51
|
Fleischer M, Endres H, Sendtner M, Volkmann J. Development of a Fully Implantable Stimulator for Deep Brain Stimulation in Mice. Front Neurosci 2020; 14:726. [PMID: 32792895 PMCID: PMC7385305 DOI: 10.3389/fnins.2020.00726] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 06/18/2020] [Indexed: 11/29/2022] Open
Abstract
INTRODUCTION Deep brain stimulation is an established method for the treatment of neurological and psychiatric disorders. To elicit the underlying mechanisms and explore new stimulation targets, rodent models are necessary. Cable bound external stimulation or portable devices limit movement of the animals and influence behavioral experiments. Therefore, implantable, individually programmed devices are required. EXPERIMENTAL PROCEDURE The stimulator consists of an 8bit-microcontroller mounted on a square electrical board (10 × 10 mm). External control is enabled by a magnetic reed contact, as running control serves a white LED, running modes are displayed by flash codes. Stimulation parameters could be programmed in the range of pulse width: 60-500 μs, amplitude: up to 300 μA and frequency: 10-500 Hz. Power is supplied by two standard batteries. The device was implanted in 8-10 weeks old BALBc-mice. Functionality was examined by electrical stimulation of nucleus accumbens area with standard parameters for mice and determination of c-fos levels in vitro in brain slices. RESULTS The implanted microstimulators were well-tolerated by the mice, without impairment of free movement. Coating, external control, and monitoring of function with LED flash code proved to be fully adequate. Stimulation with standard stimulating parameters of nucleus accumbens elicited strong c-fos elevation on simulation site. CONCLUSION We present a fully implantable stimulator for freely moving mice that meets the urgent need for further research on the effects of deep brain stimulation in rodent models. It offers the possibility to conduct behavioral experiments for up to 30 days of stimulation.
Collapse
Affiliation(s)
- Michael Fleischer
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany
- Department of Neurology, Essen University Hospital, Essen, Germany
| | - Heinz Endres
- University of Applied Science Würzburg-Schweinfurt, Schweinfurt, Germany
| | - Michael Sendtner
- Institute of Clinical Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Jens Volkmann
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
52
|
Collins AL, Saunders BT. Heterogeneity in striatal dopamine circuits: Form and function in dynamic reward seeking. J Neurosci Res 2020; 98:1046-1069. [PMID: 32056298 PMCID: PMC7183907 DOI: 10.1002/jnr.24587] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 01/08/2020] [Accepted: 01/16/2020] [Indexed: 01/03/2023]
Abstract
The striatal dopamine system has long been studied in the context of reward learning, motivation, and movement. Given the prominent role dopamine plays in a variety of adaptive behavioral states, as well as diseases like addiction, it is essential to understand the full complexity of dopamine neurons and the striatal systems they target. A growing number of studies are uncovering details of the heterogeneity in dopamine neuron subpopulations. Here, we review that work to synthesize current understanding of dopamine system heterogeneity across three levels, anatomical organization, functions in behavior, and modes of action, wherein we focus on signaling profiles and local mechanisms for modulation of dopamine release. Together, these studies reveal new and emerging dimensions of the striatal dopamine system, informing its contribution to dynamic motivational and decision-making processes.
Collapse
Affiliation(s)
- Anne L. Collins
- University of Minnesota, Department of Neuroscience, Medical Discovery Team on Addiction, Minneapolis, MN 55455
| | - Benjamin T. Saunders
- University of Minnesota, Department of Neuroscience, Medical Discovery Team on Addiction, Minneapolis, MN 55455
| |
Collapse
|
53
|
Halladay LR, Kocharian A, Piantadosi PT, Authement ME, Lieberman AG, Spitz NA, Coden K, Glover LR, Costa VD, Alvarez VA, Holmes A. Prefrontal Regulation of Punished Ethanol Self-administration. Biol Psychiatry 2020; 87:967-978. [PMID: 31937415 PMCID: PMC7217757 DOI: 10.1016/j.biopsych.2019.10.030] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 10/08/2019] [Accepted: 10/25/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND A clinical hallmark of alcohol use disorder is persistent drinking despite potential adverse consequences. The ventromedial prefrontal cortex (vmPFC) and dorsomedial prefrontal cortex (dmPFC) are positioned to exert top-down control over subcortical regions, such as the nucleus accumbens shell (NAcS) and basolateral amygdala, which encode positive and negative valence of ethanol (EtOH)-related stimuli. Prior rodent studies have implicated these regions in regulation of punished EtOH self-administration (EtOH-SA). METHODS We conducted in vivo electrophysiological recordings in mouse vmPFC and dmPFC to obtain neuronal correlates of footshock-punished EtOH-SA. Ex vivo recordings were performed in NAcS D1 receptor-expressing medium spiny neurons receiving vmPFC input to examine punishment-related plasticity in this pathway. Optogenetic photosilencing was employed to assess the functional contribution of the vmPFC, dmPFC, vmPFC projections to NAcS, or vmPFC projections to basolateral amygdala, to punished EtOH-SA. RESULTS Punishment reduced EtOH lever pressing and elicited aborted presses (lever approach followed by rapid retraction). Neurons in the vmPFC and dmPFC exhibited phasic firing to EtOH lever presses and aborts, but only in the vmPFC was there a population-level shift in coding from lever presses to aborts with punishment. Closed-loop vmPFC, but not dmPFC, photosilencing on a postpunishment probe test negated the reduction in EtOH lever presses but not in aborts. Punishment was associated with altered plasticity at vmPFC inputs to D1 receptor-expressing medium spiny neurons in the NAcS. Photosilencing vmPFC projections to the NAcS, but not to the basolateral amygdala, partially reversed suppression of EtOH lever presses on probe testing. CONCLUSIONS These findings demonstrate a key role for the vmPFC in regulating EtOH-SA after punishment, with implications for understanding the neural basis of compulsive drinking in alcohol use disorder.
Collapse
Affiliation(s)
- Lindsay R Halladay
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland; Department of Psychology, Santa Clara University, Santa Clara, California.
| | - Adrina Kocharian
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Patrick T Piantadosi
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland; Center on Compulsive Behaviors, Intramural Research Program, National Institutes of Health, Bethesda, Maryland
| | - Michael E Authement
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland; Center on Compulsive Behaviors, Intramural Research Program, National Institutes of Health, Bethesda, Maryland
| | - Abby G Lieberman
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Nathen A Spitz
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Kendall Coden
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Lucas R Glover
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Vincent D Costa
- Department of Behavioral Neuroscience, Oregon Health Sciences University, Portland, Oregon
| | - Veronica A Alvarez
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland; Center on Compulsive Behaviors, Intramural Research Program, National Institutes of Health, Bethesda, Maryland
| | - Andrew Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
54
|
ErbB4 Null Mice Display Altered Mesocorticolimbic and Nigrostriatal Dopamine Levels as well as Deficits in Cognitive and Motivational Behaviors. eNeuro 2020; 7:ENEURO.0395-19.2020. [PMID: 32354758 PMCID: PMC7242816 DOI: 10.1523/eneuro.0395-19.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 02/26/2020] [Accepted: 02/27/2020] [Indexed: 12/17/2022] Open
Abstract
Natural genetic variants of Neuregulin1 (NRG1) and its cognate receptor ErbB4 are associated with a risk for schizophrenia. Whereas most studies on NRG1-ErbB4 signaling have focused on GABAergic interneurons, ErbB4 is also expressed by midbrain dopaminergic neurons where it modulates extracellular dopamine (DA) levels. Here, we report that extracellular steady-state levels of DA are reduced in the medial prefrontal cortex (mPFC; −65%), hippocampus (−53%) and nucleus accumbens (NAc; −35%), but are elevated in the dorsal striatum (+25%) of ErbB4 knock-out mice (ErbB4 KOs) relative to wild-type controls. This pattern of DA imbalance recapitulates the reported prefrontal cortical reduction and striatal increase of DA levels in schizophrenia patients. Next, we report on a battery of behavioral tasks used to evaluate locomotor, cognitive and motivational behaviors in ErbB4 KOs relative to controls. We found that ErbB4 KOs are hyperactive in a novel open field but not in their familiar home cage, are more sensitive to amphetamine, perform poorly in the T-maze and novel object recognition (NOR) tasks, exhibit reduced spatial learning and memory on the Barnes maze, and perform markedly worse in conditioned place preference (CPP) tasks when associating cued-reward palatable food with location. However, we found that the poor performance of ErbB4 KOs in CPP are likely due to deficits in spatial memory, instead of reward seeking, as ErbB4 KOs are more motivated to work for palatable food rewards. Our findings indicate that ErbB4 signaling affects tonic DA levels and modulates a wide array of behavioral deficits relevant to psychiatric disorders, including schizophrenia.
Collapse
|
55
|
Kokane SS, Perrotti LI. Sex Differences and the Role of Estradiol in Mesolimbic Reward Circuits and Vulnerability to Cocaine and Opiate Addiction. Front Behav Neurosci 2020; 14:74. [PMID: 32508605 PMCID: PMC7251038 DOI: 10.3389/fnbeh.2020.00074] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 04/22/2020] [Indexed: 12/15/2022] Open
Abstract
Although both men and women become addicted to drugs of abuse, women transition to addiction faster, experience greater difficulties remaining abstinent, and relapse more often than men. In both humans and rodents, hormonal cycles are associated with females' faster progression to addiction. Higher concentrations and fluctuating levels of ovarian hormones in females modulate the mesolimbic reward system and influence reward-directed behavior. For example, in female rodents, estradiol (E2) influences dopamine activity within the mesolimbic reward system such that drug-directed behaviors that are normally rewarding and reinforcing become enhanced when circulating levels of E2 are high. Therefore, neuroendocrine interactions, in part, explain sex differences in behaviors motivated by drug reward. Here, we review sex differences in the physiology and function of the mesolimbic reward system in order to explore the notion that sex differences in response to drugs of abuse, specifically cocaine and opiates, are the result of molecular neuroadaptations that differentially develop depending upon the hormonal state of the animal. We also reconsider the notion that ovarian hormones, specifically estrogen/estradiol, sensitize target neurons thereby increasing responsivity when under the influence of either cocaine or opiates or in response to exposure to drug-associated cues. These adaptations may ultimately serve to guide the motivational behaviors that underlie the factors that cause women to be more vulnerable to cocaine and opiate addiction than men.
Collapse
Affiliation(s)
- Saurabh S Kokane
- Department of Psychology, University of Texas at Arlington, Arlington, TX, United States
| | - Linda I Perrotti
- Department of Psychology, University of Texas at Arlington, Arlington, TX, United States
| |
Collapse
|
56
|
Berthoud HR, Morrison CD, Münzberg H. The obesity epidemic in the face of homeostatic body weight regulation: What went wrong and how can it be fixed? Physiol Behav 2020; 222:112959. [PMID: 32422162 DOI: 10.1016/j.physbeh.2020.112959] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 04/23/2020] [Accepted: 05/04/2020] [Indexed: 12/14/2022]
Abstract
Ever since the pioneering discoveries in the mid nineteen hundreds, the hypothalamus was recognized as a crucial component of the neural system controlling appetite and energy balance. The new wave of neuron-specific research tools has confirmed this key role of the hypothalamus and has delineated many other brain areas to be part of an expanded neural system sub serving these crucial functions. However, despite significant progress in defining this complex neural circuitry, many questions remain. One of the key questions is why the sophisticated body weight regulatory system is unable to prevent the rampant obesity epidemic we are experiencing. Why are pathologically obese body weight levels defended, and what can we do about it? Here we try to find answers to these questions by 1) reminding the reader that the neural controls of ingestive behavior have evolved in a demanding, restrictive environment and encompass much of the brain's major functions, far beyond the hypothalamus and brainstem, 2) hypothesizing that the current obesogenic environment impinges mainly on a critical pathway linking hypothalamic areas with the motivational and reward systems to produce uncompensated hyperphagia, and 3) proposing adequate strategies for prevention and treatment.
