Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.386
Filter
1.
Elife ; 132024 May 15.
Article in English | MEDLINE | ID: mdl-38748470

ABSTRACT

Acetylcholine is widely believed to modulate the release of dopamine in the striatum of mammals. Experiments in brain slices clearly show that synchronous activation of striatal cholinergic interneurons is sufficient to drive dopamine release via axo-axonal stimulation of nicotinic acetylcholine receptors. However, evidence for this mechanism in vivo has been less forthcoming. Mohebi, Collins and Berke recently reported that, in awake behaving rats, optogenetic activation of striatal cholinergic interneurons with blue light readily evokes dopamine release measured with the red fluorescent sensor RdLight1 (Mohebi et al., 2023). Here, we show that blue light alone alters the fluorescent properties of RdLight1 in a manner that may be misconstrued as phasic dopamine release, and that this artefactual photoactivation can account for the effects attributed to cholinergic interneurons. Our findings indicate that measurements of dopamine using the red-shifted fluorescent sensor RdLight1 should be interpreted with caution when combined with optogenetics. In light of this and other publications that did not observe large acetylcholine-evoked dopamine transients in vivo, the conditions under which such release occurs in behaving animals remain unknown.


Subject(s)
Cholinergic Neurons , Dopamine , Interneurons , Optogenetics , Dopamine/metabolism , Animals , Interneurons/metabolism , Interneurons/physiology , Cholinergic Neurons/metabolism , Cholinergic Neurons/physiology , Rats , Optogenetics/methods , Motivation , Nucleus Accumbens/metabolism , Nucleus Accumbens/physiology , Acetylcholine/metabolism
2.
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
3.
Prog Neurobiol ; 236: 102615, 2024 May.
Article in English | MEDLINE | ID: mdl-38641041

ABSTRACT

The gut-brain peptide ghrelin and its receptor are established as a regulator of hunger and reward-processing. However, the recently recognized ghrelin receptor inverse agonist, liver-expressed antimicrobial peptide 2 (LEAP2), is less characterized. The present study aimed to elucidate LEAP2s central effect on reward-related behaviors through feeding and its mechanism. LEAP2 was administrated centrally in mice and effectively reduced feeding and intake of palatable foods. Strikingly, LEAP2s effect on feeding was correlated to the preference of the palatable food. Further, LEAP2 reduced the rewarding memory of high preference foods, and attenuated the accumbal dopamine release associated with palatable food exposure and eating. Interestingly, LEAP2 was widely expressed in the brain, and particularly in reward-related brain areas such as the laterodorsal tegmental area (LDTg). This expression was markedly altered when allowed free access to palatable foods. Accordingly, infusion of LEAP2 into LDTg was sufficient to transiently reduce acute palatable food intake. Taken together, the present results show that central LEAP2 has a profound effect on dopaminergic reward signaling associated with food and affects several aspects of feeding. The present study highlights LEAP2s effect on reward, which may have applications for obesity and other reward-related psychiatric and neurological disorders.


Subject(s)
Dopamine , Eating , Mice, Inbred C57BL , Nucleus Accumbens , Reward , Animals , Dopamine/metabolism , Male , Nucleus Accumbens/metabolism , Nucleus Accumbens/physiology , Eating/physiology , Mice , Feeding Behavior/physiology , Blood Proteins , Antimicrobial Cationic Peptides
4.
Nat Commun ; 15(1): 3661, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38688901

ABSTRACT

Optochemistry, an emerging pharmacologic approach in which light is used to selectively activate or deactivate molecules, has the potential to alleviate symptoms, cure diseases, and improve quality of life while preventing uncontrolled drug effects. The development of in-vivo applications for optochemistry to render brain cells photoresponsive without relying on genetic engineering has been progressing slowly. The nucleus accumbens (NAc) is a region for the regulation of slow-wave sleep (SWS) through the integration of motivational stimuli. Adenosine emerges as a promising candidate molecule for activating indirect pathway neurons of the NAc expressing adenosine A2A receptors (A2ARs) to induce SWS. Here, we developed a brain-permeable positive allosteric modulator of A2ARs (A2AR PAM) that can be rapidly photoactivated with visible light (λ > 400 nm) and used it optoallosterically to induce SWS in the NAc of freely behaving male mice by increasing the activity of extracellular adenosine derived from astrocytic and neuronal activity.


