their collective influence on social behaviour across physiological and pathological conditions. Understanding OXT and DA imbalance is fundamental for unravelling the neurobiological underpinnings of social interaction and reward processing deficits observed in psychiatric conditions.


Subject(s)
Dopamine , Oxytocin , Social Interaction , Oxytocin/metabolism , Oxytocin/physiology , Humans , Animals , Dopamine/metabolism , Social Behavior , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/physiology , Signal Transduction/physiology , Brain/metabolism , Brain/physiology
15.
Behav Neurosci ; 138(3): 212-220, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38635178

ABSTRACT

Associating a neutral conditioned stimulus (CS) with the absence of a biologically significant unconditioned stimulus (US) confers conditioned inhibitory properties upon the CS, referred to as conditioned inhibition. Conditioned inhibition and conditioned excitation, an association of a CS with the presence of the US, are fundamental components of associative learning. While the neural substrates of conditioned excitation are well established, those of conditioned inhibition remain poorly understood. Recent research has shed light on the lateral habenula (LHb) engagement in conditioned inhibition, along with the midbrain dopaminergic neurons. This article reviews behavioral tasks conducted to assess conditioned inhibition and how experimental LHb manipulations affect performance in these tasks. These results underscore the critical role of the LHb in conditioned inhibition. Intriguingly, stress increases LHb reactivity and impairs performances in tasks consisting of a component of conditioned inhibition in animals. Dysfunction of the LHb is observed in patients with depression. The ability of an organism to perform conditioned inhibition is closely linked to altered neuronal activity in the LHb, which has implications for mental disorders. (PsycInfo Database Record (c) 2024 APA, all rights reserved).


Subject(s)
Conditioning, Classical , Depression , Habenula , Habenula/physiology , Animals , Depression/physiopathology , Conditioning, Classical/physiology , Inhibition, Psychological , Humans , Dopaminergic Neurons/physiology , Association Learning/physiology
16.
Cell Rep ; 43(4): 114080, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38581677

ABSTRACT

Midbrain dopamine neurons are thought to play key roles in learning by conveying the difference between expected and actual outcomes. Recent evidence suggests diversity in dopamine signaling, yet it remains poorly understood how heterogeneous signals might be organized to facilitate the role of downstream circuits mediating distinct aspects of behavior. Here, we investigated the organizational logic of dopaminergic signaling by recording and labeling individual midbrain dopamine neurons during associative behavior. Our findings show that reward information and behavioral parameters are not only heterogeneously encoded but also differentially distributed across populations of dopamine neurons. Retrograde tracing and fiber photometry suggest that populations of dopamine neurons projecting to different striatal regions convey distinct signals. These data, supported by computational modeling, indicate that such distributional coding can maximize dynamic range and tailor dopamine signals to facilitate specialized roles of different striatal regions.


Subject(s)
Dopaminergic Neurons , Mesencephalon , Dopaminergic Neurons/physiology , Dopaminergic Neurons/metabolism , Animals , Mesencephalon/physiology , Mesencephalon/cytology , Male , Mice , Reward , Dopamine/metabolism , Association Learning/physiology , Mice, Inbred C57BL
17.
Eur J Neurosci ; 59(10): 2522-2534, 2024 May.
Article in English | MEDLINE | ID: mdl-38650479

ABSTRACT

Dopamine neurons signal the salience of environmental stimuli and influence learning, although it is less clear if these neurons also determine the salience of memories. Ventral tegmental area (VTA) dopamine neurons increase their firing in the presence of new objects and reduce it upon repeated, inconsequential exposures, marking the shift from novelty to familiarity. This study investigates how dopamine neuron activity during repeated familiar object exposure affects an animal's preference for new objects in a subsequent novel object recognition (NOR) test. We hypothesize that a single familiarization session will not sufficiently lower dopamine activity, such that the memory of a familiar object remains salient, leading to equal exploration of familiar and novel objects and weaker NOR discrimination. In contrast, multiple familiarization sessions likely suppress dopamine activity more effectively, reducing the salience of the familiar object and enhancing subsequent novelty discrimination. Our experiments in mice indicated that multiple familiarization sessions reduce VTA dopamine neuron activation, as measured by c-Fos expression, and enhance novelty discrimination compared with a single familiarization session. Dopamine neurons that show responsiveness to novelty were primarily located in the paranigral nucleus of the VTA and expressed vesicular glutamate transporter 2 transcripts, marking them as dopamine-glutamate neurons. Chemogenetic inhibition of dopamine neurons during a single session paralleled the effects of multiple sessions, improving NOR. These findings suggest that a critical role of dopamine neurons during the transition from novelty to familiarity is to modulate the salience of an object's memory.