Collapse
Affiliation(s)
- Hans-Rudolf Berthoud
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA.
| | - Christopher D Morrison
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - Heike Münzberg
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| |
Collapse
|
57
|
Aggarwal M, Akamine Y, Liu AW, Wickens JR. The nucleus accumbens and inhibition in the ventral tegmental area play a causal role in the Kamin blocking effect. Eur J Neurosci 2020; 52:3087-3109. [DOI: 10.1111/ejn.14732] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 03/04/2020] [Accepted: 03/26/2020] [Indexed: 01/02/2023]
Affiliation(s)
- Mayank Aggarwal
- Neurobiology Research Unit Okinawa Institute of Science and Technologys Graduate University Kunigami Okinawa Japan
| | - Yumiko Akamine
- Neurobiology Research Unit Okinawa Institute of Science and Technologys Graduate University Kunigami Okinawa Japan
| | - Andrew W. Liu
- Neurobiology Research Unit Okinawa Institute of Science and Technologys Graduate University Kunigami Okinawa Japan
| | - Jeffery R. Wickens
- Neurobiology Research Unit Okinawa Institute of Science and Technologys Graduate University Kunigami Okinawa Japan
| |
Collapse
|
58
|
Baladron J, Hamker FH. Habit learning in hierarchical cortex-basal ganglia loops. Eur J Neurosci 2020; 52:4613-4638. [PMID: 32237250 DOI: 10.1111/ejn.14730] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/21/2020] [Accepted: 03/22/2020] [Indexed: 12/17/2022]
Abstract
How do the multiple cortico-basal ganglia-thalamo-cortical loops interact? Are they parallel and fully independent or controlled by an arbitrator, or are they hierarchically organized? We introduce here a set of four key concepts, integrated and evaluated by means of a neuro-computational model, that bring together current ideas regarding cortex-basal ganglia interactions in the context of habit learning. According to key concept 1, each loop learns to select an intermediate objective at a different abstraction level, moving from goals in the ventral striatum to motor in the putamen. Key concept 2 proposes that the cortex integrates the basal ganglia selection with environmental information regarding the achieved objective. Key concept 3 claims shortcuts between loops, and key concept 4 predicts that loops compute their own prediction error signal for learning. Computational benefits of the key concepts are demonstrated. Contrasting with former concepts of habit learning, the loops collaborate to select goal-directed actions while training slower shortcuts develops habitual responses.
Collapse
Affiliation(s)
- Javier Baladron
- Department of Computer Science, Chemnitz University of Technology, Chemnitz, Germany
| | - Fred H Hamker
- Department of Computer Science, Chemnitz University of Technology, Chemnitz, Germany
| |
Collapse
|
59
|
Carr KD. Homeostatic regulation of reward via synaptic insertion of calcium-permeable AMPA receptors in nucleus accumbens. Physiol Behav 2020; 219:112850. [PMID: 32092445 PMCID: PMC7108974 DOI: 10.1016/j.physbeh.2020.112850] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 01/23/2020] [Accepted: 02/18/2020] [Indexed: 10/25/2022]
Abstract
The incentive effects of food and related cues are determined by stimulus properties and the internal state of the organism. Enhanced hedonic reactivity and incentive motivation in energy deficient subjects have been demonstrated in animal models and humans. Defining the neurobiological underpinnings of these state-based modulatory effects could illuminate fundamental mechanisms of adaptive behavior, as well as provide insight into maladaptive consequences of weight loss dieting and the relationship between disturbed eating behavior and substance abuse. This article summarizes research of our laboratory aimed at identifying neuroadaptations induced by chronic food restriction (FR) that increase the reward magnitude of drugs and associated cues. The main findings are that FR decreases basal dopamine (DA) transmission, upregulates signaling downstream of the D1 DA receptor (D1R), and triggers synaptic incorporation of calcium-permeable AMPA receptors (CP-AMPARs) in the nucleus accumbens (NAc). Selective antagonism of CP-AMPARs decreases excitatory postsynaptic currents in NAc medium spiny neurons of FR rats and blocks the enhanced rewarding effects of d-amphetamine and a D1R, but not a D2R, agonist. These results suggest that FR drives CP-AMPARs into the synaptic membrane of D1R-expressing MSNs, possibly as a homeostatic response to reward loss. FR subjects also display diminished aversion for contexts associated with LiCl treatment and centrally infused cocaine. An encompassing, though speculative, hypothesis is that NAc synaptic incorporation of CP-AMPARs in response to food scarcity and other forms of sustained reward loss adaptively increases incentive effects of reward stimuli and, at the same time, diminishes responsiveness to aversive stimuli that have potential to interfere with goal pursuit.
Collapse
Affiliation(s)
- Kenneth D Carr
- Departments of Psychiatry and Biochemistry and Molecular Pharmacology, New York University School of Medicine, 435 East 30th Street, New York, NY 10016, United States.
| |
Collapse
|
60
|
Prelimbic and Infralimbic Prefrontal Regulation of Active and Inhibitory Avoidance and Reward-Seeking. J Neurosci 2020; 40:4773-4787. [PMID: 32393535 DOI: 10.1523/jneurosci.0414-20.2020] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/23/2020] [Accepted: 05/03/2020] [Indexed: 11/21/2022] Open
Abstract
Flexible initiation or suppression of actions to avoid aversive events is crucial for survival. The prelimbic (PL) and infralimbic (IL) regions of the medial prefrontal cortex (mPFC) have been implicated in different aspects of avoidance and reward-seeking, but their respective contribution in instigating versus suppressing actions in aversive contexts remains to be clarified. We examined mPFC involvement in different forms of avoidance in rats well trained on different cued lever-press avoidance tasks. Active/inhibitory avoidance required flexible discrimination between auditory cues signaling foot-shock could be avoided by making or withholding instrumental responses. On a simpler active avoidance task, a single cue signaled when a lever press would avoid shock. PL inactivation disrupted active but not inhibitory avoidance on the discriminative task while having no effect on single-cued avoidance. In comparison, IL inactivation broadly impaired active and inhibitory avoidance. Conversely, on a cued appetitive go/no-go task, both IL and PL inactivation impaired inhibitory but not active reward-seeking, the latter effect being diametrically opposite to that observed on the avoidance task. These findings highlight the complex manner in which different mPFC regions aid in initiating or inhibiting actions in the service of avoiding aversive outcomes or obtaining rewarding ones. IL facilitates active avoidance but suppress inappropriate actions in appetitive and aversive contexts. In contrast, contextual valence plays a critical role in how the PL is recruited in initiating or suppressing actions, which may relate to the degree of cognitive control required to flexibly negotiate response or motivational conflicts and override prepotent behaviors.SIGNIFICANCE STATEMENT Choosing to make or withhold actions in a context-appropriate manner to avoid aversive events or obtain other goals is a critical survival skill. Different medial prefrontal cortex (mPFC) regions have been implicated in certain aspects of avoidance, but their contributions to instigating or suppressing actions remains to be clarified. Here, we show that the dorsal, prelimbic (PL) region of the medial PFC aids active avoidance in situations requiring flexible mitigation of response conflicts, but also aids in withholding responses to obtain rewards. In comparison the ventral infralimbic (IL) cortex plays a broader role in active and inhibitory avoidance as well as suppressing actions to obtain rewards. These findings provide insight into mechanisms underlying normal and maladaptive avoidance behaviors and response inhibition.
Collapse
|
61
|
Acute Stress Enhances Associative Learning via Dopamine Signaling in the Ventral Lateral Striatum. J Neurosci 2020; 40:4391-4400. [PMID: 32321745 DOI: 10.1523/jneurosci.3003-19.2020] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/17/2020] [Accepted: 04/08/2020] [Indexed: 01/02/2023] Open
Abstract
Acute stress transiently increases vigilance, enhancing the detection of salient stimuli in one's environment. This increased perceptual sensitivity is thought to promote the association of rewarding outcomes with relevant cues. The mesolimbic dopamine system is critical for learning cue-reward associations. Dopamine levels in the ventral striatum are elevated following exposure to stress. Together, this suggests that the mesolimbic dopamine system could mediate the influence of acute stress on cue-reward learning. To address this possibility, we examined how a single stressful experience influenced learning in an appetitive pavlovian conditioning task. Male rats underwent an episode of restraint prior to the first conditioning session. This acute stress treatment augmented conditioned responding in subsequent sessions. Voltammetry recordings of mesolimbic dopamine levels demonstrated that acute stress selectively increased reward-evoked dopamine release in the ventral lateral striatum (VLS), but not in the ventral medial striatum. Antagonizing dopamine receptors in the VLS blocked the stress-induced enhancement of conditioned responding. Collectively, these findings illustrate that stress engages dopamine signaling in the VLS to facilitate appetitive learning.SIGNIFICANCE STATEMENT Acute stress influences learning about aversive and rewarding outcomes. Dopamine neurons are sensitive to stress and critical for reward learning. However, it is unclear whether stress regulates reward learning via dopamine signaling. Using fast-scan cyclic voltammetry as rats underwent pavlovian conditioning, we demonstrate that a single stressful experience increases reward-evoked dopamine release in the ventral lateral striatum. This enhanced dopamine signal accompanies a long-lasting increase in conditioned behavioral responding. These findings highlight that the ventral lateral striatum is a node for mediating the effect of stress on reward processing.
Collapse
|
62
|
Loss of nucleus accumbens low-frequency fluctuations is a signature of chronic pain. Proc Natl Acad Sci U S A 2020; 117:10015-10023. [PMID: 32312809 PMCID: PMC7211984 DOI: 10.1073/pnas.1918682117] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The prevalence of chronic pain has reached epidemic levels. In addition to personal suffering, chronic pain is associated with psychiatric and medical comorbidities, notably substance misuse. Chronic pain does not have a cure or quantitative diagnostic or prognostic tools. Here we show that brain imaging can provide such measures. First, we show that the brain limbic system of patients with subacute back pain at risk for becoming chronic back pain patients exhibits limbic system structural alterations, which predate the onset of chronic pain. Second, we show that the nucleus accumbens activity shows loss of low-frequency fluctuations only when patients transition to the chronic phase, an observation that was reproduced in multiple datasets collected at different sites. Chronic pain is a highly prevalent disease with poorly understood pathophysiology. In particular, the brain mechanisms mediating the transition from acute to chronic pain remain largely unknown. Here, we identify a subcortical signature of back pain. Specifically, subacute back pain patients who are at risk for developing chronic pain exhibit a smaller nucleus accumbens volume, which persists in the chronic phase, compared to healthy controls. The smaller accumbens volume was also observed in a separate cohort of chronic low-back pain patients and was associated with dynamic changes in functional connectivity. At baseline, subacute back pain patients showed altered local nucleus accumbens connectivity between putative shell and core, irrespective of the risk of transition to chronic pain. At follow-up, connectivity changes were observed between nucleus accumbens and rostral anterior cingulate cortex in the patients with persistent pain. Analysis of the power spectral density of nucleus accumbens resting-state activity in the subacute and chronic back pain patients revealed loss of power in the slow-5 frequency band (0.01 to 0.027 Hz) which developed only in the chronic phase of pain. This loss of power was reproducible across two cohorts of chronic low-back pain patients obtained from different sites and accurately classified chronic low-back pain patients in two additional independent datasets. Our results provide evidence that lower nucleus accumbens volume confers risk for developing chronic pain and altered nucleus accumbens activity is a signature of the state of chronic pain.