Subject(s)
Adenosine , Nucleus Accumbens , Receptor, Adenosine A2A , Sleep, Slow-Wave , Animals , Nucleus Accumbens/metabolism , Nucleus Accumbens/drug effects , Nucleus Accumbens/physiology , Male , Receptor, Adenosine A2A/metabolism , Receptor, Adenosine A2A/genetics , Mice , Adenosine/metabolism , Adenosine/pharmacology , Allosteric Regulation , Sleep, Slow-Wave/physiology , Sleep, Slow-Wave/drug effects , Astrocytes/metabolism , Astrocytes/drug effects , Light , Neurons/metabolism , Neurons/drug effects , Mice, Inbred C57BL , Humans , Adenosine A2 Receptor Agonists/pharmacology
5.
Curr Biol ; 34(7): 1549-1560.e3, 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38458192

ABSTRACT

The successful pursuit of goals requires the coordinated execution and termination of actions that lead to positive outcomes. This process relies on motivational states that are guided by internal drivers, such as hunger or fear. However, the mechanisms by which the brain tracks motivational states to shape instrumental actions are not fully understood. The paraventricular nucleus of the thalamus (PVT) is a midline thalamic nucleus that shapes motivated behaviors via its projections to the nucleus accumbens (NAc)1,2,3,4,5,6,7,8 and monitors internal state via interoceptive inputs from the hypothalamus and brainstem.3,9,10,11,12,13,14 Recent studies indicate that the PVT can be subdivided into two major neuronal subpopulations, namely PVTD2(+) and PVTD2(-), which differ in genetic identity, functionality, and anatomical connectivity to other brain regions, including the NAc.4,15,16 In this study, we used fiber photometry to investigate the in vivo dynamics of these two distinct PVT neuronal types in mice performing a foraging-like behavioral task. We discovered that PVTD2(+) and PVTD2(-) neurons encode the execution and termination of goal-oriented actions, respectively. Furthermore, activity in the PVTD2(+) neuronal population mirrored motivation parameters such as vigor and satiety. Similarly, PVTD2(-) neurons also mirrored some of these parameters, but to a much lesser extent. Importantly, these features were largely preserved when activity in PVT projections to the NAc was selectively assessed. Collectively, our results highlight the existence of two parallel thalamo-striatal projections that participate in the dynamic regulation of goal pursuits and provide insight into the mechanisms by which the brain tracks motivational states to shape instrumental actions.


Subject(s)
Motivation , Nucleus Accumbens , Mice , Animals , Nucleus Accumbens/physiology , Thalamus , Midline Thalamic Nuclei/physiology , Hypothalamus
6.
Neuron ; 112(3): 333-335, 2024 Feb 07.
Article in English | MEDLINE | ID: mdl-38330898

ABSTRACT

In this issue of Neuron, Fetcho, Parekh, et al.1 show that neurons in the anterior cingulate cortex (ACC) projecting to the nucleus accumbens (NAc) are essential for integrating reward and effort evaluation in mice, and that this circuit is sensitive to exposure to stress hormones.


Subject(s)
Gyrus Cinguli , Nucleus Accumbens , Mice , Animals , Nucleus Accumbens/physiology , Gyrus Cinguli/physiology , Neurons
7.
Addict Biol ; 29(2): e13378, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38334006

ABSTRACT

Alcohol use disorder (AUD) is heritable. Thus, young adults with positive family histories represent an at-risk group relative to those without a family history, and if studied at a time when both groups have similar levels of alcohol use, it provides an opportunity to identify neural processing patterns associated with risk for AUD. Previous studies have shown that diminished response to potential reward is associated with genetic risk for AUD, but it is unclear how threat may modulate this response. We used a modified Monetary Incentive Delay task during fMRI to examine neural correlates of the interaction between threat and reward anticipation in a sample of young adults with (n = 31) and without (n = 44) family histories of harmful alcohol use. We found an interaction (p = 0.048) between cue and group in the right nucleus accumbens where the family history positive group showed less differentiation to the anticipation of gaining $5 and losing $5 relative to gaining $0. The family history-positive group also reported less excitement for trials to gain $5 relative to gaining $0 (p < 0.001). Family history-positive individuals showed less activation in the left insula during both safe and threat blocks compared to family history-negative individuals (p = 0.005), but the groups did not differ as a function of threat (p > 0.70). Young adults with, relative to without, enriched risk for AUD may have diminished reward processing via both neural and behavioural markers to potential rewarding and negative consequences. Neural response to threat may not be a contributing factor to risk at this stage.


