1
|
Sosis B, Rubin JE. Distinct dopaminergic spike-timing-dependent plasticity rules are suited to different functional roles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.24.600372. [PMID: 38979377 PMCID: PMC11230239 DOI: 10.1101/2024.06.24.600372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Various mathematical models have been formulated to describe the changes in synaptic strengths resulting from spike-timing-dependent plasticity (STDP). A subset of these models include a third factor, dopamine, which interacts with spike timing to contribute to plasticity at specific synapses, notably those from cortex to striatum at the input layer of the basal ganglia. Theoretical work to analyze these plasticity models has largely focused on abstract issues, such as the conditions under which they may promote synchronization and the weight distributions induced by inputs with simple correlation structures, rather than on scenarios associated with specific tasks, and has generally not considered dopamine-dependent forms of STDP. In this paper we introduce three forms of dopamine-modulated STDP adapted from previously proposed plasticity rules. We then analyze, mathematically and with simulations, their performance in three biologically relevant scenarios. We test the ability of each of the three models to maintain its weights in the face of noise and to complete simple reward prediction and action selection tasks, studying the learned weight distributions and corresponding task performance in each setting. Interestingly, we find that each plasticity rule is well suited to a subset of the scenarios studied but falls short in others. Different tasks may therefore require different forms of synaptic plasticity, yielding the prediction that the precise form of the STDP mechanism present may vary across regions of the striatum, and other brain areas impacted by dopamine, that are involved in distinct computational functions.
Collapse
Affiliation(s)
- Baram Sosis
- *Department of Mathematics, University of Pittsburgh, 301 Thackeray Hall, Pittsburgh, 15260, PA, USA
| | - Jonathan E. Rubin
- *Department of Mathematics, University of Pittsburgh, 301 Thackeray Hall, Pittsburgh, 15260, PA, USA
- Center for the Neural Basis of Cognition, University of Pittsburgh, 4400 Fifth Ave, Pittsburgh, 15213, PA, USA
| |
Collapse
|
2
|
Hoisington ZW, Salvi A, Laguesse S, Ehinger Y, Shukla C, Phamluong K, Ron D. The Small G-Protein Rac1 in the Dorsomedial Striatum Promotes Alcohol-Dependent Structural Plasticity and Goal-Directed Learning in Mice. J Neurosci 2024; 44:e1644232024. [PMID: 38886056 PMCID: PMC11255432 DOI: 10.1523/jneurosci.1644-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 04/04/2024] [Accepted: 04/10/2024] [Indexed: 06/20/2024] Open
Abstract
The small G-protein Ras-related C3 botulinum toxin substrate 1 (Rac1) promotes the formation of filamentous actin (F-actin). Actin is a major component of dendritic spines, and we previously found that alcohol alters actin composition and dendritic spine structure in the nucleus accumbens (NAc) and the dorsomedial striatum (DMS). To examine if Rac1 contributes to these alcohol-mediated adaptations, we measured the level of GTP-bound active Rac1 in the striatum of mice following 7 weeks of intermittent access to 20% alcohol. We found that chronic alcohol intake activates Rac1 in the DMS of male mice. In contrast, Rac1 is not activated by alcohol in the NAc and DLS of male mice or in the DMS of female mice. Similarly, closely related small G-proteins are not activated by alcohol in the DMS, and Rac1 activity is not increased in the DMS by moderate alcohol or natural reward. To determine the consequences of alcohol-dependent Rac1 activation in the DMS of male mice, we inhibited endogenous Rac1 by infecting the DMS of mice with an adeno-associated virus (AAV) expressing a dominant negative form of the small G-protein (Rac1-DN). We found that overexpression of AAV-Rac1-DN in the DMS inhibits alcohol-mediated Rac1 signaling and attenuates alcohol-mediated F-actin polymerization, which corresponded with a decrease in dendritic arborization and spine maturation. Finally, we provide evidence to suggest that Rac1 in the DMS plays a role in alcohol-associated goal-directed learning. Together, our data suggest that Rac1 in the DMS plays an important role in alcohol-dependent structural plasticity and aberrant learning.
Collapse
Affiliation(s)
- Zachary W Hoisington
- Alcohol and Addiction Research Group, Department of Neurology, University of California San Francisco, San Francisco, California 94107
| | - Alexandra Salvi
- Alcohol and Addiction Research Group, Department of Neurology, University of California San Francisco, San Francisco, California 94107
| | - Sophie Laguesse
- GIGA-Stem Cells and GIGA-Neurosciences, Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège, Liège 4000, Belgium
| | - Yann Ehinger
- Alcohol and Addiction Research Group, Department of Neurology, University of California San Francisco, San Francisco, California 94107
| | - Chhavi Shukla
- Alcohol and Addiction Research Group, Department of Neurology, University of California San Francisco, San Francisco, California 94107
| | - Khanhky Phamluong
- Alcohol and Addiction Research Group, Department of Neurology, University of California San Francisco, San Francisco, California 94107
| | - Dorit Ron
- Alcohol and Addiction Research Group, Department of Neurology, University of California San Francisco, San Francisco, California 94107
| |
Collapse
|
3
|
Gupta S, Tielemans A, Guevara CA, Huntley GW, Benson DL. Parkinson's-linked LRRK2-G2019S derails AMPAR trafficking, mobility, and composition in striatum with cell-type and subunit specificity. Proc Natl Acad Sci U S A 2024; 121:e2317833121. [PMID: 38968112 PMCID: PMC11252801 DOI: 10.1073/pnas.2317833121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 05/27/2024] [Indexed: 07/07/2024] Open
Abstract
Parkinson's disease (PD) is a multifactorial disease that affects multiple brain systems and circuits. While defined by motor symptoms caused by degeneration of brainstem dopamine neurons, debilitating non-motor abnormalities in fronto-striatal-based cognitive function are common, appear early, and are initially independent of dopamine. Young adult mice expressing the PD-associated G2019S missense mutation in Lrrk2 also exhibit deficits in fronto-striatal-based cognitive tasks. In mice and humans, cognitive functions require dynamic adjustments in glutamatergic synapse strength through cell-surface trafficking of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid-type glutamate receptors (AMPARs), but it is unknown how LRRK2 mutation impacts dynamic features of AMPAR trafficking in striatal projection neurons (SPNs). Here, we used Lrrk2G2019S knockin mice to show that surface AMPAR subunit stoichiometry is altered biochemically and functionally in mutant SPNs in dorsomedial striatum to favor the incorporation of GluA1 over GluA2. GluA1-containing AMPARs were resistant to internalization from the cell surface, leaving an excessive accumulation of GluA1 on the surface within and outside synapses. This negatively impacted trafficking dynamics that normally support synapse strengthening, as GluA1-containing AMPARs failed to increase at synapses in response to a potentiating stimulus and showed significantly reduced surface mobility. Surface GluA2-containing AMPARs were expressed at normal levels in synapses, indicating subunit-selective impairment. Abnormal surface accumulation of GluA1 was independent of PKA activity and was limited to D1R SPNs. Since LRRK2 mutation is thought to be part of a common PD pathogenic pathway, our data suggest that sustained, striatal cell-type specific changes in AMPAR composition and trafficking contribute to cognitive or other impairments associated with PD.
Collapse
Affiliation(s)
- Swati Gupta
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY10029
| | - Alexander Tielemans
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY10029
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY10029
| | - Christopher A. Guevara
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY10029
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY10029
| | - George W. Huntley
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY10029
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY10029
| | - Deanna L. Benson
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY10029
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY10029
| |
Collapse
|
4
|
Tanner MK, Hohorst AA, Westerman JD, Mendoza CS, Han R, Moya NA, Jaime J, Alvarez LM, Dryden MQ, Balolia A, Abdul RA, Loetz EC, Greenwood BN. Pharmacological manipulations of the dorsomedial and dorsolateral striatum during fear extinction reveal opposing roles in fear renewal. Neurobiol Learn Mem 2024; 212:107937. [PMID: 38735637 PMCID: PMC11187715 DOI: 10.1016/j.nlm.2024.107937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/17/2024] [Accepted: 05/09/2024] [Indexed: 05/14/2024]
Abstract
Systemic manipulations that enhance dopamine (DA) transmission around the time of fear extinction can strengthen fear extinction and reduce conditioned fear relapse. Prior studies investigating the brain regions where DA augments fear extinction focus on targets of mesolimbic and mesocortical DA systems originating in the ventral tegmental area, given the role of these DA neurons in prediction error. The dorsal striatum (DS), a primary target of the nigrostriatal DA system originating in the substantia nigra (SN), is implicated in behaviors beyond its canonical role in movement, such as reward and punishment, goal-directed action, and stimulus-response associations, but whether DS DA contributes to fear extinction is unknown. We have observed that chemogenetic stimulation of SN DA neurons during fear extinction prevents the return of fear in contexts different from the extinction context, a form of relapse called renewal. This effect of SN DA stimulation is mimicked by a DA D1 receptor (D1R) agonist injected into the DS, thus implicating DS DA in fear extinction. Different DS subregions subserve unique functions of the DS, but it is unclear where in the DS D1R agonist acts during fear extinction to reduce renewal. Furthermore, although fear extinction increases neural activity in DS subregions, whether neural activity in DS subregions is causally involved in fear extinction is unknown. To explore the role of DS subregions in fear extinction, adult, male Long-Evans rats received microinjections of either the D1R agonist SKF38393 or a cocktail consisting of GABAA/GABAB receptor agonists muscimol/baclofen selectively into either dorsomedial (DMS) or dorsolateral (DLS) DS subregions immediately prior to fear extinction, and extinction retention and renewal were subsequently assessed drug-free. While increasing D1R signaling in the DMS during fear extinction did not impact fear extinction retention or renewal, DMS inactivation reduced later renewal. In contrast, DLS inactivation had no effect on fear extinction retention or renewal but increasing D1R signaling in the DLS during extinction reduced fear renewal. These data suggest that DMS and DLS activity during fear extinction can have opposing effects on later fear renewal, with the DMS promoting renewal and the DLS opposing renewal. Mechanisms through which the DS could influence the contextual gating of fear extinction are discussed.