Collapse
|
63
|
Ma L, Chen W, Yu D, Han Y. Brain-Wide Mapping of Afferent Inputs to Accumbens Nucleus Core Subdomains and Accumbens Nucleus Subnuclei. Front Syst Neurosci 2020; 14:15. [PMID: 32317941 PMCID: PMC7150367 DOI: 10.3389/fnsys.2020.00015] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 03/02/2020] [Indexed: 12/20/2022] Open
Abstract
The nucleus accumbens (NAc) is the ventral part of the striatum and the interface between cognition, emotion, and action. It is composed of three major subnuclei: i.e., NAc core (NAcC), lateral shell (NAcLS), and medial shell (NAcMS), which exhibit functional heterogeneity. Thus, determining the synaptic inputs of the subregions of the NAc is important for understanding the circuit mechanisms involved in regulating different functions. Here, we simultaneously labeled subregions of the NAc with cholera toxin subunit B conjugated with multicolor Alexa Fluor, then imaged serial sections of the whole brain with a fully automated slide scanning system. Using the interactive WholeBrain framework, we characterized brain-wide inputs to the NAcC subdomains, including the rostral, caudal, dorsal, and ventral subdomains (i.e., rNAcC, cNAcC, dNAcC, and vNAcC, respectively) and the NAc subnuclei. We found diverse brain regions, distributed from the cerebrum to brain stem, projecting to the NAc. Of the 57 brain regions projecting to the NAcC, the anterior olfactory nucleus (AON) exhibited the greatest inputs. The input neurons of rNAcC and cNAcC are two distinct populations but share similar distribution over the same upstream brain regions, whereas the input neurons of dNAcC and vNAcC exhibit slightly different distributions over the same upstream regions. Of the 55 brain regions projecting to the NAcLS, the piriform area contributed most of the inputs. Of the 72 brain regions projecting to the NAcMS, the lateral septal nucleus contributed most of the inputs. The input neurons of NAcC and NAcLS share similar distributions, whereas the NAcMS exhibited brain-wide distinct distribution. Thus, the NAcC subdomains appeared to share the same upstream brain regions, although with distinct input neuron populations and slight differences in the input proportions, whereas the NAcMS subnuclei received distinct inputs from multiple upstream brain regions. These results lay an anatomical foundation for understanding the different functions of NAcC subdomains and NAc subnuclei.
Collapse
Affiliation(s)
- Liping Ma
- Department of Neurobiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Wenqi Chen
- Department of Neurobiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Danfang Yu
- Department of Neurobiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China.,Department of Neurology, Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, China
| | - Yunyun Han
- Department of Neurobiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China.,Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
64
|
Su J, Li Z, Yamashita A, Kusumoto-Yoshida I, Isomichi T, Hao L, Kuwaki T. Involvement of the Nucleus Accumbens in Chocolate-induced Cataplexy. Sci Rep 2020; 10:4958. [PMID: 32188934 PMCID: PMC7080740 DOI: 10.1038/s41598-020-61823-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 03/04/2020] [Indexed: 11/09/2022] Open
Abstract
Happiness is key for both mental and physical well-being. To further understand the brain mechanisms involved, we utilized the cataplexy that occurs in narcoleptic animal models as a quantitative behavioral measure because it is triggered by actions associated with happiness, such as laughter in humans and palatable foods in mice. Here we report that the rostral part of the nucleus accumbens (NAc) shell is strongly activated during the beginning of chocolate-induced cataplexy in orexin neuron-ablated mice. We made a local lesion in the NAc using ibotenic acid and observed the animals' behavior. The number of cataplexy bouts was negatively correlated to the lesion size. We also examined the hedonic response to palatable food by measuring the number of tongue protrusions in response to presentation of honey, which was also found to be negatively correlated to the lesion size. Next, we used clozapine N-oxide to either activate or inactivate the NAc through viral DREADD expression. As expected, the number of cataplexy bouts increased with activation and decreased with inactivation, and saline control injections showed no changes. Hedonic response in the DREADD experiment varied and showed both increases and decreases across mice. These results demonstrated that the rostral part of the NAc plays a crucial role in triggering cataplexy and hedonic orofacial movements. Since the NAc is also implicated in motivated behavior, we propose that the NAc is one of the key brain structures involved in happiness and is a driving force for positive emotion-related behaviors.
Collapse
Affiliation(s)
- Jingyang Su
- Department of Physiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Zhi Li
- Department of Physiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Akira Yamashita
- Department of Physiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Ikue Kusumoto-Yoshida
- Department of Physiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Takuto Isomichi
- Department of Physiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Liying Hao
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Tomoyuki Kuwaki
- Department of Physiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan.
| |
Collapse
|
65
|
Nucleus Accumbens Cell Type- and Input-Specific Suppression of Unproductive Reward Seeking. Cell Rep 2020; 30:3729-3742.e3. [DOI: 10.1016/j.celrep.2020.02.095] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 08/11/2019] [Accepted: 02/26/2020] [Indexed: 12/11/2022] Open
|
66
|
Cell-Type- and Endocannabinoid-Specific Synapse Connectivity in the Adult Nucleus Accumbens Core. J Neurosci 2019; 40:1028-1041. [PMID: 31831522 DOI: 10.1523/jneurosci.1100-19.2019] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 11/21/2022] Open
Abstract
The nucleus accumbens (NAc) is a mesocorticolimbic structure that integrates cognitive, emotional and motor functions. Although its role in psychiatric disorders is widely acknowledged, the understanding of its circuitry is not complete. Here, we combined optogenetic and whole-cell recordings to draw a functional portrait of excitatory disambiguated synapses onto D1 and D2 medium spiny neurons (MSNs) in the adult male mouse NAc core. Comparing synaptic properties of ventral hippocampus (vHipp), basolateral amygdala (BLA) and prefrontal cortex (PFC) inputs revealed a hierarchy of synaptic inputs that depends on the identity of the postsynaptic target MSN. Thus, the BLA is the dominant excitatory pathway onto D1 MSNs (BLA > PFC = vHipp) while PFC inputs dominate D2 MSNs (PFC > vHipp > BLA). We also tested the hypothesis that endocannabinoids endow excitatory circuits with pathway- and cell-specific plasticity. Thus, whereas CB1 receptors (CB1R) uniformly depress excitatory pathways regardless of MSNs identity, TRPV1 receptors (TRPV1R) bidirectionally control inputs onto the NAc core in a pathway-specific manner. Finally, we show that the interplay of TRPV1R/CB1R shapes plasticity at BLA-NAc synapses. Together these data shed new light on synapse and circuit specificity in the adult NAc core and illustrate how endocannabinoids contribute to pathway-specific synaptic plasticity.SIGNIFICANCE STATEMENT We examined the impact of connections from the ventral hippocampus (vHipp,) basolateral amygdala (BLA) and prefrontal cortex (PFC) onto identified medium spiny neurons (MSNs) in the adult accumbens core. We found BLA inputs were strongest at D1 MSNs while PFC inputs dominate D2 MSNs. Pathway- and cell-specific circuit control was also facilitated by endocannabinoids that endow bidirectional synaptic plasticity at identified BLA-NAc synapses. These data provide mechanistic insights on synapse and circuit specificity in the adult NAc core.
Collapse
|
67
|
Sex and Individual Differences in Alcohol Intake Are Associated with Differences in Ketamine Self-Administration Behaviors and Nucleus Accumbens Dendritic Spine Density. eNeuro 2019; 6:ENEURO.0221-19.2019. [PMID: 31740575 PMCID: PMC6893233 DOI: 10.1523/eneuro.0221-19.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 10/14/2019] [Accepted: 10/22/2019] [Indexed: 01/03/2023] Open
Abstract
Clinical and preclinical studies have shown that ketamine, an NMDA receptor antagonist, has promising therapeutic value for the treatment of alcohol use disorder (AUD). However, the maintenance of remission will ultimately require repeated infusions of ketamine, which may lead to abuse potential and may hinder its therapeutic benefits. It is therefore crucial to assess the effects of repeated treatments with ketamine on alcohol intake. Accordingly, this study aimed to examine in both sexes how individual differences in alcohol intake alter ketamine self-administration and how ketamine self-administration will alter subsequent alcohol-drinking behaviors. Male and female rats intermittently drank alcohol or water for 10 weeks and were divided into high- or low-alcohol intake groups prior to ketamine self-administration. Rats self-administered ketamine under fixed and progressive ratio schedules of reinforcement from week 4 to 7, and the incubation of ketamine craving was examined from week 8 to 10. To investigate structural plasticity in a brain region involved in reward, nucleus accumbens dendritic spine morphology was examined. Our results show that high alcohol intake in male rats attenuated ketamine self-administration, whereas in female rats high alcohol intake enhanced motivation to self-administer ketamine. Ketamine reduced alcohol intake in high-alcohol male rats but increased it in low-alcohol female rats. Incubation of ketamine craving developed in all groups except low-alcohol females. Three weeks of abstinence from ketamine was associated with increased mushroom spines in all groups except the high-alcohol male group. Overall, these data suggest that ketamine as a treatment for AUD may benefit male subjects, but not female subjects, and warrants further investigation before use as a therapeutic agent.
Collapse
|
68
|
Cofresí RU, Bartholow BD, Piasecki TM. Evidence for incentive salience sensitization as a pathway to alcohol use disorder. Neurosci Biobehav Rev 2019; 107:897-926. [PMID: 31672617 PMCID: PMC6878895 DOI: 10.1016/j.neubiorev.2019.10.009] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 10/14/2019] [Accepted: 10/15/2019] [Indexed: 12/12/2022]
Abstract
The incentive salience sensitization (ISS) theory of addiction holds that addictive behavior stems from the ability of drugs to progressively sensitize the brain circuitry that mediates attribution of incentive salience (IS) to reward-predictive cues and its behavioral manifestations. In this article, we establish the plausibility of ISS as an etiological pathway to alcohol use disorder (AUD). We provide a comprehensive and critical review of evidence for: (1) the ability of alcohol to sensitize the brain circuitry of IS attribution and expression; and (2) attribution of IS to alcohol-predictive cues and its sensitization in humans and non-human animals. We point out gaps in the literature and how these might be addressed. We also highlight how individuals with different alcohol subjective response phenotypes may differ in susceptibility to ISS as a pathway to AUD. Finally, we discuss important implications of this neuropsychological mechanism in AUD for psychological and pharmacological interventions attempting to attenuate alcohol craving and cue reactivity.
Collapse
Affiliation(s)
- Roberto U Cofresí
- University of Missouri, Department of Psychological Sciences, Columbia, MO 65211, United States.
| | - Bruce D Bartholow
- University of Missouri, Department of Psychological Sciences, Columbia, MO 65211, United States
| | - Thomas M Piasecki
- University of Missouri, Department of Psychological Sciences, Columbia, MO 65211, United States
| |
Collapse
|
69
|
Cho S, Hachmann JT, Balzekas I, In MH, Andres-Beck LG, Lee KH, Min HK, Jo HJ. Resting-state functional connectivity modulates the BOLD activation induced by nucleus accumbens stimulation in the swine brain. Brain Behav 2019; 9:e01431. [PMID: 31697455 PMCID: PMC6908867 DOI: 10.1002/brb3.1431] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 08/08/2019] [Accepted: 08/11/2019] [Indexed: 01/18/2023] Open
Abstract
INTRODUCTION While the clinical efficacy of deep brain stimulation (DBS) the treatment of motor-related symptoms is well established, the mechanism of action of the resulting cognitive and behavioral effects has been elusive. METHODS By combining functional magnetic resonance imaging (fMRI) and DBS, we investigated the pattern of blood-oxygenation-level-dependent (BOLD) signal changes induced by stimulating the nucleus accumbens in a large animal model. RESULTS We found that diffused BOLD activation across multiple functional networks, including the prefrontal, limbic, and thalamic regions during the stimulation, resulted in a significant change in inter-regional functional connectivity. More importantly, the magnitude of the modulation was closely related to the strength of the inter-regional resting-state functional connectivity. CONCLUSIONS Nucleus accumbens stimulation affects the functional activity in networks that underlie cognition and behavior. Our study provides an insight into the nature of the functional connectivity, which mediates activation effect via brain networks.