Subject(s)
Alcoholism , Humans , Young Adult , Alcoholism/diagnostic imaging , Nucleus Accumbens/physiology , Reward , Motivation , Alcohol Drinking , Magnetic Resonance Imaging
8.
Eur J Neurosci ; 59(7): 1500-1518, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38185906

ABSTRACT

Discrete alcohol cues and contexts are relapse triggers for people with alcohol use disorder exerting particularly powerful control over behaviour when they co-occur. Here, we investigated the neural substrates subserving the capacity for alcohol-associated contexts to elevate responding to an alcohol-predictive conditioned stimulus (CS). Specifically, rats were trained in a distinct 'alcohol context' to respond by entering a fluid port during a discrete auditory CS that predicted the delivery of alcohol and were familiarized with a 'neutral context' wherein alcohol was never available. When conditioned CS responding was tested by presenting the CS without alcohol, we found that augmenting glutamatergic activity in the nucleus accumbens (NAc) shell by microinfusing α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) reduced responding to an alcohol CS in an alcohol, but not neutral, context. Further, AMPA microinfusion robustly affected behaviour, attenuating the number, duration and latency of CS responses selectively in the alcohol context. Although dopaminergic inputs to the NAc shell were previously shown to be necessary for CS responding in an alcohol context, here, chemogenetic excitation of ventral tegmental area (VTA) dopamine neurons and their inputs to the NAc shell did not affect CS responding. Critically, chemogenetic excitation of VTA dopamine neurons affected feeding behaviour and elevated c-fos immunoreactivity in the VTA and NAc shell, validating the chemogenetic approach. These findings enrich our understanding of the substrates underlying Pavlovian responding for alcohol and reveal that the capacity for contexts to modulate responding to discrete alcohol cues is delicately underpinned by the NAc shell.


Subject(s)
Cues , Nucleus Accumbens , Humans , Rats , Animals , Nucleus Accumbens/physiology , Rats, Long-Evans , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid , Ethanol/pharmacology , Conditioning, Operant/physiology
9.
Nat Commun ; 15(1): 750, 2024 Jan 29.
Article in English | MEDLINE | ID: mdl-38286800

ABSTRACT

The hippocampus is pivotal in integrating emotional processing, learning, memory, and reward-related behaviors. The dorsal hippocampus (dHPC) is particularly crucial for episodic, spatial, and associative memory, and has been shown to be necessary for context- and cue-associated reward behaviors. The nucleus accumbens (NAc), a central structure in the mesolimbic reward pathway, integrates the salience of aversive and rewarding stimuli. Despite extensive research on dHPC→NAc direct projections, their sufficiency in driving reinforcement and reward-related behavior remains to be determined. Our study establishes that activating excitatory neurons in the dHPC is sufficient to induce reinforcing behaviors through its direct projections to the dorso-medial subregion of the NAc shell (dmNAcSh). Notably, dynorphin-containing neurons specifically contribute to dHPC-driven reinforcing behavior, even though both dmNAcSh dynorphin- and enkephalin-containing neurons are activated with dHPC stimulation. Our findings unveil a pathway governing reinforcement, advancing our understanding of the hippocampal circuity's role in reward-seeking behaviors.