Collapse
Affiliation(s)
- Margaret K Tanner
- Department of Psychology, University of Colorado Denver, Denver, CO, USA
| | - Alyssa A Hohorst
- Department of Integrative Biology, University of Colorado Denver, Denver, CO, USA
| | | | | | - Rebecca Han
- Department of Psychology, University of Colorado Denver, Denver, CO, USA
| | - Nicolette A Moya
- Department of Neuroscience, Northwestern Feinberg School of Medicine, Chicago, IL, USA
| | - Jennifer Jaime
- The Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lareina M Alvarez
- Department of Integrative Biology, University of Colorado Denver, Denver, CO, USA
| | - Miles Q Dryden
- Department of Psychology, University of Colorado Denver, Denver, CO, USA
| | - Aleezah Balolia
- Neuroscience Program, University of Colorado Anschutz Medical Campus, Denver, CO, USA
| | - Remla A Abdul
- Department of Psychology, University of Colorado Denver, Denver, CO, USA
| | - Esteban C Loetz
- Department of Psychology, University of Colorado Denver, Denver, CO, USA
| | | |
Collapse
|
5
|
Dong J, Wang L, Sullivan BT, Sun L, Chang L, Martinez Smith VM, Ding J, Le W, Gerfen CR, Cai H. Patch and matrix striatonigral neurons differentially regulate locomotion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.12.598675. [PMID: 38915717 PMCID: PMC11195204 DOI: 10.1101/2024.06.12.598675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Striatonigral neurons, known to promote locomotion, reside in both the patch and matrix compartments of the dorsal striatum. However, their compartment-specific contributions to locomotion remain largely unexplored. Using molecular identifier Kremen1 and Calb1 , we showed in mouse models that patch and matrix striatonigral neurons exert opposite influences on locomotion. Matrix striatonigral neurons reduced their activity before the cessation of self-paced locomotion, while patch striatonigral neuronal activity increased, suggesting an inhibitory function. Indeed, optogenetic activation of patch striatonigral neurons suppressed ongoing locomotion with reduced striatal dopamine release, contrasting with the locomotion-promoting effect of matrix striatonigral neurons, which showed an initial increase in dopamine release. Furthermore, genetic deletion of the GABA-B receptor in Aldehyde dehydrogenase 1A1-positive (ALDH1A1 + ) nigrostriatal dopaminergic neurons completely abolished the locomotion-suppressing effect of patch striatonigral neurons. Our findings unravel a compartment-specific mechanism governing locomotion in the dorsal striatum, where patch striatonigral neurons suppress locomotion by inhibiting ALDH1A1 + nigrostriatal dopaminergic neurons.
Collapse
|
6
|
Song N, Liu Z, Gao Y, Lu S, Yang S, Yuan C. NAc-DBS corrects depression-like behaviors in CUMS mouse model via disinhibition of DA neurons in the VTA. Mol Psychiatry 2024; 29:1550-1566. [PMID: 38361128 DOI: 10.1038/s41380-024-02476-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 02/17/2024]
Abstract
Major depressive disorder (MDD) is characterized by diverse debilitating symptoms that include loss of motivation and anhedonia. If multiple medications, psychotherapy, and electroconvulsive therapy fail in some patients with MDD, their condition is then termed treatment-resistant depression (TRD). MDD can be associated with abnormalities in the reward-system-dopaminergic mesolimbic pathway, in which the nucleus accumbens (NAc) and ventral tegmental area (VTA) play major roles. Deep brain stimulation (DBS) applied to the NAc alleviates the depressive symptoms of MDD. However, the mechanism underlying the effects of this DBS has remained elusive. In this study, using the chronic unpredictable mild stress (CUMS) mouse model, we investigated the behavioral and neurobiological effects of NAc-DBS on the multidimensional depression-like phenotypes induced by CUMS by integrating behavioral, in vivo microdialysis coupled with high-performance liquid chromatography-electrochemical detector (HPLC-ECD), calcium imaging, pharmacological, and genetic manipulation methods in freely moving mice. We found that long-term and repeated, but not single, NAc-DBS induced robust antidepressant responses in CUMS mice. Moreover, even a single trial NAc-DBS led to the elevation of the γ-aminobutyric acid (GABA) neurotransmitter, accompanied by the increase in dopamine (DA) neuron activity in the VTA. Both the inhibition of the GABAA receptor activity and knockdown of the GABAA-α1 gene in VTA-GABA neurons blocked the antidepressant effect of NAc-DBS in CUMS mice. Our results showed that NAc-DBS could disinhibit VTA-DA neurons by regulating the level of GABA and the activity of VTA-GABA in the VTA and could finally correct the depression-like behaviors in the CUMS mouse model.
Collapse
Affiliation(s)
- Nan Song
- Center of Cognition and Brain Science, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China.
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China.
| | - Zhenhong Liu
- Institute for Brain Disorders, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Yan Gao
- Center of Cognition and Brain Science, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Shanshan Lu
- Center of Cognition and Brain Science, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Shenglian Yang
- Center of Cognition and Brain Science, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Chao Yuan
- Center of Cognition and Brain Science, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| |
Collapse
|
7
|
Hoisington ZW, Salvi A, Laguesse S, Ehinger Y, Shukla C, Phamluong K, Ron D. The small G-protein Rac1 in the dorsomedial striatum promotes alcohol-dependent structural plasticity and goal-directed learning in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.30.555562. [PMID: 37693512 PMCID: PMC10491244 DOI: 10.1101/2023.08.30.555562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
The small G-protein Rac1 promotes the formation of filamentous actin (F-Actin). Actin is a major component of dendritic spines, and we previously found that alcohol alters actin composition and dendritic spine structure in the nucleus accumbens (NAc) and the dorsomedial striatum (DMS). To examine if Rac1 contributes to these alcohol-mediated adaptations, we measured the level of GTP-bound active Rac1 in the striatum of mice following 7 weeks of intermittent access to 20% alcohol. We found that chronic alcohol intake activates Rac1 in the DMS of male mice. In contrast, Rac1 is not activated by alcohol in the NAc and DLS of male mice, or in the DMS of female mice. Similarly, closely related small G-proteins are not activated by alcohol in the DMS, and Rac1 activity is not increased in the DMS by moderate alcohol or natural reward. To determine the consequences of alcohol-dependent Rac1 activation in the DMS of male mice, we inhibited endogenous Rac1 by infecting the DMS of mice with an AAV expressing a dominant negative form of the small G-protein (Rac1-DN). We found that overexpression of AAV-Rac1-DN in the DMS inhibits alcohol-mediated Rac1 signaling and attenuates alcohol-mediated F-actin polymerization, which corresponded with a decrease in dendritic arborization and spine maturation. Finally, we provide evidence to suggest that Rac1 in the DMS plays a role in alcohol-associated goal-directed learning. Together, our data suggest that Rac1 in the DMS plays an important role in alcohol-dependent structural plasticity and aberrant learning. Significance Statement Addiction, including alcohol use disorder, is characterized by molecular and cellular adaptations that promote maladaptive behaviors. We found that Rac1 was activated by alcohol in the dorsomedial striatum (DMS) of male mice. We show that alcohol-mediated Rac1 signaling is responsible for alterations in actin dynamics and neuronal morphology. We also present data to suggest that Rac1 is important for alcohol-associated learning processes. These results suggest that Rac1 in the DMS is an important contributor to adaptations that promote alcohol use disorder.
Collapse
|
8
|
Wu M, Liu F, Wang H, Yao L, Wei C, Zheng Q, Han J, Liu Z, Liu Y, Duan H, Ren W, Sun Z. Characterizing the dynamic learning process: Implications of a quantitative analysis. Behav Brain Res 2024; 463:114915. [PMID: 38368954 DOI: 10.1016/j.bbr.2024.114915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 02/05/2024] [Accepted: 02/15/2024] [Indexed: 02/20/2024]
Abstract
Understanding the neural mechanisms involved in learning processes is crucial for unraveling the complexities of behavior and cognition. Sudden change from the untrained level to the fully-learned level is a pivotal feature of instrumental learning. However, the concept of change point and suitable methods to conveniently analyze the characteristics of sudden change in groups remain elusive, which might hinder a fuller understanding of the neural mechanism underlying dynamic leaning process. In the current study, we investigated the learning processes of mice that were trained in an aversive instrumental learning task, and introduced a novel strategy to analyze behavioral variations in instrumental learning, leading to improved clarity on the concept of sudden change and enabling comprehensive group analysis. By applying this novel strategy, we examined the effects of cocaine and a cannabinoid receptor agonist on instrumental learning. Intriguingly, our analysis revealed significant differences in timing and occurrence of sudden changes that were previously overlooked using traditional analysis. Overall, our research advances understanding of behavioral variation during instrumental learning and the interplay between learning behaviors and neurotransmitter systems, contributing to a deeper comprehension of learning processes and informing future investigations and therapeutic interventions.