Collapse
Affiliation(s)
- Shinho Cho
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA.,Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota at Twin Cities, Minneapolis, MN, USA
| | - Jan T Hachmann
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA.,Department of Neurologic Surgery, Virginia Commonwealth University Health System, Richmond, VA, USA
| | - Irena Balzekas
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Myung-Ho In
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA.,Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Lindsey G Andres-Beck
- Department of Biomedical Engineering and Physiology, Mayo Clinic, Rochester, MN, USA
| | - Kendall H Lee
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA.,Department of Biomedical Engineering and Physiology, Mayo Clinic, Rochester, MN, USA
| | - Hoon-Ki Min
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA.,Department of Radiology, Mayo Clinic, Rochester, MN, USA.,Department of Biomedical Engineering and Physiology, Mayo Clinic, Rochester, MN, USA
| | - Hang Joon Jo
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA.,Department of Neurology, Mayo Clinic, Rochester, MN, USA.,Department of Radiology, Mayo Clinic, Rochester, MN, USA.,Department of Physiology, College of Medicine, Hanyang University, Seoul, South Korea
| |
Collapse
|
70
|
Kardos J, Dobolyi Á, Szabó Z, Simon Á, Lourmet G, Palkovits M, Héja L. Molecular Plasticity of the Nucleus Accumbens Revisited-Astrocytic Waves Shall Rise. Mol Neurobiol 2019; 56:7950-7965. [PMID: 31134458 PMCID: PMC6834761 DOI: 10.1007/s12035-019-1641-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 05/06/2019] [Indexed: 12/11/2022]
Abstract
Part of the ventral striatal division, the nucleus accumbens (NAc) drives the circuit activity of an entire macrosystem about reward like a "flagship," signaling and leading diverse conducts. Accordingly, NAc neurons feature complex inhibitory phenotypes that assemble to process circuit inputs and generate outputs by exploiting specific arrays of opposite and/or parallel neurotransmitters, neuromodulatory peptides. The resulting complex combinations enable versatile yet specific forms of accumbal circuit plasticity, including maladaptive behaviors. Although reward signaling and behavior are elaborately linked to neuronal circuit activities, it is plausible to propose whether these neuronal ensembles and synaptic islands can be directly controlled by astrocytes, a powerful modulator of neuronal activity. Pioneering studies showed that astrocytes in the NAc sense citrate cycle metabolites and/or ATP and may induce recurrent activation. We argue that the astrocytic calcium, GABA, and Glu signaling and altered sodium and chloride dynamics fundamentally shape metaplasticity by providing active regulatory roles in the synapse- and network-level flexibility of the NAc.
Collapse
Affiliation(s)
- Julianna Kardos
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok körútja 2, Budapest, 1117, Hungary.
| | - Árpád Dobolyi
- Laboratory of Neuromorphology, Department of Anatomy, Histology and Embryology, Semmelweis University, Üllői út 26, Budapest, 1086, Hungary
- MTA-ELTE Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Eötvös Loránd University and the Hungarian Academy of Sciences, Pázmány Péter sétány 1C, Budapest, 1117, Hungary
| | - Zsolt Szabó
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok körútja 2, Budapest, 1117, Hungary
| | - Ágnes Simon
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok körútja 2, Budapest, 1117, Hungary
| | - Guillaume Lourmet
- Laboratory of Neuromorphology, Department of Anatomy, Histology and Embryology, Semmelweis University, Üllői út 26, Budapest, 1086, Hungary
| | - Miklós Palkovits
- Human Brain Tissue Bank, Semmelweis University, Tűzoltó utca 58, Budapest, H-1094, Hungary
| | - László Héja
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok körútja 2, Budapest, 1117, Hungary
| |
Collapse
|
71
|
Cartmell SC, Tian Q, Thio BJ, Leuze C, Ye L, Williams NR, Yang G, Ben-Dor G, Deisseroth K, Grill WM, McNab JA, Halpern CH. Multimodal characterization of the human nucleus accumbens. Neuroimage 2019; 198:137-149. [PMID: 31077843 PMCID: PMC7341972 DOI: 10.1016/j.neuroimage.2019.05.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 04/27/2019] [Accepted: 05/07/2019] [Indexed: 01/03/2023] Open
Abstract
Dysregulation of the nucleus accumbens (NAc) is implicated in numerous neuropsychiatric disorders. Treatments targeting this area directly (e.g. deep brain stimulation) demonstrate variable efficacy, perhaps owing to non-specific targeting of a functionally heterogeneous nucleus. Here we provide support for this notion, first observing disparate behavioral effects in response to direct simulation of different locations within the NAc in a human patient. These observations motivate a segmentation of the NAc into subregions, which we produce from a diffusion-tractography based analysis of 245 young, unrelated healthy subjects. We further explore the mechanism of these stimulation-induced behavioral responses by identifying the most probable subset of axons activated using a patient-specific computational model. We validate our diffusion-based segmentation using evidence from several modalities, including MRI-based measures of function and microstructure, human post-mortem immunohistochemical staining, and cross-species comparison of cortical-NAc projections that are known to be conserved. Finally, we visualize the passage of individual axon bundles through one NAc subregion in a post-mortem human sample using CLARITY 3D histology corroborated by 7T tractography. Collectively, these findings extensively characterize human NAc subregions and provide insight into their structural and functional distinctions with implications for stereotactic treatments targeting this region.
Collapse
Affiliation(s)
- Samuel Cd Cartmell
- Department of Neurosurgery, Stanford University, Stanford, CA, 94305, USA
| | - Qiyuan Tian
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA; Department of Electrical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Brandon J Thio
- Department of Biomedical Engineering, Duke University, Stanford University, Stanford, CA, 94305, USA
| | - Christoph Leuze
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA
| | - Li Ye
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Nolan R Williams
- Department of Psychiatry, Stanford University, Stanford, CA, 94305, USA
| | - Grant Yang
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA; Department of Electrical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Gabriel Ben-Dor
- Department of Psychiatry, Stanford University, Stanford, CA, 94305, USA
| | - Karl Deisseroth
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA; Department of Psychiatry, Stanford University, Stanford, CA, 94305, USA
| | - Warren M Grill
- Department of Biomedical Engineering, Duke University, Stanford University, Stanford, CA, 94305, USA
| | - Jennifer A McNab
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA
| | - Casey H Halpern
- Department of Neurosurgery, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
72
|
Shi X, Barr JL, von Weltin E, Wolsh C, Unterwald EM. Differential Roles of Accumbal GSK3 β in Cocaine versus Morphine-Induced Place Preference, U50,488H-Induced Place Aversion, and Object Memory. J Pharmacol Exp Ther 2019; 371:339-347. [PMID: 31420527 DOI: 10.1124/jpet.119.259283] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 07/31/2019] [Indexed: 11/22/2022] Open
Abstract
Previous research has demonstrated that activity of glycogen synthase kinase-3 (GSK3) is necessary for the rewarding effects of cocaine. In the present study, a conditional GSK3β gene knockdown model was used to determine if GSK3β activity specifically in the nucleus accumbens is important for cocaine conditioned reward. The roles of accumbal GSK3β in morphine conditioned reward, trans-(±)-3,4-dichloro-N-methyl-N-[2-(1-pyrrolidinyl)cyclohexyl]benzeneacetamide methanesulfonate salt (U50,488H)-induced conditioned place aversion, and cognitive function were also studied. Adult male and female GSK3β-floxed or wild-type mice were injected with adeno-associated virus/Cre into the nucleus accumbens to reduce expression of GSK3β and underwent behavioral testing 4 weeks later. The development of cocaine-induced conditioned place preference was significantly attenuated in mice with reduced levels of GSK3β in the nucleus accumbens, whereas the development of morphine-induced place preference remained intact. Conditional knockdown of GSK3β in the accumbens prevented the development of conditioned aversion produced by U50,488H, a κ-opioid receptor agonist. Cognitive memory tests revealed deficits in object location memory, but not novel object recognition in mice with accumbal GSK3β knockdown. These data demonstrate that GSK3β in the nucleus accumbens is required for cocaine conditioned place preference and U50,488H conditioned place aversion, as well as spatial memory in object location task, indicating differential roles of GSK3β in the psychostimulant and opiate reward process, as well as in memory for spatial locations and object identity. SIGNIFICANCE STATEMENT: Knockdown of GSK3β in the nucleus accumbens attenuated the development of cocaine-induced place preference, as well as conditioned place aversion to U50,488H, a κ-opioid receptor agonist. In contrast, the development of morphine place preference was not altered by GSK3β knockdown. GSK3β knockdown in nucleus accumbens impaired performance in the object location task, but not the novel object recognition task. These results elucidate different physiological roles of accumbal GSKβ in conditioned reward, aversion, and memory.
Collapse
Affiliation(s)
- Xiangdang Shi
- Center for Substance Abuse Research and Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Jeffrey L Barr
- Center for Substance Abuse Research and Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Eva von Weltin
- Center for Substance Abuse Research and Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Cassandra Wolsh
- Center for Substance Abuse Research and Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Ellen M Unterwald
- Center for Substance Abuse Research and Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
73
|
Abstract
The striatum is essential for learning which actions lead to reward and for implementing those actions. Decades of experimental and theoretical work have led to several influential theories and hypotheses about how the striatal circuit mediates these functions. However, owing to technical limitations, testing these hypotheses rigorously has been difficult. In this Review, we briefly describe some of the classic ideas of striatal function. We then review recent studies in rodents that take advantage of optical and genetic methods to test these classic ideas by recording and manipulating identified cell types within the circuit. This new body of work has provided experimental support of some longstanding ideas about the striatal circuit and has uncovered critical aspects of the classic view that are incorrect or incomplete.
Collapse
Affiliation(s)
- Julia Cox
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Ilana B Witten
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA.
- Department of Psychology, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
74
|
Top-down control of the medial orbitofrontal cortex to nucleus accumbens core pathway in decisional impulsivity. Brain Struct Funct 2019; 224:2437-2452. [DOI: 10.1007/s00429-019-01913-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 06/14/2019] [Indexed: 02/03/2023]
|
75
|
Parekh PK, Logan RW, Ketchesin KD, Becker-Krail D, Shelton MA, Hildebrand MA, Barko K, Huang YH, McClung CA. Cell-Type-Specific Regulation of Nucleus Accumbens Synaptic Plasticity and Cocaine Reward Sensitivity by the Circadian Protein, NPAS2. J Neurosci 2019; 39:4657-4667. [PMID: 30962277 PMCID: PMC6561687 DOI: 10.1523/jneurosci.2233-18.2019] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 03/16/2019] [Accepted: 04/03/2019] [Indexed: 12/23/2022] Open
Abstract
The circadian transcription factor neuronal PAS domain 2 (NPAS2) is linked to psychiatric disorders associated with altered reward sensitivity. The expression of Npas2 is preferentially enriched in the mammalian forebrain, including the nucleus accumbens (NAc), a major neural substrate of motivated and reward behavior. Previously, we demonstrated that downregulation of NPAS2 in the NAc reduces the conditioned behavioral response to cocaine in mice. We also showed that Npas2 is preferentially enriched in dopamine receptor 1 containing medium spiny neurons (D1R-MSNs) of the striatum. To extend these studies, we investigated the impact of NPAS2 disruption on accumbal excitatory synaptic transmission and strength, along with the behavioral sensitivity to cocaine reward in a cell-type-specific manner. Viral-mediated knockdown of Npas2 in the NAc of male and female C57BL/6J mice increased the excitatory drive onto MSNs. Using Drd1a-tdTomato mice in combination with viral knockdown, we determined these synaptic adaptations were specific to D1R-MSNs relative to non-D1R-MSNs. Interestingly, NAc-specific knockdown of Npas2 blocked cocaine-induced enhancement of synaptic strength and glutamatergic transmission specifically onto D1R-MSNs. Last, we designed, validated, and used a novel Cre-inducible short-hairpin RNA virus for MSN-subtype-specific knockdown of Npas2 Cell-type-specific Npas2 knockdown in D1R-MSNs, but not D2R-MSNs, in the NAc reduced cocaine conditioned place preference. Together, our results demonstrate that NPAS2 regulates excitatory synapses of D1R-MSNs in the NAc and cocaine reward-related behavior.SIGNIFICANCE STATEMENT Drug addiction is a widespread public health concern often comorbid with other psychiatric disorders. Disruptions of the circadian clock can predispose or exacerbate substance abuse in vulnerable individuals. We demonstrate a role for the core circadian protein, NPAS2, in mediating glutamatergic neurotransmission at medium spiny neurons (MSNs) in the nucleus accumbens (NAc), a region critical for reward processing. We find that NPAS2 negatively regulates functional excitatory synaptic plasticity in the NAc and is necessary for cocaine-induced plastic changes in MSNs expressing the dopamine 1 receptor (D1R). We further demonstrate disruption of NPAS2 in D1R-MSNs produces augmented cocaine preference. These findings highlight the significance of cell-type-specificity in mechanisms underlying reward regulation by NPAS2 and extend our knowledge of its function.