Subject(s)
Dynorphins , Nucleus Accumbens , Phospholipid Ethers , Nucleus Accumbens/physiology , Hippocampus/physiology , Reward , Neurons/physiology
10.
Brain Struct Funct ; 229(2): 403-429, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38193917

ABSTRACT

The influence of novelty on feeding behavior is significant and can override both homeostatic and hedonic drives due to the uncertainty of potential danger. Previous work found that novel food hypophagia is enhanced in a novel environment and that males habituate faster than females. The current study's aim was to identify the neural substrates of separate effects of food and context novelty. Adult male and female rats were tested for consumption of a novel or familiar food in either a familiar or in a novel context. Test-induced Fos expression was measured in the amygdalar, thalamic, striatal, and prefrontal cortex regions that are important for appetitive responding, contextual processing, and reward motivation. Food and context novelty induced strikingly different activation patterns. Novel context induced Fos robustly in almost every region analyzed, including the central (CEA) and basolateral complex nuclei of the amygdala, the thalamic paraventricular (PVT) and reuniens nuclei, the nucleus accumbens (ACB), the medial prefrontal cortex prelimbic and infralimbic areas, and the dorsal agranular insular cortex (AI). Novel food induced Fos in a few select regions: the CEA, anterior basomedial nucleus of the amygdala, anterior PVT, and posterior AI. There were also sex differences in activation patterns. The capsular and lateral CEA had greater activation for male groups and the anterior PVT, ACB ventral core and shell had greater activation for female groups. These activation patterns and correlations between regions, suggest that distinct functional circuitries control feeding behavior when food is novel and when eating occurs in a novel environment.


Subject(s)
Amygdala , Prefrontal Cortex , Rats , Female , Male , Animals , Prefrontal Cortex/physiology , Amygdala/physiology , Thalamus/physiology , Prosencephalon , Nucleus Accumbens/physiology
11.
Neuron ; 112(4): 611-627.e8, 2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38086372

ABSTRACT

Social animals compete for limited resources, resulting in a social hierarchy. Although different neuronal subpopulations in the medial prefrontal cortex (mPFC), which has been mechanistically implicated in social dominance behavior, encode distinct social competition behaviors, their identities and associated molecular underpinnings have not yet been identified. In this study, we found that mPFC neurons projecting to the nucleus accumbens (mPFC-NAc) encode social winning behavior, whereas mPFC neurons projecting to the ventral tegmental area (mPFC-VTA) encode social losing behavior. High-throughput single-cell transcriptomic analysis and projection-specific genetic manipulation revealed that the expression level of POU domain, class 3, transcription factor 1 (Pou3f1) in mPFC-VTA neurons controls social hierarchy. Optogenetic activation of mPFC-VTA neurons increases Pou3f1 expression and lowers social rank. Together, these data demonstrate that discrete activity and gene expression in separate mPFC projections oppositely orchestrate social competition and hierarchy.


Subject(s)
Nucleus Accumbens , Ventral Tegmental Area , Animals , Ventral Tegmental Area/physiology , Nucleus Accumbens/physiology , Social Behavior , Prefrontal Cortex/physiology , Neurons
12.
Neuron ; 112(3): 473-487.e4, 2024 Feb 07.
Article in English | MEDLINE | ID: mdl-37963470

ABSTRACT

Effort valuation-a process for selecting actions based on the anticipated value of rewarding outcomes and expectations about the work required to obtain them-plays a fundamental role in decision-making. Effort valuation is disrupted in chronic stress states and is supported by the anterior cingulate cortex (ACC), but the circuit-level mechanisms by which the ACC regulates effort-based decision-making are unclear. Here, we show that ACC neurons projecting to the nucleus accumbens (ACC-NAc) play a critical role in effort valuation behavior in mice. Activity in ACC-NAc cells integrates both reward- and effort-related information, encoding a reward-related signal that scales with effort requirements and is necessary for supporting future effortful decisions. Chronic corticosterone exposure reduces motivation, suppresses effortful reward-seeking, and disrupts ACC-NAc signals. Together, our results delineate a stress-sensitive ACC-NAc circuit that supports effortful reward-seeking behavior by integrating reward and effort signals and reinforcing effort allocation in the service of maximizing reward.