Collapse
Affiliation(s)
- Meilin Wu
- MOE Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi'an 710062, China
| | - Fuhong Liu
- MOE Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi'an 710062, China
| | - Hao Wang
- College of Life Sciences, Shaanxi Normal University, Xi'an 710062, China
| | - Li Yao
- MOE Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi'an 710062, China
| | - Chunling Wei
- MOE Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi'an 710062, China
| | - Qiaohua Zheng
- MOE Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi'an 710062, China
| | - Jing Han
- MOE Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi'an 710062, China
| | - Zhiqiang Liu
- MOE Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi'an 710062, China
| | - Yihui Liu
- MOE Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi'an 710062, China
| | - Haijun Duan
- MOE Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi'an 710062, China
| | - Wei Ren
- MOE Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi'an 710062, China; Faculty of Education, Shaanxi Normal University, Xi'an 710062, China.
| | - Zongpeng Sun
- School of Psychology, Shaanxi Normal University, Xi'an 710062, China.
| |
Collapse
|
9
|
Roth AM, Lokesh R, Tang J, Buggeln JH, Smith C, Calalo JA, Sullivan SR, Ngo T, Germain LS, Carter MJ, Cashaback JGA. Punishment Leads to Greater Sensorimotor Learning But Less Movement Variability Compared to Reward. Neuroscience 2024; 540:12-26. [PMID: 38220127 PMCID: PMC10922623 DOI: 10.1016/j.neuroscience.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 01/16/2024]
Abstract
When a musician practices a new song, hitting a correct note sounds pleasant while striking an incorrect note sounds unpleasant. Such reward and punishment feedback has been shown to differentially influence the ability to learn a new motor skill. Recent work has suggested that punishment leads to greater movement variability, which causes greater exploration and faster learning. To further test this idea, we collected 102 participants over two experiments. Unlike previous work, in Experiment 1 we found that punishment did not lead to faster learning compared to reward (n = 68), but did lead to a greater extent of learning. Surprisingly, we also found evidence to suggest that punishment led to less movement variability, which was related to the extent of learning. We then designed a second experiment that did not involve adaptation, allowing us to further isolate the influence of punishment feedback on movement variability. In Experiment 2, we again found that punishment led to significantly less movement variability compared to reward (n = 34). Collectively our results suggest that punishment feedback leads to less movement variability. Future work should investigate whether punishment feedback leads to a greater knowledge of movement variability and or increases the sensitivity of updating motor actions.
Collapse
Affiliation(s)
- Adam M Roth
- Department of Mechanical Engineering, University of Delaware, United States
| | - Rakshith Lokesh
- Department of Biomedical Engineering, University of Delaware, United States
| | - Jiaqiao Tang
- Department of Kinesiology, McMaster University, Canada
| | - John H Buggeln
- Department of Biomedical Engineering, University of Delaware, United States
| | - Carly Smith
- Department of Biomedical Engineering, University of Delaware, United States
| | - Jan A Calalo
- Department of Mechanical Engineering, University of Delaware, United States
| | - Seth R Sullivan
- Department of Biomedical Engineering, University of Delaware, United States
| | - Truc Ngo
- Department of Biomedical Engineering, University of Delaware, United States
| | | | | | - Joshua G A Cashaback
- Department of Mechanical Engineering, University of Delaware, United States; Department of Biomedical Engineering, University of Delaware, United States; Kinesiology and Applied Physiology, University of Delaware, United States; Interdisciplinary Neuroscience Graduate Program, University of Delaware, United States; Biomechanics and Movement Science Program, University of Delaware, United States; Department of Kinesiology, McMaster University, Canada.
| |
Collapse
|
10
|
Derman RC, Bryda EC, Ferrario CR. Role of nucleus accumbens D1-type medium spiny neurons in the expression and extinction of sign-tracking. Behav Brain Res 2024; 459:114768. [PMID: 37984521 PMCID: PMC10842774 DOI: 10.1016/j.bbr.2023.114768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/31/2023] [Accepted: 11/16/2023] [Indexed: 11/22/2023]
Abstract
While sign-tracking, also known as autoshaping, has been studied for many decades, only recently has the tendency to show sign-tracking behavior been linked to the development and persistence of addiction. Sign-tracking is dependent upon dopamine activity in the nucleus accumbens (NAc). The NAc is comprised predominantly of medium spiny projection neurons (MSN) that can be differentiated by their D1-like or D2-like dopamine receptor expression. Here we determined how reducing activity of D1-type MSNs in the NAc affects the expression and extinction of sign-tracking. To address this, we transfected the NAc of transgenic male and female rats that selectively express Cre recombinase in D1-type MSNs with a DIO viral vector expressing hM4Di. Cre- rats were given the same viral infusion but did not express the hM4Di receptor and therefore served as controls. Rats were then conditioned to associate lever presentations with pellet delivery. After sign-tracking was established, all rats were administered clozapine-n-oxide (CNO) prior to three additional conditioning sessions to assess the effects of NAc D1-MSNs inhibition on sign-tracking in the presence of reward. CNO treatment did not alter the expression of sign-tracking in Cre+ or Cre- rats. Next rats underwent extinction training where lever presentations occurred without pellet delivery and all rats received a CNO injection prior to each extinction session. In these extinction conditions, Cre+ rats exhibited robust extinction of sign-tracking across sessions, whereas Cre- rats did not. To determine if D1-MSN inhibition merely produced a temporary cessation of sign-tracking or instead had facilitated a persistent loss of sign-tracking, we evaluated the reemergence of sign-tracking in a test for reconditioning. During testing, reintroduction of the CS-US pairing did not promote the reemergence of sign-tracking in Cre+ rats, but restored sign-tracking in Cre- rats. Thus, chemogenetic inhibition of NAc D1-MSNs promoted extinction of sign-tracking. Collectively, these data suggest that D1-MSNs play an important role in resistance to extinction that typifies sign-tracking behavior.
Collapse
Affiliation(s)
- Rifka C Derman
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA; Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR 97239, USA.
| | - Elizabeth C Bryda
- Rat Resource and Research Center, Animal Modeling Core, Veterinary Pathobiology, University of Missouri, Columbia, MO 65201, USA
| | - Carrie R Ferrario
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA; Department of Pharmacology, Psychology Department, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
11
|
Gupta S, Guevara CA, Tielemans A, Huntley GW, Benson DL. Parkinson's-linked LRRK2-G2019S derails AMPAR trafficking, mobility and composition in striatum with cell-type and subunit specificity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.13.562231. [PMID: 37905106 PMCID: PMC10614818 DOI: 10.1101/2023.10.13.562231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Parkinson's (PD) is a multi-factorial disease that affects multiple brain systems and circuits. While defined by motor symptoms caused by degeneration of brainstem dopamine neurons, debilitating non-motor abnormalities in fronto-striatal based cognitive function are common, appear early and are initially independent of dopamine. Young adult mice expressing the PD-associated G2019S missense mutation in Lrrk2 also exhibit deficits in fronto-striatal-based cognitive tasks. In mice and humans, cognitive functions require dynamic adjustments in glutamatergic synapse strength through cell-surface trafficking of AMPA-type glutamate receptors (AMPARs), but it is unknown how LRRK2 mutation impacts dynamic features of AMPAR trafficking in striatal projection neurons (SPNs). Here, we used Lrrk2 G2019S knockin mice to show that surface AMPAR subunit stoichiometry is altered biochemically and functionally in mutant SPNs to favor incorporation of GluA1 over GluA2. GluA1-containing AMPARs were resistant to internalization from the cell surface, leaving an excessive accumulation of GluA1 on the surface within and outside synapses. This negatively impacted trafficking dynamics that normally support synapse strengthening, as GluA1-containing AMPARs failed to increase at synapses in response to a potentiating stimulus and showed significantly reduced surface mobility. Surface GluA2-containing AMPARs were expressed at normal levels in synapses, indicating subunit-selective impairment. Abnormal surface accumulation of GluA1 was independent of PKA activity and was limited to D 1 R SPNs. Since LRRK2 mutation is thought to be part of a common PD pathogenic pathway, our data suggest that sustained, striatal cell-type specific changes in AMPAR composition and trafficking contribute to cognitive or other impairments associated with PD. SIGNIFICANCE STATEMENT Mutations in LRRK2 are common genetic risks for PD. Lrrk2 G2019S mice fail to exhibit long-term potentiation at corticostriatal synapses and show significant deficits in frontal-striatal based cognitive tasks. While LRRK2 has been implicated generally in protein trafficking, whether G2019S derails AMPAR trafficking at synapses on striatal neurons (SPNs) is unknown. We show that surface GluA1-AMPARs fail to internalize and instead accumulate excessively within and outside synapses. This effect is selective to D 1 R SPNs and negatively impacts synapse strengthening as GluA1-AMPARs fail to increase at the surface in response to potentiation and show limited surface mobility. Thus, LRRK2-G2019S narrows the effective range of plasticity mechanisms, supporting the idea that cognitive symptoms reflect an imbalance in AMPAR trafficking mechanisms within cell-type specific projections.
Collapse
|
12
|
Xie X, Chen R, Wang X, Smith L, Wang J. Activity-dependent labeling and manipulation of fentanyl-recruited striatal ensembles using ArcTRAP approach. STAR Protoc 2023; 4:102369. [PMID: 37354458 PMCID: PMC10320278 DOI: 10.1016/j.xpro.2023.102369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/27/2023] [Accepted: 05/17/2023] [Indexed: 06/26/2023] Open
Abstract
Understanding the memory substrates underlying substance abuse requires the permanent tagging and manipulation of drug-recruited neural ensembles. Here, we present a protocol for activity-dependent labeling and chemogenetic manipulation of fentanyl-activated striatal ensembles using the ArcTRAP approach. We outline the necessary steps to breed ArcTRAP mice, prepare drugs and reagents, conduct behavioral training, and perform tagging and manipulation. This approach can be adapted to investigate drug-recruited ensembles in other brain regions, providing a versatile tool for exploring the neural mechanisms underlying addiction. For complete details on the use and execution of this protocol, please refer to Wang et al.1.