Collapse
Affiliation(s)
- Puja K Parekh
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15219, and
| | - Ryan W Logan
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15219, and
- Center for Systems Genetics of Addiction, The Jackson Laboratory, Bar Harbor, Maine 04609
| | - Kyle D Ketchesin
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15219, and
| | - Darius Becker-Krail
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15219, and
| | - Micah A Shelton
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15219, and
| | - Mariah A Hildebrand
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15219, and
| | - Kelly Barko
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15219, and
| | - Yanhua H Huang
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15219, and
| | - Colleen A McClung
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15219, and
- Center for Systems Genetics of Addiction, The Jackson Laboratory, Bar Harbor, Maine 04609
| |
Collapse
|
76
|
Dopamine-glutamate neuron projections to the nucleus accumbens medial shell and behavioral switching. Neurochem Int 2019; 129:104482. [PMID: 31170424 DOI: 10.1016/j.neuint.2019.104482] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 05/14/2019] [Accepted: 05/27/2019] [Indexed: 12/29/2022]
Abstract
Dopamine (DA) neuron projections to the striatum are functionally heterogeneous with diverse behavioral roles. We focus here on DA neuron projections to the nucleus accumbens (NAc) medial Shell, their distinct anatomical and functional connections, and discuss their role in motivated behavior. We first review rodent studies showing that a subpopulation of DA neurons in the medial ventral tegmental area (VTA) project to the NAc medial Shell. Using a combinatorial strategy, we show that the majority of DA neurons projecting to the NAc Shell express vesicular glutamate transporter 2 (VGLUT2) making them capable of glutamate co-transmission (DA-GLU neurons). In the NAc dorsal medial Shell, all of the DA neuron terminals arise from DA-GLU neurons, while in the lateral NAc Shell, DA neuron terminals arise from both DA-GLU neurons and DA-only neurons, without VGLUT2. DA-GLU neurons make excitatory connections to the three major cells types, spiny projection neurons, fast-spiking interneuron and cholinergic interneurons (ChIs). The strongest DA-GLU neuron excitatory connections are to ChIs. Photostimulation of DA-GLU neuron terminals in the slice drives ChIs to burst fire. Finally, we review studies that address specially the behavioral function of this subpopulation of DA neurons in extinction learning and latent inhibition. Taking into account findings from anatomical and functional connectome studies, we propose that DA-GLU neuron connections to ChIs in the medial Shell play a crucial role in switching behavioral responses under circumstances of altered cue-reinforcer contingencies.
Collapse
|
77
|
Orsini CA, Hernandez CM, Bizon JL, Setlow B. Deconstructing value-based decision making via temporally selective manipulation of neural activity: Insights from rodent models. COGNITIVE, AFFECTIVE & BEHAVIORAL NEUROSCIENCE 2019; 19:459-476. [PMID: 30341621 PMCID: PMC6472996 DOI: 10.3758/s13415-018-00649-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The ability to choose among options that differ in their rewards and costs (value-based decision making) has long been a topic of interest for neuroscientists, psychologists, and economists alike. This is likely because this is a cognitive process in which all animals (including humans) engage on a daily basis, be it routine (which road to take to work) or consequential (which graduate school to attend). Studies of value-based decision making (particularly at the preclinical level) often treat it as a uniform process. The results of such studies have been invaluable for our understanding of the brain substrates and neurochemical systems that contribute to decision making involving a range of different rewards and costs. Value-based decision making is not a unitary process, however, but is instead composed of distinct cognitive operations that function in concert to guide choice behavior. Within this conceptual framework, it is therefore important to consider that the known neural substrates supporting decision making may contribute to temporally distinct and dissociable components of the decision process. This review will describe this approach for investigating decision making, drawing from published studies that have used techniques that allow temporal dissection of the decision process, with an emphasis on the literature in animal models. The review will conclude with a discussion of the implications of this work for understanding pathological conditions that are characterized by impaired decision making.
Collapse
Affiliation(s)
- Caitlin A Orsini
- Department of Psychiatry, University of Florida College of Medicine, P.O. Box 100256, Gainesville, FL, 32610-0256, USA.
- Center for Addiction Research and Education, University of Florida, Gainesville, FL, 32610, USA.
| | - Caesar M Hernandez
- Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA
| | - Jennifer L Bizon
- Department of Psychiatry, University of Florida College of Medicine, P.O. Box 100256, Gainesville, FL, 32610-0256, USA
- Center for Addiction Research and Education, University of Florida, Gainesville, FL, 32610, USA
- Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA
| | - Barry Setlow
- Department of Psychiatry, University of Florida College of Medicine, P.O. Box 100256, Gainesville, FL, 32610-0256, USA
- Center for Addiction Research and Education, University of Florida, Gainesville, FL, 32610, USA
- Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA
- Department of Psychology, University of Florida, Gainesville, FL, 32610, USA
| |
Collapse
|
78
|
Hernandez NS, Schmidt HD. Central GLP-1 receptors: Novel molecular targets for cocaine use disorder. Physiol Behav 2019; 206:93-105. [PMID: 30930091 DOI: 10.1016/j.physbeh.2019.03.026] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 03/20/2019] [Accepted: 03/26/2019] [Indexed: 12/25/2022]
Abstract
Given that the search for effective pharmacotherapies for cocaine use disorder has, thus far, been fruitless, there remains a critical need for conceptually innovative approaches toward identifying new medications to treat this disease. A better understanding of the neurocircuits and neurobiological mechanisms underlying cocaine taking and seeking may identify molecular substrates that could serve as targets for novel pharmacotherapies to treat cocaine use disorder. Recent preclinical evidence suggests that glucagon-like peptide-1 (GLP-1) receptor agonists could be re-purposed to treat cocaine craving-induced relapse. This review endeavors to comprehensively summarize the current literature investigating the efficacy of GLP-1 receptor agonists in reducing the rewarding and reinforcing effects of cocaine in animal models of cocaine use disorder. The role of central endogenous GLP-1 circuits in voluntary cocaine taking and seeking is also discussed. Behavioral, neurochemical, electrophysiological and molecular biology studies indicate that central GLP-1 receptor activation functionally modulates the mesolimbic reward system and decreases addiction-like phenotypes in rodents. Overall, an emerging preclinical literature provides compelling evidence to advance GLP-1 receptor agonists into clinical trials testing the efficacy of these medications in preventing cocaine craving-induced relapse.
Collapse
Affiliation(s)
- N S Hernandez
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States of America; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States of America
| | - H D Schmidt
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States of America; Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA 19104, United States of America.
| |
Collapse
|
79
|
Prolonged ad libitum access to low-concentration sucrose changes the neurochemistry of the nucleus accumbens in male Sprague-Dawley rats. Physiol Behav 2019; 201:95-103. [DOI: 10.1016/j.physbeh.2018.12.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 12/12/2018] [Accepted: 12/12/2018] [Indexed: 01/12/2023]
|
80
|
Park YS, Sammartino F, Young NA, Corrigan J, Krishna V, Rezai AR. Anatomic Review of the Ventral Capsule/Ventral Striatum and the Nucleus Accumbens to Guide Target Selection for Deep Brain Stimulation for Obsessive-Compulsive Disorder. World Neurosurg 2019; 126:1-10. [PMID: 30790738 DOI: 10.1016/j.wneu.2019.01.254] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 01/23/2019] [Accepted: 01/24/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Disturbances in the reward network of the brain underlie addiction, depression, and obsessive-compulsive disorder. The ventral capsule/ventral striatum and nucleus accumbens (NAc) region is a clinically approved target for deep brain stimulation for obsessive-compulsive disorder. METHODS We performed a comprehensive literature review to define clinically relevant anatomy and connectivity of the ventral capsule/ventral striatum and NAc region to guide target selection for deep brain stimulation. RESULTS Architecturally and functionally, the NAc is divided into the core and the shell, with each area having different connections. The shell primarily receives limbic information, and the core typically receives information from the motor system. In general, afferents from the prefrontal cortex, hippocampus, and amygdala are excitatory. The dopaminergic projections to the NAc from the ventral tegmental area modulate the balance of these excitatory inputs. Several important inputs to the NAc converge at the junction of the internal capsule (IC) and the anterior commissure (AC): the ventral amygdalofugal pathways that run parallel to and underneath the AC, the precommissural fornical fibers that run anterior to the AC, axons from the ventral prefrontal cortex and medial orbitofrontal cortex that occupy the most ventral part of the IC and embedding within the NAc and AC, and the superolateral branch of the medial forebrain bundle located parallel to the anterior thalamic radiation in the IC. CONCLUSIONS The caudal part of the NAc passing through the IC-AC junction may be an effective target for deep brain stimulation to improve behavioral symptoms associated with obsessive-compulsive disorder.
Collapse
Affiliation(s)
- Yong-Sook Park
- Department of Neurosurgery, Chung-Ang University Hospital, Seoul, Korea
| | | | - Nicole A Young
- Department of Neurosurgery, The Ohio State University, Columbus, Ohio, USA
| | - John Corrigan
- Department of Neurosurgery, The Ohio State University, Columbus, Ohio, USA
| | - Vibhor Krishna
- Department of Neurosurgery, The Ohio State University, Columbus, Ohio, USA.
| | - Ali R Rezai
- Department of Neurosurgery, West Virginia University Hospital, Morgantown, West Virginia, USA
| |
Collapse
|
81
|
Morris LS, Kundu P, Costi S, Collins A, Schneider M, Verma G, Balchandani P, Murrough JW. Ultra-high field MRI reveals mood-related circuit disturbances in depression: a comparison between 3-Tesla and 7-Tesla. Transl Psychiatry 2019; 9:94. [PMID: 30770788 PMCID: PMC6377652 DOI: 10.1038/s41398-019-0425-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 01/10/2019] [Indexed: 12/30/2022] Open
Abstract
Ultra-high field 7-Tesla (7 T) MRI has the potential to advance our understanding of neuropsychiatric disorders, including major depressive disorder (MDD). To date, few studies have quantified the advantage of resting state functional MRI (fMRI) at 7 T compared to 3-Tesla (3 T). We conducted a series of experiments that demonstrate the improvement in temporal signal-to-noise ratio (TSNR) of a multi-echo multi-band fMRI protocol with ultra-high field 7 T MRI, compared to a similar protocol using 3 T MRI in healthy controls (HC). We also directly tested the enhancement in ultra-high field 7 T fMRI signal power by examining the ventral tegmental area (VTA), a small midbrain structure that is critical to the expected neuropathology of MDD but difficult to discern with standard 3 T MRI. We demonstrate up to 300% improvement in TSNR and resting state functional connectivity coefficients provided by ultra-high field 7 T fMRI compared to 3 T, indicating enhanced power for detection of functional neural architecture. A multi-echo based acquisition protocol and signal denoising pipeline afforded greater gain in signal power compared to classic acquisition and denoising pipelines. Furthermore, ultra-high field fMRI revealed mood-related neurocircuit disturbances in patients with MDD compared to HC, which were not detectable with 3 T fMRI. Ultra-high field 7 T fMRI may provide an effective tool for studying functional neural architecture relevant to MDD and other neuropsychiatric disorders.