Subject(s)
Motivation , Nucleus Accumbens , Mice , Animals , Nucleus Accumbens/physiology , Reward , Neurons/physiology , Gyrus Cinguli/physiology , Decision Making/physiology
13.
J Pain ; 25(2): 331-349, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37673193

ABSTRACT

Persistent pain conditions and sleep disorders are public health problems worldwide. It is widely accepted that sleep disruption increases pain sensitivity; however, the underlying mechanisms are poorly understood. In this study, we used a protocol of 6 hours a day of total sleep deprivation for 3 days in rats to advance the understanding of these mechanisms. We focused on gender differences and the dopaminergic mesocorticolimbic system. The findings demonstrated that sleep restriction (SR) increased pain sensitivity in a similar way in males and females, without inducing a significant stress response. This pronociceptive effect depends on a nucleus accumbens (NAc) neuronal ensemble recruited during SR and on the integrity of the anterior cingulate cortex (ACC). Data on indirect dopaminergic parameters, dopamine transporter glycosylation, and dopamine and cyclic adenosine monophosphate (AMP)-regulated phosphoprotein-32 phosphorylation, as well as dopamine, serotonin, and norepinephrine levels, suggest that dopaminergic function decreases in the NAc and ACC after SR. Complementarily, pharmacological activation of dopamine D2, but not D1 receptors either in the ACC or in the NAc prevents SR from increasing pain sensitivity. The ACC and NAc are the main targets of dopaminergic mesocorticolimbic projections with a key role in pain modulation. This study showed their integrative role in the pronociceptive effect of SR, pointing to dopamine D2 receptors as a potential target for pain management in patients with sleep disorders. These findings narrow the focus of future studies on the mechanisms by which sleep impairment increases pain sensitivity. PERSPECTIVE: This study demonstrates that the pronociceptive effect of SR affects similarly males and females and depends on a NAc neuronal ensemble recruited during SR and on the integrity of the ACC. Findings on dopaminergic function support dopamine D2 receptors as targets for pain management in sleep disorders patients.


Subject(s)
Dopamine , Nucleus Accumbens , Humans , Male , Rats , Animals , Nucleus Accumbens/physiology , Dopamine/pharmacology , Gyrus Cinguli , Pain , Sleep Deprivation/complications
14.
Brain Struct Funct ; 229(1): 97-114, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37910300

ABSTRACT

The basolateral amygdala (BLA) and the nucleus accumbens core (NAcc) share some similar behavioral functions, such as associative learning, Pavlovian to instrumental transfer, and choice behavior. However, their prefrontal anatomical inputs have not been well characterized before, especially the collateral projections. In this study, we analyzed the distribution and collateralization of projections to the BLA and the NAcc from the prefrontal cortices (PFC), including the prelimbic (PL) and the infralimbic (IL) divisions of the medial prefrontal cortex (mPFC) and the subregions of the orbitofrontal cortex (OFC), such as the medial OFC (MO), the lateral OFC (LO), and the ventral OFC (VO). Double retrograde tracing approach was used, in which Cholera toxin subunit B conjugated with the Alexa Fluor 488 (CTB-AF488) or Alexa Fluor 594 (CTB-AF594) were unilaterally injected into the BLA and the NAcc, respectively, in male Long-Evans rats (n = 6). Among the sampled neurons, prefrontal projection to the BLA or the NAcc is more robust on the ipsilateral side, and more robust from the PL, the IL, and the MO compared to from the LO and the VO. The majority of the projections from the PFC to the BLA and/or the NAcc are confined in deep layer. In addition, for each of the prefrontal areas, about 15-25% BLA-projecting neurons send collateral projections to the NAcc, and vice versa. In conclusion, our data suggested that prefrontal control over the BLA and the NAcc is not entirely independent. The functional importance of the collateral projections awaits further examination.