Collapse
Affiliation(s)
- Xueyi Xie
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA.
| | - Ruifeng Chen
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA
| | - Xuehua Wang
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA
| | - Laura Smith
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA
| | - Jun Wang
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA.
| |
Collapse
|
13
|
Djientcheu Tientcheu JP, Ngueguim Tsofack F, Gounoue RK, Fifen RN, Dzeufiet PDD, Dimo T. The Aqueous Extract of Sclerocarya birrea, Nauclea latifolia, and Piper longum Mixture Protects Striatal Neurons and Movement-Associated Functionalities in a Rat Model of Diabetes-Induced Locomotion Dysfunction. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2023; 2023:7865919. [PMID: 37441190 PMCID: PMC10335873 DOI: 10.1155/2023/7865919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 05/30/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023]
Abstract
Among the many complications of type 2 diabetes (T2D), locomotor disorders have been poorly studied and understood. Therefore, no disease-modifying treatment is usually considered. The study aimed to investigate the effect of the aqueous extract of Sclerocarya birrea, Nauclea latifolia, and Piper longum (SNP) mixture on locomotor activity in fructose/streptozotocin-induced diabetic rats. T2D was induced by 10% fructose orally (6 weeks) and streptozotocin (STZ, 35 mg/kg, i.v.) in 25 male rats. Diabetic animals received distilled water, metformin (200 mg/kg), or the aqueous extract of the SNP mixture (75, 150, or 300 mg/kg). A 10-minute open field test was performed in diabetic rats (glycemia: 126 and 350 mg/dL) to assess locomotor activity before and after treatment. A group of 5 normal rats (NC) served as controls throughout the study. Rats were sacrificed, and the striatum was removed for biochemical and histological studies. In untreated diabetic rats, fructose/STZ administration resulted in hyperglycemia that altered locomotor function as characterized by increased freezing time, decreased mobility time, number of lines crossed, and total travel time compared to NC. MDA, TNF-α, INF-γ, and nitrite levels were elevated in the striatum of diabetic rats, while catalase activity and GSH levels were decreased, indicating oxidative stress and neuroinflammatory changes. In untreated diabetic rats, the microstructure of the HE-stained striatum revealed lipid vacuolation (hydropic degeneration) of the parenchyma, indicating a loss of neuronal integrity. The locomotor dysfunction was significantly improved by the aqueous extract of the SNP mixture, both biochemically and histologically. As a result, our findings support the mixture's ability to correct diabetes-related locomotion disorders as a glucose-lowering product and antioxidant, anti-inflammatory, and neuroprotective agent. These results justify the use of the aqueous extract of a combination of these three plants to manage diabetes and neuroinflammatory complications in Northern Cameroon.
Collapse
Affiliation(s)
| | - Florence Ngueguim Tsofack
- Laboratory of Animal Physiology, Faculty of Science, University of Yaoundé 1, P.O. Box 812, Yaoundé, Cameroon
| | - Racéline Kamkumo Gounoue
- Laboratory of Animal Physiology, Faculty of Science, University of Yaoundé 1, P.O. Box 812, Yaoundé, Cameroon
| | - Rodrigue Ngapout Fifen
- Laboratory of Animal Physiology, Faculty of Science, University of Yaoundé 1, P.O. Box 812, Yaoundé, Cameroon
| | | | - Théophile Dimo
- Laboratory of Animal Physiology, Faculty of Science, University of Yaoundé 1, P.O. Box 812, Yaoundé, Cameroon
| |
Collapse
|
14
|
Santoscoy MC, Espinoza P, De La Cruz D, Mahamdeh M, Starr JR, Patel N, Maguire CA. An AAV capsid increases transduction of striatum and a ChAT promoter allows selective cholinergic neuron transduction. Mol Ther Methods Clin Dev 2023; 29:532-540. [PMID: 37359416 PMCID: PMC10285237 DOI: 10.1016/j.omtm.2023.05.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 05/04/2023] [Indexed: 06/28/2023]
Abstract
Adeno-associated virus (AAV) vectors are currently the most efficient option for intracranial gene therapies to treat neurodegenerative disease. Increased efficacy and safety will depend upon robust and specific expression of therapeutic genes into target cell-types within the human brain. In this study, we set out with two objectives: (1) to identify capsids with broader transduction of the striatum upon intracranial injection in mice and (2) to test a truncated human choline acetyltransferase (ChAT) promoter that would allow efficient and selective transduction of cholinergic neurons. We compared AAV9 and an engineered capsid, AAV-S, to mediate widespread reporter gene expression throughout the striatum. We observed that AAV-S transduced a significantly greater area of the injected hemisphere primarily in the rostral direction compared with AAV9 (CAG promoter). We tested AAV9 vectors packaging a reporter gene expression cassette driven by either the ChAT or CAG promoter. Specificity of transgene expression of ChAT neurons over other cells was 7-fold higher, and efficiency was 3-fold higher for the ChAT promoter compared with the CAG promoter. The AAV-ChAT transgene expression cassette should be a useful tool for the study of cholinergic neurons in mice, and the broader transduction area of AAV-S warrants further evaluation of this capsid.
Collapse
Affiliation(s)
- Miguel C. Santoscoy
- Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown, MA, USA
| | - Paula Espinoza
- Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown, MA, USA
| | - Demitri De La Cruz
- Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown, MA, USA
| | - Mohammed Mahamdeh
- Harvard Medical School, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA, USA
| | - Jacqueline R. Starr
- Harvard Medical School, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Nikita Patel
- Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown, MA, USA
| | - Casey A. Maguire
- Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown, MA, USA
| |
Collapse
|
15
|
Jesús Trujillo M, Ilarraz C, Kasanetz F. Plasticity of cortico-striatal neurons of the caudal anterior cingulate cortex during experimental neuropathic pain. Neuroscience 2023:S0306-4522(23)00230-0. [PMID: 37236391 DOI: 10.1016/j.neuroscience.2023.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/26/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023]
Abstract
Maladaptive neuronal plasticity is a main mechanism for the development and maintenance of pathological pain. Affective, motivational and cognitive deficits that are comorbid with pain involve cellular and synaptic modifications in the anterior cingulate cortex (ACC), a major brain mediator of pain perception. Here we use a model of neuropathic pain (NP) in male mice and ex-vivo electrophysiology to investigate whether layer 5 caudal ACC (cACC) neurons projecting to the dorsomedial striatum (DMS), a critical region for motivational regulation of behavior, are involved in aberrant neuronal plasticity. We found that while the intrinsic excitability of cortico-striatal cACC neurons (cACC-CS) was preserved in NP animals, excitatory postsynaptic potentials (EPSP) induced after stimulation of distal inputs were enlarged. The highest synaptic responses were evident both after single stimuli and in each of the EPSP that compose responses to trains of stimuli, and were accompanied by increased synaptically-driven action potentials. EPSP temporal summation was intact in ACC-CS neurons from NP mice, suggesting that the plastic changes were not due to alterations in dendritic integration but rather through synaptic mechanisms. These results demonstrate for the first time that NP affects cACC neurons that project to the DMS and reinforce the notion that maladaptive plasticity of the cortico-striatal pathway may be a key factor in sustaining pathological pain.
Collapse
Affiliation(s)
- María Jesús Trujillo
- Universidad de Buenos Aires, Facultad de Ciencias Médicas, Departamento de Ciencias Fisiológicas, Grupo de Neurociencia de Sistemas, Buenos Aires, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay), Buenos Aires, Argentina
| | - Constanza Ilarraz
- Universidad de Buenos Aires, Facultad de Ciencias Médicas, Departamento de Ciencias Fisiológicas, Grupo de Neurociencia de Sistemas, Buenos Aires, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay), Buenos Aires, Argentina
| | - Fernando Kasanetz
- Universidad de Buenos Aires, Facultad de Ciencias Médicas, Departamento de Ciencias Fisiológicas, Grupo de Neurociencia de Sistemas, Buenos Aires, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay), Buenos Aires, Argentina.
| |
Collapse
|
16
|
Lin J, Xu Y, Guo P, Chen YJ, Zhou J, Xia M, Tan B, Liu X, Feng H, Chen Y. CCL5/CCR5-mediated peripheral inflammation exacerbates blood‒brain barrier disruption after intracerebral hemorrhage in mice. J Transl Med 2023; 21:196. [PMID: 36918921 PMCID: PMC10015963 DOI: 10.1186/s12967-023-04044-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 03/08/2023] [Indexed: 03/16/2023] Open
Abstract
BACKGROUND Owing to metabolic disequilibrium and immune suppression, intracerebral hemorrhage (ICH) patients are prone to infections; according to a recent global analysis of stroke cases, approximately 10 million new-onset ICH patients had experienced concurrent infection. However, the intrinsic mechanisms underlying the effects of infection related peripheral inflammation after ICH remain unclear. METHODS Lipopolysaccharide (LPS) was intraperitoneally injected into ICH model mice to induce peripheral inflammation. Neurobehavioral deficits, blood‒brain barrier (BBB) disruption, and the expression of CCR5, JAK2, STAT3, and MMP9 were evaluated after treatment with recombinant CCL5 (rCCL5) (a CCR5 ligand), maraviroc (MVC) (an FDA-approved selective CCR5 antagonist), or JAK2 CRISPR plasmids. RESULTS Our study revealed that severe peripheral inflammation increased CCL5/CCR5 axis activation in multiple inflammatory cell types, including microglia, astrocytes, and monocytes, and aggravated BBB disruption and neurobehavioral dysfunction after ICH, possibly in part through the JAK2/STAT3 signaling pathway. CONCLUSIONS CCR5 might be a potential target for the clinical treatment of infection-induced exacerbation of BBB disruption following ICH.