Collapse
Affiliation(s)
- Laurel S. Morris
- 0000 0001 0670 2351grid.59734.3cThe Mood and Anxiety Disorders Program, Department of Psychiatry, The Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA ,0000 0001 0670 2351grid.59734.3cThe Translational and Molecular Imaging Institute, Department of Radiology, The Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Prantik Kundu
- 0000 0001 0670 2351grid.59734.3cThe Mood and Anxiety Disorders Program, Department of Psychiatry, The Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA ,0000 0001 0670 2351grid.59734.3cThe Translational and Molecular Imaging Institute, Department of Radiology, The Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Sara Costi
- 0000 0001 0670 2351grid.59734.3cThe Mood and Anxiety Disorders Program, Department of Psychiatry, The Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA ,0000 0001 0670 2351grid.59734.3cThe Translational and Molecular Imaging Institute, Department of Radiology, The Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Abigail Collins
- 0000 0001 0670 2351grid.59734.3cThe Mood and Anxiety Disorders Program, Department of Psychiatry, The Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA ,0000 0001 0670 2351grid.59734.3cThe Translational and Molecular Imaging Institute, Department of Radiology, The Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Molly Schneider
- 0000 0001 0670 2351grid.59734.3cThe Mood and Anxiety Disorders Program, Department of Psychiatry, The Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA ,0000 0001 0670 2351grid.59734.3cThe Translational and Molecular Imaging Institute, Department of Radiology, The Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Gaurav Verma
- 0000 0001 0670 2351grid.59734.3cThe Mood and Anxiety Disorders Program, Department of Psychiatry, The Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA ,0000 0001 0670 2351grid.59734.3cThe Translational and Molecular Imaging Institute, Department of Radiology, The Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Priti Balchandani
- 0000 0001 0670 2351grid.59734.3cThe Mood and Anxiety Disorders Program, Department of Psychiatry, The Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA ,0000 0001 0670 2351grid.59734.3cThe Translational and Molecular Imaging Institute, Department of Radiology, The Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - James W. Murrough
- 0000 0001 0670 2351grid.59734.3cThe Mood and Anxiety Disorders Program, Department of Psychiatry, The Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA ,0000 0001 0670 2351grid.59734.3cThe Translational and Molecular Imaging Institute, Department of Radiology, The Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| |
Collapse
|
82
|
Reiner DJ, Fredriksson I, Lofaro OM, Bossert JM, Shaham Y. Relapse to opioid seeking in rat models: behavior, pharmacology and circuits. Neuropsychopharmacology 2019; 44:465-477. [PMID: 30293087 PMCID: PMC6333846 DOI: 10.1038/s41386-018-0234-2] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 09/17/2018] [Accepted: 09/27/2018] [Indexed: 12/13/2022]
Abstract
Lifetime relapse rates remain a major obstacle in addressing the current opioid crisis. Relapse to opioid use can be modeled in rodent studies where drug self-administration is followed by a period of abstinence and a subsequent test for drug seeking. Abstinence can be achieved through extinction training, forced abstinence, or voluntary abstinence. Voluntary abstinence can be accomplished by introducing adverse consequences of continued drug self-administration (e.g., punishment or electric barrier) or by introducing an alternative nondrug reward in a discrete choice procedure (drug versus palatable food or social interaction). In this review, we first discuss pharmacological and circuit mechanisms of opioid seeking, as assessed in the classical extinction-reinstatement model, where reinstatement is induced by reexposure to the self-administered drug (drug priming), discrete cues, discriminative cues, drug-associated contexts, different forms of stress, or withdrawal states. Next, we discuss pharmacological and circuit mechanisms of relapse after forced or voluntary abstinence, including the phenomenon of "incubation of heroin craving" (the time-dependent increases in heroin seeking during abstinence). We conclude by discussing future directions of preclinical relapse-related studies using opioid drugs.
Collapse
Affiliation(s)
- David J. Reiner
- Behavioral Neuroscience Research Branch, IRP-NIDA-NIH, Baltimore, MD USA
| | - Ida Fredriksson
- Behavioral Neuroscience Research Branch, IRP-NIDA-NIH, Baltimore, MD USA
| | - Olivia M. Lofaro
- Behavioral Neuroscience Research Branch, IRP-NIDA-NIH, Baltimore, MD USA
| | | | - Yavin Shaham
- Behavioral Neuroscience Research Branch, IRP-NIDA-NIH, Baltimore, MD, USA.
| |
Collapse
|
83
|
Ventral Pallidum Is the Primary Target for Accumbens D1 Projections Driving Cocaine Seeking. J Neurosci 2019; 39:2041-2051. [PMID: 30622165 PMCID: PMC6507080 DOI: 10.1523/jneurosci.2822-18.2018] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 12/11/2018] [Accepted: 12/21/2018] [Indexed: 01/18/2023] Open
Abstract
Outputs from the nucleus accumbens (NAc) include projections to the ventral pallidum and the ventral tegmental area and subtantia nigra in the ventral mesencephalon. The medium spiny neurons (MSN) that give rise to these pathways are GABAergic and consist of two populations of equal number that are segregated by differentially expressed proteins, including D1- and D2-dopamine receptors. Afferents to the ventral pallidum arise from both D1- and D2-MSNs, whereas the ventral mesencephalon is selectively innervated by D1-MSN. To determine the extent of collateralization of D1-MSN to these axon terminal fields we used retrograde labeling in transgenic mice expressing tdTomato selectively in D1-MSN, and found that a large majority of D1-MSN in either the shell or core subcompartments of the accumbens collateralized to both output structures. Approximately 70% of D1-MSNs projecting to the ventral pallidum collateralized to the ventral mesencephalon, whereas >90% of mesencephalic D1-MSN afferents collateralized to the ventral pallidum. In contrast, <10% of dorsal striatal D1-MSNs collateralized to both the globus pallidus and ventral mesencephalon. D1-MSN activation is required for conditioned cues to induce cocaine seeking. To determine which D1-MSN projection mediates cued cocaine seeking, we selectively transfected D1-MSNs in transgenic rats with an inhibitory Gi-coupled DREADD. Activation of the transfected Gi-DREADD with clozapine-N-oxide administered into the ventral pallidum, but not into the ventral mesencephalon, blocked cue-induced cocaine seeking. These data show that, although accumbens D1-MSNs largely collateralize to both the ventral pallidum and ventral mesencephalon, only D1-MSN innervation of the ventral pallidum is necessary for cue-induced cocaine seeking.SIGNIFICANCE STATEMENT Activity in D1 dopamine receptor-expressing neurons in the NAc is required for rodents to respond to cocaine-conditioned cues and relapse to drug seeking behaviors. The D1-expressing neurons project to both the ventral pallidum and ventral mesencephalon, and we found that a majority of the neurons that innervate the ventral pallidum also collateralize to the ventral mesencephalon. However, despite innervating both structures, only D1 innervation of the ventral pallidum mediates cue-induced cocaine seeking.
Collapse
|
84
|
Noorbakhshnia M, Zarrinimehr N. The role of nucleus accumbens shell on acquisition and retrieval stages of morphine state dependent learning. Asian J Psychiatr 2019; 39:150-156. [PMID: 30639974 DOI: 10.1016/j.ajp.2018.12.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 12/15/2018] [Accepted: 12/20/2018] [Indexed: 01/28/2023]
Abstract
AIM In the present study, the effect of transient inactivation of the shell subregion of the nucleus accumbens (NAC shell) by lidocaine on the acquisition and retrieval stages of passive avoidance learning (PAL) and memory and morphine state-dependent learning (SDL) in male wistar rats was investigated. METHODOLOGY Adult male wistar rats weighing (220-250 g) were used. Lidocaine hydrochloride was bilaterally injected into the shell area of the nucleus accumbens 5 min before of subcutaneous morphine administration. RESULTS pre-training and pre-test infusion of lidocaine into the NAC shell significantly impaired PAL and memory. Furthermore, Pre-training administration of morphine (5 mg/kg, s.c.) in a step-through passive avoidance task induced state-dependent learning with impaired memory retrieval on the test day. The impairment of memory was restored after pre-test administration of the same dose of morphine. This phenomenon has been named as morphine state dependent learning (SDL). Moreover, Pre-training and pre-test inactivation of the NAC shell impaired morphine SDL. CONCLUSIONS The results suggest the role of NAC shell as a common structure in the PAL and morphine SDL. It is suggested that NAC shell as a common area plays a critical role in the acquisition and retrieval stages of PAL and also morphine SDL.
Collapse
Affiliation(s)
- Maryam Noorbakhshnia
- Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran.
| | - Nahid Zarrinimehr
- Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran
| |
Collapse
|
85
|
Huang Y, Yu R. Common and distinct neural substrates of the money illusion in win and loss domains. Neuroimage 2019; 184:109-118. [PMID: 30219291 DOI: 10.1016/j.neuroimage.2018.09.032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Revised: 09/03/2018] [Accepted: 09/12/2018] [Indexed: 11/30/2022] Open
Abstract
People often evaluate money based on its face value and overlook its real purchasing power, a phenomenon known as the money illusion. In the present study, using functional magnetic resonance imaging (fMRI) combined with a gambling task, we examined the neural signatures of the money illusion in both win and loss domains. Behavioral results showed that self-reported satisfaction with outcomes was modulated by the face value but not the true value of money in both win and loss domains. At the neural level, activity in the posterior insula was associated with the true value of money in the win domain, but not in the loss domain. Importantly, we found that the ventral striatum, ventromedial prefrontal cortex (vmPFC) and amygdala encoded the money illusion in both domains, indicating a domain-general rather than domain-specific neural signature. Moreover, participants with a larger degree of money illusion at the behavioral level showed stronger functional connectivity between the ventral striatum and ventral anterior cingulate cortex (vACC) in the win domain, but stronger functional connectivity between the ventral striatum and amygdala in the loss domain. Our findings highlight the overlapping and distinct neural substrates underlying the money illusion in the context of wins and losses.
Collapse
Affiliation(s)
- Yi Huang
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore
| | - Rongjun Yu
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore; Department of Psychology, National University of Singapore, Singapore.
| |
Collapse
|
86
|
Solinas M, Belujon P, Fernagut PO, Jaber M, Thiriet N. Dopamine and addiction: what have we learned from 40 years of research. J Neural Transm (Vienna) 2018; 126:481-516. [PMID: 30569209 DOI: 10.1007/s00702-018-1957-2] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 11/17/2018] [Indexed: 12/22/2022]
Abstract
Among the neurotransmitters involved in addiction, dopamine (DA) is clearly the best known. The critical role of DA in addiction is supported by converging evidence that has been accumulated in the last 40 years. In the present review, first we describe the dopaminergic system in terms of connectivity, functioning and involvement in reward processes. Second, we describe the functional, structural, and molecular changes induced by drugs within the DA system in terms of neuronal activity, synaptic plasticity and transcriptional and molecular adaptations. Third, we describe how genetic mouse models have helped characterizing the role of DA in addiction. Fourth, we describe the involvement of the DA system in the vulnerability to addiction and the interesting case of addiction DA replacement therapy in Parkinson's disease. Finally, we describe how the DA system has been targeted to treat patients suffering from addiction and the result obtained in clinical settings and we discuss how these different lines of evidence have been instrumental in shaping our understanding of the physiopathology of drug addiction.
Collapse
Affiliation(s)
- Marcello Solinas
- Université de Poitiers, INSERM, U-1084, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France.
| | - Pauline Belujon
- Université de Poitiers, INSERM, U-1084, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France
| | - Pierre Olivier Fernagut
- Université de Poitiers, INSERM, U-1084, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France
| | - Mohamed Jaber
- Université de Poitiers, INSERM, U-1084, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France
- CHU de Poitiers, Poitiers, France
| | - Nathalie Thiriet
- Université de Poitiers, INSERM, U-1084, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France
| |
Collapse
|
87
|
Ventral pallidum encodes relative reward value earlier and more robustly than nucleus accumbens. Nat Commun 2018; 9:4350. [PMID: 30341305 PMCID: PMC6195583 DOI: 10.1038/s41467-018-06849-z] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 09/26/2018] [Indexed: 12/21/2022] Open
Abstract
The ventral striatopallidal system, a basal ganglia network thought to convert limbic information into behavioral action, includes the nucleus accumbens (NAc) and the ventral pallidum (VP), typically described as a major output of NAc. Here, to investigate how reward-related information is transformed across this circuit, we measure the activity of neurons in NAc and VP when rats receive two highly palatable but differentially preferred rewards, allowing us to track the reward-specific information contained within the neural activity of each region. In VP, we find a prominent preference-related signal that flexibly reports the relative value of reward outcomes across multiple conditions. This reward-specific firing in VP is present in a greater proportion of the population and arises sooner following reward delivery than in NAc. Our findings establish VP as a preeminent value signaler and challenge the existing model of information flow in the ventral basal ganglia.