Subject(s)
Basolateral Nuclear Complex , Nucleus Accumbens , Rats , Male , Animals , Nucleus Accumbens/physiology , Rats, Long-Evans , Prefrontal Cortex/physiology , Cerebral Cortex
15.
Nat Methods ; 21(4): 680-691, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38036855

ABSTRACT

Dopamine (DA) plays multiple roles in a wide range of physiological and pathological processes via a large network of dopaminergic projections. To dissect the spatiotemporal dynamics of DA release in both dense and sparsely innervated brain regions, we developed a series of green and red fluorescent G-protein-coupled receptor activation-based DA (GRABDA) sensors using a variety of DA receptor subtypes. These sensors have high sensitivity, selectivity and signal-to-noise ratio with subsecond response kinetics and the ability to detect a wide range of DA concentrations. We then used these sensors in mice to measure both optogenetically evoked and behaviorally relevant DA release while measuring neurochemical signaling in the nucleus accumbens, amygdala and cortex. Using these sensors, we also detected spatially resolved heterogeneous cortical DA release in mice performing various behaviors. These next-generation GRABDA sensors provide a robust set of tools for imaging dopaminergic activity under a variety of physiological and pathological conditions.


Subject(s)
Dopamine , Nucleus Accumbens , Mice , Animals , Nucleus Accumbens/physiology , Receptors, Dopamine , Brain , Receptors, G-Protein-Coupled
16.
Neuron ; 112(3): 500-514.e5, 2024 Feb 07.
Article in English | MEDLINE | ID: mdl-38016471

ABSTRACT

Striatal dopamine (DA) release has long been linked to reward processing, but it remains controversial whether DA release reflects costs or benefits and how these signals vary with motivation. Here, we measure DA release in the nucleus accumbens (NAc) and dorsolateral striatum (DLS) while independently varying costs and benefits and apply behavioral economic principles to determine a mouse's level of motivation. We reveal that DA release in both structures incorporates both reward magnitude and sunk cost. Surprisingly, motivation was inversely correlated with reward-evoked DA release. Furthermore, optogenetically evoked DA release was also heavily dependent on sunk cost. Our results reconcile previous disparate findings by demonstrating that striatal DA release simultaneously encodes cost, benefit, and motivation but in distinct manners over different timescales. Future work will be necessary to determine whether the reduction in phasic DA release in highly motivated animals is due to changes in tonic DA levels.


Subject(s)
Dopamine , Motivation , Mice , Animals , Dopamine/physiology , Corpus Striatum/physiology , Neostriatum , Nucleus Accumbens/physiology , Reward
17.
Article in English | MEDLINE | ID: mdl-37871776

ABSTRACT

BACKGROUND: Childhood maltreatment is associated with reduced activation of the nucleus accumbens, a central region in the reward network, and overactivity in the amygdala, a key region in threat processing. However, the long-lasting impact of these associations in the context of later-life stress is not well understood. The current study explored the association between childhood threat and deprivation and functional connectivity of threat and reward regions in an adult trauma sample. METHODS: Trauma survivors (N = 169; mean age [SD] = 32.2 [10.3] years; female = 55.6%) were recruited from a level I trauma center. Two weeks after injury, participants completed the Childhood Trauma Questionnaire (measuring experiences of threat and deprivation) and underwent resting-state functional magnetic resonance imaging. Seed-to-voxel analyses evaluated the effect of childhood threat and deprivation on amygdala and nucleus accumbens resting-state connectivity. RESULTS: Higher levels of threat were associated with increased connectivity between the right nucleus accumbens with temporal fusiform gyrus/parahippocampal gyrus and the left amygdala and the precuneus (false discovery rate-corrected p < .05). After controlling for posttraumatic symptoms 2 weeks posttrauma and lifetime trauma exposure, only the nucleus accumbens findings survived. There were no significant relationships between experiences of childhood deprivation and amygdala or nucleus accumbens connectivity. CONCLUSIONS: Experiences of threat are associated with increased nucleus accumbens and amygdala connectivity, which may reflect a preparedness to detect salient and visual stimuli. This may also reflect a propensity toward dysregulated reward processing. Overall, these results suggest that childhood threat may be contributing to aberrant neural baseline reward and threat sensitivity later in life in an adult trauma sample.