Collapse
Affiliation(s)
- Jie Lin
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Ya Xu
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Peiwen Guo
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yù-Jié Chen
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jiru Zhou
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Min Xia
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Binbin Tan
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xin Liu
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Hua Feng
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China. .,Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China. .,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Yujie Chen
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China. .,Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China. .,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| |
Collapse
|
17
|
Task parameters influence operant response variability in mice. Psychopharmacology (Berl) 2023; 240:213-225. [PMID: 36572717 PMCID: PMC9894580 DOI: 10.1007/s00213-022-06298-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 12/12/2022] [Indexed: 12/28/2022]
Abstract
RATIONALE During operant conditioning, animals associate actions with outcomes. However, patterns and rates of operant responding change over learning, which makes it difficult to distinguish changes in learning from general changes in performance or movement. Thus, understanding how task parameters influence movement execution is essential. OBJECTIVES To understand how specific operant task parameters influenced the repetition of future operant responses, we investigated the ability of operant conditioning schedules and contingencies to promote reproducible bouts of five lever presses in mice. METHODS Mice were trained on one of the four operant tasks to test three distinct hypotheses: (1) whether a cue presented concurrently with sucrose delivery influenced the pattern of lever pressing; (2) whether requiring animals to collect earned sucrose promoted the organization of responses into bouts; and (3) whether only reinforcing bouts where interresponse time (IRT) variances were below a target promoted reproducible patterns of operant behavior. RESULTS (1) Signaling reinforcer delivery with a cue increased learning rates but resulted in mice pressing the lever in fast succession until the cue turned on, rather than executing discrete bouts. (2) Requiring mice to collect the reinforcer between bouts had little effect on behavior. (3) A training strategy that directly reinforced bouts with low variance IRTs was not more effective than a traditional fixed ratio schedule at promoting reproducible action execution. CONCLUSIONS Together, our findings provide insights into the parameters of behavioral training that promote reproducible actions and that should be carefully selected when designing operant conditioning experiments.
Collapse
|
18
|
Tanner MK, Davis JKP, Jaime J, Moya NA, Hohorst AA, Bonar K, Abrams KA, Jamil N, Han R, Hubert TJ, Brown N, Loetz EC, Greenwood BN. Duration- and sex-dependent neural circuit control of voluntary physical activity. Psychopharmacology (Berl) 2022; 239:3697-3709. [PMID: 36195731 PMCID: PMC9768838 DOI: 10.1007/s00213-022-06243-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/20/2022] [Indexed: 10/10/2022]
Abstract
RATIONALE Exercise participation remains low despite clear benefits. Rats engage in voluntary wheel running (VWR) that follows distinct phases of acquisition, during which VWR escalates, and maintenance, during which VWR remains stable. Understanding mechanisms driving acquisition and maintenance of VWR could lead to novel strategies to promote exercise. The two phases of VWR resemble those that occur during operant conditioning and, therefore, might involve similar neural substrates. The dorsomedial (DMS) dorsal striatum (DS) supports the acquisition of operant conditioning, whereas the dorsolateral striatum (DLS) supports its maintenance. OBJECTIVES Here we sought to characterize the roles of DS subregions in VWR. Females escalate VWR and operant conditioning faster than males. Thus, we also assessed for sex differences. METHODS To determine the causal role of DS subregions in VWR, we pharmacologically inactivated the DMS or DLS of adult, male and female, Long-Evans rats during the two phases of VWR. The involvement of DA receptor 1 (D1)-expressing neurons in the DS was investigated by quantifying cfos mRNA within this neuronal population. RESULTS We observed that, in males, the DMS and DLS are critical for VWR exclusively during acquisition and maintenance, respectively. In females, the DMS is also critical only during acquisition, but the DLS contributes to VWR during both VWR phases. DLS D1 neurons could be an important driver of VWR escalation during acquisition. CONCLUSIONS The acquisition and maintenance of VWR involve unique neural substrates in the DS that vary by sex. Results reveal targets for sex-specific strategies to promote exercise.
Collapse
Affiliation(s)
- Margaret K Tanner
- Department of Psychology, University of Colorado Denver, PO Box 173364, North Classroom Rm. 5005 F, CB 173, Denver, CO, 80217-3364, USA
| | - Jazmyne K P Davis
- Department of Psychology, University of Colorado Denver, PO Box 173364, North Classroom Rm. 5005 F, CB 173, Denver, CO, 80217-3364, USA
| | - Jennifer Jaime
- Department of Psychology, University of Colorado Denver, PO Box 173364, North Classroom Rm. 5005 F, CB 173, Denver, CO, 80217-3364, USA
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Nicolette A Moya
- Department of Psychology, University of Colorado Denver, PO Box 173364, North Classroom Rm. 5005 F, CB 173, Denver, CO, 80217-3364, USA
- Department of Neuroscience, Northwestern Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Alyssa A Hohorst
- Department of Psychology, University of Colorado Denver, PO Box 173364, North Classroom Rm. 5005 F, CB 173, Denver, CO, 80217-3364, USA
| | - Kelsey Bonar
- Department of Psychology, University of Colorado Denver, PO Box 173364, North Classroom Rm. 5005 F, CB 173, Denver, CO, 80217-3364, USA
| | - Kelsey A Abrams
- Department of Psychology, University of Colorado Denver, PO Box 173364, North Classroom Rm. 5005 F, CB 173, Denver, CO, 80217-3364, USA
- University of Colorado Anschutz Medical Campus, Cell Biology, Stem Cells, and Development Graduate Program, Aurora, CO, USA
| | - Nashra Jamil
- Department of Psychology, University of Colorado Denver, PO Box 173364, North Classroom Rm. 5005 F, CB 173, Denver, CO, 80217-3364, USA
| | - Rebecca Han
- Department of Psychology, University of Colorado Denver, PO Box 173364, North Classroom Rm. 5005 F, CB 173, Denver, CO, 80217-3364, USA
| | - Troy J Hubert
- Department of Psychology, University of Colorado Denver, PO Box 173364, North Classroom Rm. 5005 F, CB 173, Denver, CO, 80217-3364, USA
- Department of Psychology, University of Kentucky, Lexington, KY, 40506, USA
| | - Nadja Brown
- Department of Psychology, University of Colorado Denver, PO Box 173364, North Classroom Rm. 5005 F, CB 173, Denver, CO, 80217-3364, USA
| | - Esteban C Loetz
- Department of Psychology, University of Colorado Denver, PO Box 173364, North Classroom Rm. 5005 F, CB 173, Denver, CO, 80217-3364, USA
| | - Benjamin N Greenwood
- Department of Psychology, University of Colorado Denver, PO Box 173364, North Classroom Rm. 5005 F, CB 173, Denver, CO, 80217-3364, USA.
| |
Collapse
|
19
|
Di Y, Diao Z, Zheng Q, Li J, Cheng Q, Li Z, Fang S, Wang H, Wei C, Zheng Q, Liu Y, Han J, Liu Z, Fan J, Ren W, Tian Y. Differential Alterations in Striatal Direct and Indirect Pathways Mediate Two Autism-like Behaviors in Valproate-Exposed Mice. J Neurosci 2022; 42:7833-7847. [PMID: 36414013 PMCID: PMC9581566 DOI: 10.1523/jneurosci.0623-22.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 08/20/2022] [Accepted: 08/24/2022] [Indexed: 12/14/2022] Open
Abstract
Autism is characterized by two key diagnostic criteria including social deficits and repetitive behaviors. Although recent studies implicated ventral striatum in social deficits and dorsal striatum in repetitive behaviors, here we revealed coexisting and opposite morphologic and functional alterations in the dorsostriatal direct and indirect pathways, and such alterations in these two pathways were found to be responsible, respectively, for the two abovementioned different autism-like behaviors exhibited by male mice prenatally exposed to valproate. The alteration in direct pathway was characterized by a potentiated state of basal activity, with impairment in transient responsiveness of D1-MSNs during social exploration. Concurrent alteration in indirect pathway was a depressed state of basal activity, with enhancement in transient responsiveness of D2-MSNs during repetitive behaviors. A causal relationship linking such differential alterations in these two pathways to the coexistence of these two autism-like behaviors was demonstrated by the cell type-specific correction of abnormal basal activity in the D1-MSNs and D2-MSNs of valproate-exposed mice. The findings support those differential alterations in two striatal pathways mediate the two coexisting autism-like behavioral abnormalities, respectively. This result will help in developing therapeutic options targeting these circuit alterations.SIGNIFICANCE STATEMENT Autism is characterized by two key diagnostic criteria including social deficits and repetitive behaviors. Although a number of recent studies have implicated ventral striatum in social deficits and dorsal striatum in repetitive behaviors, but social behaviors need to be processed by a series of actions, and repetitive behaviors, especially the high-order repetitive behaviors such as restrictive interests, have its scope to cognitive and emotional domains. The current study, for the first time, revealed that prenatal valproate exposure induced coexisting and differential alterations in the dorsomedial striatal direct and indirect pathways, and that these alterations mediate the two coexisting autism-like behavioral abnormalities, respectively. This result will help in developing therapeutic options targeting these circuit alterations to address the behavioral abnormalities.