Collapse
|
88
|
Li Z, Chen Z, Fan G, Li A, Yuan J, Xu T. Cell-Type-Specific Afferent Innervation of the Nucleus Accumbens Core and Shell. Front Neuroanat 2018; 12:84. [PMID: 30459564 PMCID: PMC6232828 DOI: 10.3389/fnana.2018.00084] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 09/25/2018] [Indexed: 01/21/2023] Open
Abstract
The nucleus accumbens (NAc) is clearly implicated in reward processing and drug addiction, as well as in numerous neurological and psychiatric disorders; nevertheless, the circuit mechanisms underlying the diverse functions of the NAc remain poorly understood. Here, we characterized the whole-brain and monosynaptic inputs to two main projection cell types – D1 dopamine receptor expressing medium spiny neurons (D1R-MSNs) and D2 dopamine receptor expressing medium spiny neurons (D2R-MSNs) – within the NAc core and NAc shell by rabies-mediated trans-synaptic tracing. We discovered that D1R-MSNs and D2R-MSNs in both NAc subregions receive similar inputs from diverse sources. Inputs to the NAc core are broadly scattered, whereas inputs to the NAc shell are relatively concentrated. Furthermore, we identified numerous brain areas providing important contrasting inputs to different NAc subregions. The anterior cortex preferentially innervates the NAc core for both D1R-MSNs and D2R-MSNs, whereas the lateral hypothalamic area (LH) preferentially targets D1R-MSNs in the NAc shell. Characterizing the cell-type-specific connectivity of different NAc subregions lays a foundation for studying how diverse functions of the NAc are mediated by specific pathways.
Collapse
Affiliation(s)
- Zhao Li
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Zhilong Chen
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Guoqing Fan
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Anan Li
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Yuan
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Tonghui Xu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
89
|
Ketamine and its metabolite (2R,6R)-hydroxynorketamine induce lasting alterations in glutamatergic synaptic plasticity in the mesolimbic circuit. Mol Psychiatry 2018; 23:2066-2077. [PMID: 29158578 DOI: 10.1038/mp.2017.239] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 08/24/2017] [Accepted: 09/07/2017] [Indexed: 12/17/2022]
Abstract
Low doses of ketamine trigger rapid and lasting antidepressant effects after one injection in treatment-resistant patients with major depressive disorder. Modulation of AMPA receptors (AMPARs) in the hippocampus and prefrontal cortex is suggested to mediate the antidepressant action of ketamine and of one of its metabolites (2R,6R)-hydroxynorketamine ((2R,6R)-HNK). We have examined whether ketamine and (2R,6R)-HNK affect glutamatergic transmission and plasticity in the mesolimbic system, brain regions known to have key roles in reward-motivated behaviors, mood and hedonic drive. We found that one day after the injection of a low dose of ketamine, long-term potentiation (LTP) in the nucleus accumbens (NAc) was impaired. Loss of LTP was maintained for 7 days and was not associated with an altered basal synaptic transmission mediated by AMPARs and N-methyl-D-aspartate receptors (NMDARs). Inhibition of mammalian target of rapamycin signaling with rapamycin did not prevent the ketamine-induced loss of LTP but inhibited LTP in saline-treated mice. However, ketamine blunted the increase in the phosphorylation of the GluA1 subunit of AMPARs at a calcium/calmodulin-dependent protein kinase II/protein kinase C site induced by an LTP induction protocol. Moreover, ketamine caused a persistent increased phosphorylation of GluA1 at a protein kinase A site. (2R,6R)-HNK also impaired LTP in the NAc. In dopaminergic neurons of the ventral tegmental area from ketamine- or (2R,6R)-HNK-treated mice, AMPAR-mediated responses were depressed, while those mediated by NMDARs were unaltered, which resulted in a reduced AMPA/NMDA ratio, a measure of long-term synaptic depression. These results demonstrate that a single injection of ketamine or (2R,6R)-HNK induces enduring alterations in the function of AMPARs and synaptic plasticity in brain regions involved in reward-related behaviors.
Collapse
|
90
|
Cao J, Dorris DM, Meitzen J. Electrophysiological properties of medium spiny neurons in the nucleus accumbens core of prepubertal male and female Drd1a-tdTomato line 6 BAC transgenic mice. J Neurophysiol 2018; 120:1712-1727. [PMID: 29975170 PMCID: PMC6230806 DOI: 10.1152/jn.00257.2018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 06/27/2018] [Accepted: 06/30/2018] [Indexed: 12/12/2022] Open
Abstract
The nucleus accumbens core (AcbC) is a striatal brain region essential for integrating motivated behavior and reward processing with premotor function. In humans and rodents, research has identified sex differences and sex steroid hormone sensitivity in AcbC-mediated behaviors, in disorders, and in rats in the electrophysiological properties of the AcbC output neuron type, the medium spiny neuron (MSN). It is unknown whether the sex differences detected in MSN electrophysiological properties extend to mice. Furthermore, MSNs come in distinct subtypes with subtle differences in electrophysiological properties, and it is unknown whether MSN subtype-specific electrophysiology varies by sex. To address these questions, we used male and female Drd1a-tdTomato line 6 bacterial artificial chromosome transgenic mice. We made acute brain slices of the AcbC, and performed whole cell patch-clamp recordings across MSN subtypes to comprehensively assess AcbC MSN subtype electrophysiological properties. We found that ( 1 mice MSNs did not exhibit the sex differences detected in rat MSNs, and 2) electrophysiological properties differed between MSN subtypes in both sexes, including rheobase, resting membrane potential, action potential properties, intrinsic excitability, input resistance in both the linear and rectified ranges, and miniature excitatory postsynaptic current properties. These findings significantly extend previous studies of MSN subtypes performed in males or animals of undetermined sex and indicate that the influence of sex upon AcbC MSN properties varies between rodent species. NEW & NOTEWORTHY This research provides the most comprehensive assessment of medium spiny neuron subtype electrophysiological properties to date in a critical brain region, the nucleus accumbens core. It additionally represents the first evaluation of whether mouse medium spiny neuron subtype electrophysiological properties differ by sex.
Collapse
Affiliation(s)
- Jinyan Cao
- Department of Biological Sciences, North Carolina State University , Raleigh, North Carolina
- W. M. Keck Center for Behavioral Biology, North Carolina State University , Raleigh, North Carolina
| | - David M Dorris
- Department of Biological Sciences, North Carolina State University , Raleigh, North Carolina
| | - John Meitzen
- Department of Biological Sciences, North Carolina State University , Raleigh, North Carolina
- W. M. Keck Center for Behavioral Biology, North Carolina State University , Raleigh, North Carolina
- Center for Human Health and the Environment, North Carolina State University , Raleigh, North Carolina
- Comparative Medicine Institute, North Carolina State University , Raleigh, North Carolina
| |
Collapse
|
91
|
Fitzgerald ML, Pickel VM. Adolescent isolation rearing produces a prepulse inhibition deficit correlated with expression of the NMDA GluN1 subunit in the nucleus accumbens. Brain Struct Funct 2018; 223:3169-3181. [PMID: 29779156 PMCID: PMC6626533 DOI: 10.1007/s00429-018-1673-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 04/27/2018] [Indexed: 12/18/2022]
Abstract
Adolescence is a transition period during which social interaction is necessary for normal brain and behavior development. Severely abnormal social interactions during adolescence can increase the incidence of lifelong psychiatric disease. Decreased prepulse inhibition (PPI) is a quantifiable hallmark of some psychiatric illnesses in humans and can be elicited in rodents by isolation rearing throughout the adolescent transition period. PPI is a measure of sensorimotor gating in which the nucleus accumbens (Acb) is crucially involved. The Acb is comprised of core and shell subregions, which receive convergent dopaminergic and glutamatergic inputs. To gain insight into the neurobiological correlates of adolescent adversity, we conducted electron microscopic immunolabeling of dopamine D1 receptors (D1Rs) and the GluN1 subunit of glutamate NMDA receptors in the Acb of isolation-reared (IR) adult male rats. In all animals, GluN1 was primarily located in dendritic profiles, many of which also contained D1Rs. GluN1 was also observed in perisynaptic glia and axon terminals. In IR rats compared with group-reared controls, GluN1 density was selectively decreased in D1R-containing dendrites of the Acb core. Across all animals, dendritic GluN1 density correlated with average percent PPI, implicating endogenous expression of NMDA receptors of the Acb as a possible substrate of the PPI response. These results suggest that adolescent isolation dampens NMDA-mediated excitation in direct (D1R-containing) output neurons of the Acb, and that these changes influence the operational measure of PPI.
Collapse
Affiliation(s)
- Megan L Fitzgerald
- Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY, 10065, USA
- New York State Psychiatric Institute, Columbia University, New York, NY, 10032, USA
| | - Virginia M Pickel
- Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY, 10065, USA.
| |
Collapse
|
92
|
Gibson GD, Millan EZ, McNally GP. The nucleus accumbens shell in reinstatement and extinction of drug seeking. Eur J Neurosci 2018; 50:2014-2022. [PMID: 30044017 DOI: 10.1111/ejn.14084] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 07/05/2018] [Accepted: 07/17/2018] [Indexed: 12/31/2022]
Abstract
The contexts where drugs are self-administered have important control over relapse and extinction of drug-seeking behavior. The nucleus accumbens shell (AcbSh) is essential to this contextual control over drug-seeking behavior. It has been consistently implicated in both the expression of context-induced reinstatement and the expression of extinction, across a variety of drug classes and other rewards. Here, we review the evidence linking AcbSh to the extinction and reinstatement of drug seeking. We consider whether this dual role can be linked to known heterogeneities in AcbSh cell types, their major afferents, and their major efferents. We show that although these heterogeneities are each important and can determine extinction vs. reinstatement, they do not seem adequate to explain the body of findings from the behavioral literature. Rather, we suggest that this functional specialization of AcbSh may be more profitably viewed in terms of the segregation and compartmentalization of AcbSh channels.
Collapse
Affiliation(s)
| | - E Zayra Millan
- School of Psychology, UNSW Sydney, Sydney, 2052, NSW, Australia
| | - Gavan P McNally
- School of Psychology, UNSW Sydney, Sydney, 2052, NSW, Australia
| |
Collapse
|
93
|
Microinjection of the mGluR2/3 agonist, LY379268, into the nucleus accumbens attenuates extinction latencies and the reinstatement of morphine-induced conditioned place preference in rats. Behav Pharmacol 2018; 29:385-392. [DOI: 10.1097/fbp.0000000000000375] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
94
|
Fortin SM, Roitman MF. Challenges to Body Fluid Homeostasis Differentially Recruit Phasic Dopamine Signaling in a Taste-Selective Manner. J Neurosci 2018; 38:6841-6853. [PMID: 29934352 PMCID: PMC6070668 DOI: 10.1523/jneurosci.0399-18.2018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 06/01/2018] [Accepted: 06/05/2018] [Indexed: 01/13/2023] Open
Abstract
The internal environment of an organism must remain stable to ensure optimal performance and ultimately survival. The generation of motivated behaviors is an adaptive mechanism for defending homeostasis. Although physiological state modulates motivated behaviors, the influence of physiological state on phasic dopamine signaling, an underlying neurobiological substrate of reward-driven behavior, is underexplored. Here, we use sodium depletion and water restriction, manipulations of body fluid homeostasis, to determine the flexibility and specificity of dopamine responses. Changes in dopamine concentration were measured using fast-scan cyclic voltammetry in the nucleus accumbens shell of male rats in response to intraoral infusions of fluids that either satisfied or did not satisfy homeostatic need. Increases in dopamine concentration during intraoral infusions were observed only under conditions of physiological deficit. Furthermore, dopamine increases were selective and limited to those that satisfied the need state of the animal. Thus, dopamine neurons track fluid balance and respond to salt and water stimuli in a state- and taste-dependent manner. Using Fluoro-Gold tracing and immunohistochemistry for c-Fos and Foxp2, a marker of sodium-deprivation responsive neurons, we revealed brainstem populations of neurons that are activated by sodium depletion and project directly to the ventral tegmental area. The identified projections may modulate dopamine neuron excitability and consequently the state-specific dopamine release observed in our experiments. This work illustrates the impact of physiological state on mesolimbic dopamine signaling and a potential circuit by which homeostatic disruptions are communicated to mesolimbic circuitry to drive the selective reinforcement of biologically-required stimuli under conditions of physiological need.SIGNIFICANCE STATEMENT Motivated behaviors arise during physiological need and are highly selective for homeostasis-restoring stimuli. Although phasic dopamine signaling has been shown to contribute to the generation of motivated behaviors, the state and stimulus specificity of phasic dopamine signaling is less clear. These studies use thirst and sodium appetite to show that dopamine neurons dynamically track body fluid homeostasis and respond to water and salt stimuli in a state- and taste-dependent manner. We also identify hindbrain sodium deprivation-responsive neurons that project directly to the ventral tegmental area, where dopamine neuron cell bodies reside. This work demonstrates command of homeostasis over dopamine signaling and proposes a circuit by which physiological need drives motivated behavior by state- and taste-selective recruitment of phasic dopamine signaling.