Subject(s)
Dopaminergic Neurons , Mice, Inbred C57BL , Recognition, Psychology , Ventral Tegmental Area , Animals , Recognition, Psychology/physiology , Dopaminergic Neurons/physiology , Dopaminergic Neurons/metabolism , Ventral Tegmental Area/physiology , Mice , Male , Proto-Oncogene Proteins c-fos/metabolism , Vesicular Glutamate Transport Protein 2/metabolism , Vesicular Glutamate Transport Protein 2/genetics
18.
Neuroscience ; 547: 56-73, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38636897

ABSTRACT

Dopamine (DA) neurons of the substantia nigra (SN) and ventral tegmental area generally respond to aversive stimuli or the absence of expected rewards with transient inhibition of firing rates, which can be recapitulated with activation of the lateral habenula (LHb) and eliminated by lesioning the intermediating rostromedial tegmental nucleus (RMTg). However, a minority of DA neurons respond to aversive stimuli, such as foot shock, with a transient increase in firing rate, an outcome that rarely occurs with LHb stimulation. The degree to which individual neurons respond to these two stimulation modalities with the same response phenotype and the role of the RMTg is not known. Here, we record responses from single SN DA neurons to alternating activation of the LHb and foot shock in male rats. Lesions of the RMTg resulted in a shift away from inhibition to no response during both foot shock and LHb stimulation. Furthermore, lesions unmasked an excitatory response during LHb stimulation. The response correspondence within the same neuron between the two activation sources was no different from chance in sham controls, suggesting that external inputs rather than intrinsic DA neuronal properties are more important to response outcome. These findings contribute to a literature that shows a complex neurocircuitry underlies the regulation of DA activity and, by extension, behaviors related to learning, anhedonia, and cognition.


Subject(s)
Dopaminergic Neurons , Habenula , Substantia Nigra , Animals , Male , Habenula/physiology , Dopaminergic Neurons/physiology , Substantia Nigra/physiology , Electroshock , Action Potentials/physiology , Rats , Electric Stimulation , Rats, Sprague-Dawley , Ventral Tegmental Area/physiology
19.
Article in English | MEDLINE | ID: mdl-38498742

ABSTRACT

Depression is one of the most serious mental disorders affecting modern human life and is often caused by chronic stress. Dopamine system dysfunction is proposed to contribute to the pathophysiology of chronic stress, especially the ventral tegmental area (VTA) which mainly consists of dopaminergic neurons. Focused ultrasound stimulation (FUS) is a promising neuromodulation modality and multiple studies have demonstrated effective ultrasonic activation of cortical, subcortical, and related networks. However, the effects of FUS on the dopamine system and the potential link to chronic stress-induced depressive behaviors are relatively unknown. Here, we measured the effects of FUS targeting VTA on the improvement of depression-like behavior and evaluated the dopamine concentration in the downstream region - medial prefrontal cortex (mPFC). We found that targeting VTA FUS treatment alleviated chronic restraint stress (CRS) -induced anhedonia and despair behavior. Using an in vivo photometry approach, we analyzed the dopamine signal of mPFC and revealed a significant increase following the FUS, positively associated with the improvement of anhedonia behavior. FUS also protected the dopaminergic neurons in VTA from the damage caused by CRS exposure. Thus, these results demonstrated that targeting VTA FUS treatment significantly rescued the depressive-like behavior and declined dopamine level of mPFC induced by CRS. These beneficial effects of FUS might be due to protection in the DA neuron of VTA. Our findings suggest that FUS treatment could serve as a new therapeutic strategy for the treatment of stress-related disorders.


Subject(s)
Anhedonia , Dopamine , Humans , Prefrontal Cortex/physiology , Ventral Tegmental Area/physiology , Neurons/physiology , Dopaminergic Neurons/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...