Collapse
Affiliation(s)
- Yuanyuan Di
- Key Laboratory of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Zhijun Diao
- Key Laboratory of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Qi Zheng
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Jin Li
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Qiangqiang Cheng
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Zhongqi Li
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Suwen Fang
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Hao Wang
- Key Laboratory of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Chunling Wei
- Key Laboratory of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Qiaohua Zheng
- Key Laboratory of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Yingxun Liu
- Key Laboratory of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Jing Han
- Key Laboratory of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Zhiqiang Liu
- Key Laboratory of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Juan Fan
- Key Laboratory of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Wei Ren
- Key Laboratory of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
- Faculty of Education, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Yingfang Tian
- Key Laboratory of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| |
Collapse
|
20
|
Divergent encoding of active avoidance behavior in corticostriatal and corticolimbic projections. Sci Rep 2022; 12:10731. [PMID: 35750718 PMCID: PMC9232563 DOI: 10.1038/s41598-022-14930-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 05/17/2022] [Indexed: 11/09/2022] Open
Abstract
Active avoidance behavior, in which an animal performs an action to avoid a stressor, is crucial for survival and may provide insight into avoidance behaviors seen in anxiety disorders. Active avoidance requires the dorsomedial prefrontal cortex (dmPFC), which is thought to regulate avoidance via downstream projections to the striatum and amygdala. However, the endogenous activity of dmPFC projections during active avoidance learning has never been recorded. Here we utilized fiber photometry to record from the dmPFC and its axonal projections to the dorsomedial striatum (DMS) and the basolateral amygdala (BLA) during active avoidance learning in both male and female mice. We examined neural activity during conditioned stimulus (CS) presentations and during clinically relevant behaviors such as active avoidance or cued freezing. Both prefrontal projections showed learning-related increases in activity during CS onset throughout active avoidance training. The dmPFC as a whole showed increased and decreased patterns of activity during avoidance and cued freezing, respectively. Finally, dmPFC-DMS and dmPFC-BLA projections show divergent encoding of active avoidance behavior, with the dmPFC-DMS projection showing increased activity and the dmPFC-BLA projection showing decreased activity during active avoidance. Our results demonstrate task-relevant encoding of active avoidance in projection-specific dmPFC subpopulations that play distinct but complementary roles in active avoidance learning.
Collapse
|
21
|
Wu M, Di Y, Diao Z, Yan C, Cheng Q, Huang H, Liu Y, Wei C, Zheng Q, Fan J, Han J, Liu Z, Tian Y, Duan H, Ren W, Sun Z. Acute cannabinoids impair association learning via selectively enhancing synaptic transmission in striatonigral neurons. BMC Biol 2022; 20:108. [PMID: 35550070 PMCID: PMC9102575 DOI: 10.1186/s12915-022-01307-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 04/22/2022] [Indexed: 11/30/2022] Open
Abstract
Background Cannabinoids and their derivatives attract strong interest due to the tremendous potential of their psychoactive effects for treating psychiatric disorders and symptoms. However, their clinical application is restricted by various side-effects such as impaired coordination, anxiety, and learning and memory disability. Adverse impact on dorsal striatum-dependent learning is an important side-effect of cannabinoids. As one of the most important forms of learning mediated by the dorsal striatum, reinforcement learning is characterized by an initial association learning phase, followed by habit learning. While the effects of cannabinoids on habit learning have been well-studied, little is known about how cannabinoids influence the initial phase of reinforcement learning. Results We found that acute activation of cannabinoid receptor type 1 (CB1R) by the synthetic cannabinoid HU210 induced dose-dependent impairment of association learning, which could be alleviated by intra-dorsomedial striatum (DMS) injection of CB1R antagonist. Moreover, acute exposure to HU210 elicited enhanced synaptic transmission in striatonigral “direct” pathway medium spiny neurons (MSNs) but not indirect pathway neurons in DMS. Intriguingly, enhancement of synaptic transmission that is also observed after learning was abolished by HU210, indicating cannabinoid system might disrupt reinforcement learning by confounding synaptic plasticity normally required for learning. Remarkably, the impaired response-reinforcer learning was also induced by selectively enhancing the D1-MSN (MSN that selectively expresses the dopamine receptor type 1) activity by virally expressing excitatory hM3Dq DREADD (designer receptor exclusively activated by a designer drug), which could be rescued by specifically silencing the D1-MSN activity via hM4Di DREADD. Conclusion Our findings demonstrate dose-dependent deleterious effects of cannabinoids on association learning by disrupting plasticity change required for learning associated with the striatal direct pathway, which furthers our understanding of the side-effects of cannabinoids and the underlying mechanisms. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-022-01307-1.
Collapse
Affiliation(s)
- Meilin Wu
- MOE Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi'an, 710062, China
| | - Yuanyuan Di
- MOE Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi'an, 710062, China
| | - Zhijun Diao
- MOE Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi'an, 710062, China
| | - Chuanting Yan
- MOE Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi'an, 710062, China
| | - Qiangqiang Cheng
- College of Life Sciences, Shaanxi Normal University, Xi'an, 710062, China
| | - Huan Huang
- School of Psychology, Shaanxi Normal University, Xi'an, 710062, China
| | - Yingxun Liu
- MOE Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi'an, 710062, China
| | - Chunling Wei
- MOE Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi'an, 710062, China
| | - Qiaohua Zheng
- MOE Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi'an, 710062, China
| | - Juan Fan
- College of Life Sciences, Shaanxi Normal University, Xi'an, 710062, China
| | - Jing Han
- MOE Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi'an, 710062, China
| | - Zhiqiang Liu
- MOE Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi'an, 710062, China
| | - Yingfang Tian
- College of Life Sciences, Shaanxi Normal University, Xi'an, 710062, China
| | - Haijun Duan
- MOE Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi'an, 710062, China
| | - Wei Ren
- MOE Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi'an, 710062, China. .,School of Education, Shaanxi Normal University, Xi'an, 710062, China.
| | - Zongpeng Sun
- School of Psychology, Shaanxi Normal University, Xi'an, 710062, China.
| |
Collapse
|
22
|
Relationship of the Nigrostriatal Tract with the Motor Function and the Corticospinal Tract in Chronic Hemiparetic Stroke Patients: A Diffusion Tensor Imaging Study. Healthcare (Basel) 2022; 10:healthcare10040731. [PMID: 35455908 PMCID: PMC9028700 DOI: 10.3390/healthcare10040731] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/06/2022] [Accepted: 04/12/2022] [Indexed: 01/25/2023] Open
Abstract
This study investigated the relationship of the nigrostriatal tract (NST) with motor function and the corticospinal tract (CST) using diffusion tensor tractography in chronic hemiparetic stroke patients. Forty-three consecutive patients with putaminal hemorrhage in the chronic stage were recruited. The Motricity Index was used to evaluate the motor function of affected hemiparetic extremities. The fractional anisotropy and the tract volume of ipsilesional NST and ipsilesional CST were acquired. The tract volume (Rho = 0.824) of ipsilesional NST and fractional anisotropy (r = 0.682) and the tract volume (Rho = 0.886) of ipsilesional CST showed a strong positive correlation with the Motricity Index score. The fractional anisotropy of ipsilesional NST showed moderate positive correlations with the fractional anisotropy (r = 0.449) and tract volume (Rho = 0.353) of ipsilesional CST. The tract volume of ipsilesional NST showed strong positive correlations with the fractional anisotropy (Rho = 0.716) and the tract volume (Rho = 0.799) of ipsilesional CST. The regression model showed that the tract volumes of ipsilesional NST and ipsilesional CST were positively associated with the Motricity Index score (Adjusted R2 = 0.763, F = 45.998). Mediation analysis showed that the tract volume of ipsilesional CST partially mediated the effects of the tract volume of ipsilesional NST on the Motricity Index score (z = 3.34). A close relationship was found between ipsilesional NST and the motor function of affected extremities in chronic hemiparetic patients with putaminal hemorrhage. Moreover, ipsilesional NST influenced the motor function of affected extremities indirectly through ipsilesional CST.
Collapse
|
23
|
Ruiz-Tejada A, Neisewander J, Katsanos CS. Regulation of Voluntary Physical Activity Behavior: A Review of Evidence Involving Dopaminergic Pathways in the Brain. Brain Sci 2022; 12:brainsci12030333. [PMID: 35326289 PMCID: PMC8946175 DOI: 10.3390/brainsci12030333] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/23/2022] [Accepted: 02/26/2022] [Indexed: 02/04/2023] Open
Abstract
Physical activity leads to well-established health benefits. Current efforts to enhance physical activity have targeted mainly socioeconomic factors. However, despite these efforts, only a small number of adults engage in regular physical activity to the point of meeting current recommendations. Evidence collected in rodent models and humans establish a strong central nervous system component that regulates physical activity behavior. In particular, dopaminergic pathways in the central nervous system are among the best-characterized biological mechanisms to date with respect to regulating reward, motivation, and habit formation, which are critical for establishing regular physical activity. Herein, we discuss evidence for a role of brain dopamine in the regulation of voluntary physical activity behavior based on selective breeding and pharmacological studies in rodents, as well as genetic studies in both rodents and humans. While these studies establish a role of dopamine and associated mechanisms in the brain in the regulation of voluntary physical activity behavior, there is clearly need for more research on the underlying biology involved in motivation for physical activity and the formation of a physical activity habit. Such knowledge at the basic science level may ultimately be translated into better strategies to enhance physical activity levels within the society.