Collapse
Affiliation(s)
| | - Mitchell F Roitman
- Graduate Program in Neuroscience and
- Department of Psychology, University of Illinois at Chicago (UIC), Chicago, Illinois, 60607
| |
Collapse
|
95
|
Expression and localization of CB1R, NAPE-PLD, and FAAH in the vervet monkey nucleus accumbens. Sci Rep 2018; 8:8689. [PMID: 29875385 PMCID: PMC5989267 DOI: 10.1038/s41598-018-26826-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 05/15/2018] [Indexed: 11/14/2022] Open
Abstract
Extensive rodent literature suggests that the endocannabinoid (eCB) system present in the nucleus accumbens (NAc) modulates dopamine (DA) release in this area. However, expression patterns of the cannabinoid receptor type 1 (CB1R), the synthesizing enzyme N-acyl phosphatidylethanolamine phospholipase D (NAPE-PLD), and the degradation enzyme fatty acid amide hydrolase (FAAH) in the NAc have not yet been described in non-human primates. The goal of this study is therefore to characterize the expression and localization of the eCB system within the NAc of vervet monkeys (Chlorocebus sabaeus) using Western blots and immunohistochemistry. Results show that CB1R, NAPE-PLD, and FAAH are expressed across the NAc rostrocaudal axis, both in the core and shell. CB1R, NAPE-PLD, and FAAH are localized in medium spiny neurons (MSNs) and fast-spiking GABAergic interneurons (FSIs). Dopaminergic projections and astrocytes did not express CB1R, NAPE-PLD, or FAAH. These data show that the eCB system is present in the vervet monkey NAc and supports its role in the primate brain reward circuit.
Collapse
|
96
|
Oscillatory local field potentials of the nucleus accumbens and the anterior limb of the internal capsule in heroin addicts. Clin Neurophysiol 2018; 129:1242-1253. [DOI: 10.1016/j.clinph.2018.03.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Revised: 02/23/2018] [Accepted: 03/05/2018] [Indexed: 12/22/2022]
|
97
|
Jang H, Jung K, Jeong J, Park SK, Kralik JD, Jeong J. Nucleus accumbens shell moderates preference bias during voluntary choice behavior. Soc Cogn Affect Neurosci 2018; 12:1428-1436. [PMID: 28992274 PMCID: PMC5629817 DOI: 10.1093/scan/nsx072] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 05/23/2017] [Indexed: 11/29/2022] Open
Abstract
The nucleus accumbens (NAc) shell lies anatomically at a critical intersection within the brain’s reward system circuitry, however, its role in voluntary choice behavior remains unclear. Rats with electrolytic lesions in the NAc shell were tested in a novel foraging paradigm. Over a continuous two-week period they freely chose among four nutritionally identical but differently flavored food pellets by pressing corresponding levers. We examined the lesion’s effects on three behavioral dynamics components: motivation (when to eat), preference bias (what to choose) and persistence (how long to repeat the same choice). The lesion led to a marked increase in the preference bias: i.e., increased selection of the most-preferred choice option, and decreased selection of the others. We found no effects on any other behavioral measures, suggesting no effect on motivation or choice persistence. The results implicate the NAc shell in moderating the instrumental valuation process by inhibiting excessive bias toward preferred choice options.
Collapse
Affiliation(s)
- Hyeran Jang
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Korea
| | - Kanghoon Jung
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Korea.,Department of Psychological and Brain Sciences, Dartmouth College, Hanover, NH 03755, USA
| | - Jaehoon Jeong
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Korea
| | - Sang Ki Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Korea
| | - Jerald D Kralik
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Korea.,Department of Psychological and Brain Sciences, Dartmouth College, Hanover, NH 03755, USA
| | - Jaeseung Jeong
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Korea
| |
Collapse
|
98
|
Gibson GD, Prasad AA, Jean-Richard-dit-Bressel P, Yau JO, Millan EZ, Liu Y, Campbell EJ, Lim J, Marchant NJ, Power JM, Killcross S, Lawrence AJ, McNally GP. Distinct Accumbens Shell Output Pathways Promote versus Prevent Relapse to Alcohol Seeking. Neuron 2018; 98:512-520.e6. [DOI: 10.1016/j.neuron.2018.03.033] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 01/30/2018] [Accepted: 03/19/2018] [Indexed: 12/22/2022]
|
99
|
Tonn Eisinger KR, Larson EB, Boulware MI, Thomas MJ, Mermelstein PG. Membrane estrogen receptor signaling impacts the reward circuitry of the female brain to influence motivated behaviors. Steroids 2018; 133:53-59. [PMID: 29195840 PMCID: PMC5864533 DOI: 10.1016/j.steroids.2017.11.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 11/20/2017] [Accepted: 11/23/2017] [Indexed: 12/12/2022]
Abstract
Within the adult female, estrogen signaling is well-described as an integral component of the physiologically significant hypothalamic-pituitary-gonadal axis. In rodents, the timing of ovulation is intrinsically entwined with the display of sexual receptivity. For decades, the importance of estradiol activating intracellular estrogen receptors within the hypothalamus and midbrain/spinal cord lordosis circuits has been appreciated. These signaling pathways primarily account for the ability of the female to reproduce. Yet, often overlooked is that the desire to reproduce is also tightly regulated by estrogen receptor signaling. This lack of emphasis can be attributed to an absence of nuclear estrogen receptors in brain regions associated with reward, such as the nucleus accumbens, which are associated with motivated behaviors. This review outlines how membrane-localized estrogen receptors affect metabotropic glutamate receptor signaling within the rodent nucleus accumbens. In addition, we discuss how, as estrogens drive increased motivation for reproduction, they also produce the untoward side effect of heightening female vulnerability to drug addiction.
Collapse
Affiliation(s)
- Katherine R Tonn Eisinger
- Department of Neuroscience and Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Erin B Larson
- Department of Neuroscience and Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Marissa I Boulware
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Mark J Thomas
- Department of Neuroscience and Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Paul G Mermelstein
- Department of Neuroscience and Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
100
|
Wouterlood FG, Engel A, Daal M, Houwen G, Meinderts A, Jordà Siquier T, Beliën JAM, van Dongen YC, Scheel-Krüger J, Thierry AM, Groenewegen HJ, Deniau JM. Mesencephalic dopamine neurons interfacing the shell of nucleus accumbens and the dorsolateral striatum in the rat. J Neurosci Res 2018; 96:1518-1542. [PMID: 29696690 PMCID: PMC6099426 DOI: 10.1002/jnr.24242] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 02/26/2018] [Accepted: 03/15/2018] [Indexed: 11/18/2022]
Abstract
Parallel corticostriatonigral circuits have been proposed that separately process motor, cognitive, and emotional‐motivational information. Functional integration requires that interactions exist between neurons participating in these circuits. This makes it imperative to study the complex anatomical substrate underlying corticostriatonigral circuits. It has previously been proposed that dopaminergic neurons in the ventral mesencephalon may play a role in this circuit interaction. Therefore, we studied in rats convergence of basal ganglia circuits by depositing an anterograde neuroanatomical tracer into the ventral striatum together with a retrograde fluorescent tracer ipsilaterally in the dorsolateral striatum. In the mesencephalon, using confocal microscopy, we looked for possible appositions of anterogradely labeled fibers and retrogradely labeled neurons, “enhancing” the latter via intracellular injection of Lucifer Yellow. Tyrosine hydroxylase (TH) immunofluorescence served to identify dopaminergic neurons. In neurophysiological experiments, we combined orthodromic stimulation in the medial ventral striatum with recording from ventral mesencephalic neurons characterized by antidromic stimulation from the dorsal striatum. We observed terminal fields of anterogradely labeled fibers that overlap populations of retrogradely labeled nigrostriatal cell bodies in the substantia nigra pars compacta and lateral ventral tegmental area (VTA), with numerous close appositions between boutons of anterogradely labeled fibers and nigrostriatal, TH‐immunopositive neurons. Neurophysiological stimulation in the medial ventral striatum caused inhibition of dopaminergic nigrostriatal neurons projecting to the ventrolateral striatal territory. Responding nigrostriatal neurons were located in the medial substantia nigra and adjacent VTA. Our results strongly suggest a functional link between ventromedial, emotional‐motivational striatum, and the sensorimotor dorsal striatum via dopaminergic nigrostriatal neurons.
Collapse
Affiliation(s)
- Floris G Wouterlood
- Department of Anatomy & Neurosciences, VU University Medical Center, Neuroscience Campus Amsterdam, 1007 MB Amsterdam, The Netherlands
| | - Angela Engel
- Department of Anatomy & Neurosciences, VU University Medical Center, Neuroscience Campus Amsterdam, 1007 MB Amsterdam, The Netherlands
| | - Mariah Daal
- Department of Anatomy & Neurosciences, VU University Medical Center, Neuroscience Campus Amsterdam, 1007 MB Amsterdam, The Netherlands
| | - Gertjan Houwen
- Department of Anatomy & Neurosciences, VU University Medical Center, Neuroscience Campus Amsterdam, 1007 MB Amsterdam, The Netherlands
| | - Aileen Meinderts
- Department of Anatomy & Neurosciences, VU University Medical Center, Neuroscience Campus Amsterdam, 1007 MB Amsterdam, The Netherlands
| | - Tomàs Jordà Siquier
- Department of Anatomy & Neurosciences, VU University Medical Center, Neuroscience Campus Amsterdam, 1007 MB Amsterdam, The Netherlands
| | - Jeroen A M Beliën
- Department of Pathology, VU University Medical Center, Neuroscience Campus Amsterdam, 1007 MB, Amsterdam, The Netherlands
| | - Yvette C van Dongen
- Department of Anatomy & Neurosciences, VU University Medical Center, Neuroscience Campus Amsterdam, 1007 MB Amsterdam, The Netherlands.,Institut National de la Santé et de la Recherche Médicale, U114, Chaire de Neuropharmacologie, Collège de France, 11 Place Marcelin Berthelot, 75231 Paris Cedex 05, France
| | - Jørgen Scheel-Krüger
- Department of Clinical Medicine, Center of Functionally Integrative Neuroscience, Nørrebrogade 44, 8000 Aarhus C, Denmark
| | - Anne-Marie Thierry
- Institut National de la Santé et de la Recherche Médicale, U114, Chaire de Neuropharmacologie, Collège de France, 11 Place Marcelin Berthelot, 75231 Paris Cedex 05, France
| | - Henk J Groenewegen
- Department of Anatomy & Neurosciences, VU University Medical Center, Neuroscience Campus Amsterdam, 1007 MB Amsterdam, The Netherlands
| | - Jean-Michel Deniau
- Institut National de la Santé et de la Recherche Médicale, U114, Chaire de Neuropharmacologie, Collège de France, 11 Place Marcelin Berthelot, 75231 Paris Cedex 05, France
| |
Collapse
|