Subject(s)
Magnetic Resonance Imaging , Nucleus Accumbens , Psychological Tests , Self Report , Humans , Adult , Female , Child , Nucleus Accumbens/physiology , Amygdala , Reward
18.
Steroids ; 201: 109344, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37979822

ABSTRACT

The striatal brain regions encompassing the nucleus accumbens core (NAcc), shell (NAcs) and caudate-putamen (CPu) regulate cognitive functions including motivated behaviors, habit, learning, and sensorimotor action, among others. Sex steroid hormone sensitivity and sex differences have been documented in all of these functions in both normative and pathological contexts, including anxiety, depression and addiction. The neurotransmitter glutamate has been implicated in regulating these behaviors as well as striatal physiology, and there are likewise documented sex differences in glutamate action upon the striatal output neurons, the medium spiny neurons (MSNs). Here we review the available data regarding the role of steroid sex hormones such as 17ß-estradiol (estradiol), progesterone, and testosterone in rapidly modulating MSN glutamatergic synapse properties, presented in the context of the estrous cycle as appropriate. Estradiol action upon glutamatergic synapse properties in female NAcc MSNs is most comprehensively discussed. In the female NAcc, MSNs exhibit development period-specific sex differences and estrous cycle variations in glutamatergic synapse properties as shown by multiple analyses, including that of miniature excitatory postsynaptic currents (mEPSCs). Estrous cycle-differences in NAcc MSN mEPSCs can be mimicked by acute exposure to estradiol or an ERα agonist. The available evidence, or lack thereof, is also discussed concerning estrogen action upon MSN glutamatergic synapse in the other striatal regions as well as the underexplored roles of progesterone and testosterone. We conclude that there is strong evidence regarding estradiol action upon glutamatergic synapse function in female NAcs MSNs and call for more research regarding other hormones and striatal regions.


Subject(s)
Nucleus Accumbens , Progesterone , Female , Humans , Male , Brain , Estradiol/pharmacology , Estrous Cycle , Glutamates , Nucleus Accumbens/physiology , Putamen/chemistry , Synapses , Testosterone , Caudate Nucleus/chemistry , Caudate Nucleus/physiology
19.
Neuron ; 112(5): 772-785.e9, 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38141605

ABSTRACT

Lack of behavioral suppression typifies substance use disorders, yet the neural circuit underpinnings of drug-induced behavioral disinhibition remain unclear. Here, we employ deep-brain two-photon calcium imaging in heroin self-administering mice, longitudinally tracking adaptations within a paraventricular thalamus to nucleus accumbens behavioral inhibition circuit from the onset of heroin use to reinstatement. We find that select thalamo-accumbal neuronal ensembles become profoundly hypoactive across the development of heroin seeking and use. Electrophysiological experiments further reveal persistent adaptations at thalamo-accumbal parvalbumin interneuronal synapses, whereas functional rescue of these synapses prevents multiple triggers from initiating reinstatement of heroin seeking. Finally, we find an enrichment of µ-opioid receptors in output- and cell-type-specific paraventricular thalamic neurons, which provide a mechanism for heroin-induced synaptic plasticity and behavioral disinhibition. These findings reveal key circuit adaptations that underlie behavioral disinhibition in opioid dependence and further suggest that recovery of this system would reduce relapse susceptibility.


Subject(s)
Heroin , Opioid-Related Disorders , Rats , Mice , Animals , Heroin/pharmacology , Rats, Sprague-Dawley , Self Administration/methods , Neurons , Nucleus Accumbens/physiology
20.
Neuron ; 112(5): 835-849.e7, 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38134921

ABSTRACT

At the core of value-based learning is the nucleus accumbens (NAc). D1- and D2-receptor-containing medium spiny neurons (MSNs) in the NAc core are hypothesized to have opposing valence-based roles in behavior. Using optical imaging and manipulation approaches in mice, we show that neither D1 nor D2 MSNs signal valence. D1 MSN responses were evoked by stimuli regardless of valence or contingency. D2 MSNs were evoked by both cues and outcomes, were dynamically changed with learning, and tracked valence-free prediction error at the population and individual neuron level. Finally, D2 MSN responses to cues were necessary for associative learning. Thus, D1 and D2 MSNs work in tandem, rather than in opposition, by signaling specific properties of stimuli to control learning.


Subject(s)
Medium Spiny Neurons , Receptors, Dopamine D1 , Mice , Animals , Mice, Transgenic , Receptors, Dopamine D1/metabolism , Nucleus Accumbens/physiology , Neurons/physiology , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...