Collapse
|
24
|
Saito N, Itakura M, Sasaoka T. D1 Receptor Mediated Dopaminergic Neurotransmission Facilitates Remote Memory of Contextual Fear Conditioning. Front Behav Neurosci 2022; 16:751053. [PMID: 35309682 PMCID: PMC8925912 DOI: 10.3389/fnbeh.2022.751053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 01/21/2022] [Indexed: 11/13/2022] Open
Abstract
Dopaminergic neurotransmission via dopamine D1 receptors (D1Rs) is considered to play an important role not only in reward-based learning but also in aversive learning. The contextual and auditory cued fear conditioning tests involve the processing of classical fear conditioning and evaluates aversive learning memory. It is possible to evaluate aversive learning memory in two different types of neural transmission circuits. In addition, when evaluating the role of dopaminergic neurotransmission via D1R, to avoid the effects in D1R-mediated neural circuitry alterations during development, it is important to examine using mice who D1R expression in the mature stage is suppressed. Herein, we investigated the role of dopaminergic neurotransmission via D1Rs in aversive memory formation in contextual and auditory cued fear conditioning tests using D1R knockdown (KD) mice, in which the expression of D1Rs could be conditionally and reversibly controlled with doxycycline (Dox) treatment. For aversive memory, we examined memory formation using recent memory 1 day after conditioning, and remote memory 4 weeks after conditioning. Furthermore, immunostaining of the brain tissues of D1RKD mice was performed after aversive footshock stimulation to investigate the distribution of activated c-Fos, an immediate-early gene, in the hippocampus (CA1, CA3, dentate gyrus), striatum, amygdala, and prefrontal cortex during aversive memory formation. After aversive footshock stimulation, immunoblotting was performed using hippocampal, striatal, and amygdalar samples from D1RKD mice to investigate the increase in the amount of c-Fos and phosphorylated SNAP-25 at Ser187 residue. When D1R expression was suppressed using Dox, behavioral experiments revealed impaired contextual fear learning in remote aversion memory following footshock stimulation. Furthermore, expression analysis showed a slight increase in the post-stimulation amount of c-Fos in the hippocampus and striatum, and a significant increase in the amount of phosphorylated SNAP-25 in the hippocampus, striatum, and prefrontal cortex before and after stimulation. These findings indicate that deficiency in D1R-mediated dopaminergic neurotransmission is an important factor in impairing contextual fear memory formation for remote memory.
Collapse
Affiliation(s)
- Nae Saito
- Department of Comparative and Experimental Medicine, Brain Research Institute, Niigata University, Niigata, Japan
- Department of Molecular and Cellular Medicine, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Makoto Itakura
- Department of Biochemistry, Kitasato University School of Medicine, Sagamihara, Japan
| | - Toshikuni Sasaoka
- Department of Comparative and Experimental Medicine, Brain Research Institute, Niigata University, Niigata, Japan
- *Correspondence: Toshikuni Sasaoka,
| |
Collapse
|
25
|
Cell-type- and region-specific modulation of cocaine seeking by micro-RNA-1 in striatal projection neurons. Mol Psychiatry 2022; 27:918-928. [PMID: 34785784 PMCID: PMC9054679 DOI: 10.1038/s41380-021-01328-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 09/16/2021] [Accepted: 09/28/2021] [Indexed: 12/14/2022]
Abstract
The persistent and experience-dependent nature of drug addiction may result in part from epigenetic alterations, including non-coding micro-RNAs (miRNAs), which are both critical for neuronal function and modulated by cocaine in the striatum. Two major striatal cell populations, the striato-nigral and striato-pallidal projection neurons, express, respectively, the D1 (D1-SPNs) and D2 (D2-SPNs) dopamine receptor, and display distinct but complementary functions in drug-evoked responses. However, a cell-type-specific role for miRNAs action has yet to be clarified. Here, we evaluated the expression of a subset of miRNAs proposed to modulate cocaine effects in the nucleus accumbens (NAc) and dorsal striatum (DS) upon sustained cocaine exposure in mice and showed that these selected miRNAs were preferentially upregulated in the NAc. We focused on miR-1 considering the important role of some of its predicted mRNA targets, Fosb and Npas4, in the effects of cocaine. We validated these targets in vitro and in vivo. We explored the potential of miR-1 to regulate cocaine-induced behavior by overexpressing it in specific striatal cell populations. In DS D1-SPNs miR-1 overexpression downregulated Fosb and Npas4 and reduced cocaine-induced CPP reinstatement, but increased cue-induced cocaine seeking. In DS D2-SPNs miR-1 overexpression reduced the motivation to self-administer cocaine. Our results indicate a role of miR1 and its target genes, Fosb and Npas4, in these behaviors and highlight a precise cell-type- and region-specific modulatory role of miR-1, illustrating the importance of cell-specific investigations.
Collapse
|
26
|
Yan J, Xu W, Lenahan C, Huang L, Wen J, Li G, Hu X, Zheng W, Zhang JH, Tang J. CCR5 Activation Promotes NLRP1-Dependent Neuronal Pyroptosis via CCR5/PKA/CREB Pathway After Intracerebral Hemorrhage. Stroke 2021; 52:4021-4032. [PMID: 34719258 PMCID: PMC8607924 DOI: 10.1161/strokeaha.120.033285] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 07/11/2021] [Accepted: 07/30/2021] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND PURPOSE Neuronal pyroptosis is a type of regulated cell death triggered by proinflammatory signals. CCR5 (C-C chemokine receptor 5)-mediated inflammation is involved in the pathology of various neurological diseases. This study investigated the impact of CCR5 activation on neuronal pyroptosis and the underlying mechanism involving cAMP-dependent PKA (protein kinase A)/CREB (cAMP response element binding)/NLRP1 (nucleotide-binding domain leucine-rich repeat pyrin domain containing 1) pathway after experimental intracerebral hemorrhage (ICH). METHODS A total of 194 adult male CD1 mice were used. ICH was induced by autologous whole blood injection. Maraviroc (MVC)-a selective antagonist of CCR5-was administered intranasally 1 hour after ICH. To elucidate the underlying mechanism, a specific CREB inhibitor, 666-15, was administered intracerebroventricularly before MVC administration in ICH mice. In a set of naive mice, rCCL5 (recombinant chemokine ligand 5) and selective PKA activator, 8-Bromo-cAMP, were administered intracerebroventricularly. Short- and long-term neurobehavioral assessments, Western blot, Fluoro-Jade C, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL), and immunofluorescence staining were performed. RESULTS The brain expression of CCL5 (chemokine ligand 5), CCR5, PKA-Cα (protein kinase A-Cα), p-CREB (phospho-cAMP response element binding), and NLRP1 was increased, peaking at 24 hours after ICH. CCR5 was expressed on neurons, microglia, and astrocytes. MVC improved the short- and long-term neurobehavioral deficits and decreased neuronal pyroptosis in ipsilateral brain tissues at 24 hours after ICH, which were accompanied by increased PKA-Cα and p-CREB expression, and decreased expression of NLRP1, ASC (apoptosis-associated speck-like protein containing a CARD), C-caspase-1, GSDMD (gasdermin D), and IL (interleukin)-1β/IL-18. Such effects of MVC were abolished by 666-15. At 24 hours after injection in naive mice, rCCL5 induced neurological deficits, decreased PKA-Cα and p-CREB expression in the brain, and upregulated NLRP1, ASC, C-caspase-1, N-GSDMD, and IL-1β/IL-18 expression. Those effects of rCCL5 were reversed by 8-Bromo-cAMP. CONCLUSIONS CCR5 activation promoted neuronal pyroptosis and neurological deficits after ICH in mice, partially through the CCR5/PKA/CREB/NLRP1 signaling pathway. CCR5 inhibition with MVC may provide a promising therapeutic approach in managing patients with ICH.
Collapse
Affiliation(s)
- Jun Yan
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China (J.Y.)
| | - Weilin Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China (W.X.)
| | - Cameron Lenahan
- Department of Biomedical Sciences, Burrell College of Osteopathic Medicine, Las Cruces, NM (C.L.)
| | - Lei Huang
- Department of Neurosurgery (L.H., J.H.Z.), Loma Linda University, CA
- Department of Physiology and Pharmacology (L.H., J.H.Z., J.T.), Loma Linda University, CA
| | - Jing Wen
- Department of Rheumatism, First Affiliated Hospital of Guangxi Medical University, Nanning, China (J.W.)
| | - Gaigai Li
- Department of Neurology, Tongji Hospital, Tongji Medical college, Huazhong University of Science and Technology, Wuhan, China (G.L.)
| | - Xin Hu
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, China (X.H.)
| | - Wen Zheng
- Department of Neurology, The Third Xiangya Hospital of Central South University, Changsha, China (W.Z.)
| | - John H. Zhang
- Department of Neurosurgery (L.H., J.H.Z.), Loma Linda University, CA
- Department of Physiology and Pharmacology (L.H., J.H.Z., J.T.), Loma Linda University, CA
- Department of Anesthesiology (J.H.Z.), Loma Linda University, CA
| | - Jiping Tang
- Department of Physiology and Pharmacology (L.H., J.H.Z., J.T.), Loma Linda University, CA
| |
Collapse
|
27
|
Involvement of Midbrain Dopamine Neuron Activity in Negative Reinforcement Learning in Mice. Mol Neurobiol 2021; 58:5667-5681. [PMID: 34387814 DOI: 10.1007/s12035-021-02515-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 08/03/2021] [Indexed: 10/20/2022]
Abstract
The activity of the midbrain dopamine system reflects the valence of environmental events and modulates various brain structures to modify an organism's behavior. A series of recent studies reported that the direct and indirect pathways in the striatum are critical for instrumental learning, but the dynamic changes in dopamine neuron activity that occur during negative reinforcement learning are still largely unclear. In the present study, by using a negative reinforcement learning paradigm employing foot shocks as aversive stimuli, bidirectional changes in substantia nigra pars compacta (SNc) dopamine neuron activity in the learning and habituation phases were observed. The results showed that in the learning phase, before mice had mastered the skill of escaping foot shocks, the presence of foot shocks induced a transient reduction in the activity of SNc dopamine neurons; however, in the habituation phase, in which the learned skill was automated, it induced a transient increase. Microinjection of a dopamine D1 receptor (D1R) or D2 receptor (D2R) antagonist into the dorsomedial striatum (DMS) significantly impaired learning behavior, suggesting that the modulatory effects of dopamine on both the direct and indirect pathways are required. Moreover, during the learning phase, excitatory synaptic transmission to DMS D2R-expressing medium spiny neurons (D2-MSNs) was potentiated. However, upon completion of the learning and habituation phases, the synapses onto D1R-expressing medium spiny neurons (D1-MSNs) were potentiated, and those onto D2-MSNs were restored to normal levels. The bidirectional changes in both SNc dopamine neuron activity and DMS synaptic plasticity might be the critical neural correlates for negative reinforcement learning.
Collapse
|
28
|
Loewke AC, Minerva AR, Nelson AB, Kreitzer AC, Gunaydin LA. Frontostriatal Projections Regulate Innate Avoidance Behavior. J Neurosci 2021; 41:5487-5501. [PMID: 34001628 PMCID: PMC8221601 DOI: 10.1523/jneurosci.2581-20.2021] [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: 10/05/2020] [Revised: 04/13/2021] [Accepted: 04/16/2021] [Indexed: 11/21/2022] Open
Abstract
The dorsomedial prefrontal cortex (dmPFC) has been linked to avoidance and decision-making under conflict, key neural computations altered in anxiety disorders. However, the heterogeneity of prefrontal projections has obscured identification of specific top-down projections involved. While the dmPFC-amygdala circuit has long been implicated in controlling reflexive fear responses, recent work suggests that dmPFC-dorsomedial striatum (DMS) projections may be more important for regulating avoidance. Using fiber photometry recordings in both male and female mice during the elevated zero maze task, we show heightened neural activity in frontostriatal but not frontoamygdalar projection neurons during exploration of the anxiogenic open arms. Additionally, using optogenetics, we demonstrate that this frontostriatal projection preferentially excites postsynaptic D1 receptor-expressing neurons in the DMS and causally controls innate avoidance behavior. These results support a model for prefrontal control of defensive behavior in which the dmPFC-amygdala projection controls reflexive fear behavior and the dmPFC-striatum projection controls anxious avoidance behavior.SIGNIFICANCE STATEMENT The medial prefrontal cortex has been extensively linked to several behavioral symptom domains related to anxiety disorders, with much of the work centered around reflexive fear responses. Comparatively little is known at the mechanistic level about anxious avoidance behavior, a core feature across anxiety disorders. Recent work has suggested that the striatum may be an important hub for regulating avoidance behaviors. Our work uses optical circuit dissection techniques to identify a specific corticostriatal circuit involved in encoding and controlling avoidance behavior. Identifying neural circuits for avoidance will enable the development of more targeted symptom-specific treatments for anxiety disorders.
Collapse
Affiliation(s)
- Adrienne C Loewke
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, California 94158
| | - Adelaide R Minerva
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, California 94158
| | - Alexandra B Nelson
- Department of Neurology, University of California, San Francisco, San Francisco, California 94158
- Kavli Institute for Fundamental Neuroscience is at University of California, San Francisco, San Francisco, California 94158
| | - Anatol C Kreitzer
- Department of Neurology, University of California, San Francisco, San Francisco, California 94158
- Kavli Institute for Fundamental Neuroscience is at University of California, San Francisco, San Francisco, California 94158
- Department of Physiology, University of California, San Francisco, San Francisco, California 94158
- Neurological Disease Institute, Gladstone Institutes, San Francisco, California 94158
| | - Lisa A Gunaydin
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, California 94158
- Kavli Institute for Fundamental Neuroscience is at University of California, San Francisco, San Francisco, California 94158
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, California 94158
| |
Collapse
|
29
|
He Y, Li Y, Pu Z, Chen M, Gao Y, Chen L, Ruan Y, Pan X, Zhou Y, Ge Y, Zhou J, Zheng W, Huang Z, Li Z, Chen JF. Striatopallidal Pathway Distinctly Modulates Goal-Directed Valuation and Acquisition of Instrumental Behavior via Striatopallidal Output Projections. Cereb Cortex 2021; 30:1366-1381. [PMID: 31690946 DOI: 10.1093/cercor/bhz172] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 07/03/2019] [Accepted: 07/03/2019] [Indexed: 12/19/2022] Open
Abstract
The striatopallidal pathway is specialized for control of motor and motivational behaviors, but its causal role in striatal control of instrumental learning remains undefined (partly due to the confounding motor effects). Here, we leveraged the transient and "time-locked" optogenetic manipulations with the reward delivery to minimize motor confounding effect, to better define the striatopallidal control of instrumental behaviors. Optogenetic (Arch) silencing of the striatopallidal pathway in the dorsomedial striatum (DMS) and dorsolateral striatum (DLS) promoted goal-directed and habitual behaviors, respectively, without affecting acquisition of instrumental behaviors, indicating striatopallidal pathway suppression of instrumental behaviors under physiological condition. Conversely, striatopallidal pathway activation mainly affected the acquisition of instrumental behaviors with the acquisition suppression achieved by either optogenetic (ChR2) or chemicogenetic (hM3q) activation, by strong (10 mW, but not weak 1 mW) optogenetic activation, by the time-locked (but not random) optogenetic activation with the reward and by the DMS (but not DLS) striatopallidal pathway. Lastly, striatopallidal pathway modulated instrumental behaviors through striatopallidal output projections into the external globus pallidus (GPe) since optogenetic activation of the striatopallidal pathway in the DMS and of the striatopallidal output projections in the GPe similarly suppressed goal-directed behavior. Thus, the striatopallidal pathway confers distinctive and inhibitory controls of animal's sensitivity to goal-directed valuation and acquisition of instrumental behaviors under normal and over-activation conditions, through the output projections into GPe.
Collapse
Affiliation(s)
- Yan He
- School of Optometry and Ophthalmology and Eye Hospital, The Institute of Molecular Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Yan Li
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Zhilan Pu
- School of Optometry and Ophthalmology and Eye Hospital, The Institute of Molecular Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Mozi Chen
- School of Optometry and Ophthalmology and Eye Hospital, The Institute of Molecular Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Ying Gao
- School of Optometry and Ophthalmology and Eye Hospital, The Institute of Molecular Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Li Chen
- Department of Pharmacology, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Yang Ruan
- School of Optometry and Ophthalmology and Eye Hospital, The Institute of Molecular Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Xinran Pan
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Yuling Zhou
- School of Optometry and Ophthalmology and Eye Hospital, The Institute of Molecular Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Yuanyuan Ge
- School of Optometry and Ophthalmology and Eye Hospital, The Institute of Molecular Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Jianhong Zhou
- School of Optometry and Ophthalmology and Eye Hospital, The Institute of Molecular Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Wu Zheng
- School of Optometry and Ophthalmology and Eye Hospital, The Institute of Molecular Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Zhili Huang
- Department of Pharmacology, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Zhihui Li
- School of Optometry and Ophthalmology and Eye Hospital, The Institute of Molecular Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Jiang-Fan Chen
- School of Optometry and Ophthalmology and Eye Hospital, The Institute of Molecular Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| |
Collapse
|
30
|
Ramis MR, Sarubbo F, Moranta D, Tejada S, Lladó J, Miralles A, Esteban S. Neurochemical and Cognitive Beneficial Effects of Moderate Physical Activity and Catechin in Aged Rats. Antioxidants (Basel) 2021; 10:antiox10040621. [PMID: 33921628 PMCID: PMC8072822 DOI: 10.3390/antiox10040621] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/13/2021] [Accepted: 04/16/2021] [Indexed: 12/12/2022] Open
Abstract
A healthy aging process is a requirement for good life quality. A relationship between physical activity, the consumption of antioxidants and brain health has been stablished via the activation of pathways that reduce the harmful effects of oxidative stress, by inducing enzymes such as SIRT1, which is a protector of brain function. We analyzed the cognitive and neurochemical effects of applying physical exercise in elderly rats, alone or in combination with the antioxidant catechin. Several tests of spatial and episodic memory and motor coordination were evaluated. In addition, brain monoaminergic neurotransmitters and SIRT1 protein levels were assessed in the brains of the same rats. The results show that physical activity by itself improved age-related memory and learning deficits, correlating with the restoration of brain monoaminergic neurotransmitters and SIRT1 protein levels in the hippocampus. The administration of the antioxidant catechin along with the exercise program enhanced further the monoaminergic pathways, but not the other parameters studied. These results agree with previous reports revealing a neuroprotective effect of physical activity, probably based on its ability to improve the redox status of the brain, demonstrating that exercise at an advanced age, combined with the consumption of antioxidants, could produce favorable effects in terms of brain health.
Collapse
Affiliation(s)
- Margarita R. Ramis
- Laboratory of Neurophysiology, Biology Department, University of Balearic Islands (UIB), Ctra. Valldemossa Km 7.5, E-07122 Palma de Mallorca, Spain; (M.R.R.); (F.S.); (D.M.); (S.T.); (A.M.)
| | - Fiorella Sarubbo
- Laboratory of Neurophysiology, Biology Department, University of Balearic Islands (UIB), Ctra. Valldemossa Km 7.5, E-07122 Palma de Mallorca, Spain; (M.R.R.); (F.S.); (D.M.); (S.T.); (A.M.)
- Research Unit, University Hospital Son Llàtzer, Crta. Manacor Km 4, 07198 Palma, Spain
| | - David Moranta
- Laboratory of Neurophysiology, Biology Department, University of Balearic Islands (UIB), Ctra. Valldemossa Km 7.5, E-07122 Palma de Mallorca, Spain; (M.R.R.); (F.S.); (D.M.); (S.T.); (A.M.)
| | - Silvia Tejada
- Laboratory of Neurophysiology, Biology Department, University of Balearic Islands (UIB), Ctra. Valldemossa Km 7.5, E-07122 Palma de Mallorca, Spain; (M.R.R.); (F.S.); (D.M.); (S.T.); (A.M.)
- CIBERON (Physiopathology of Obesity and Nutrition), 28029 Madrid, Spain
- Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma, Spain
| | - Jerònia Lladó
- Department of Biology and University Institute of Health Sciences Research (IUNICS-IdISBa), University of Balearic Islands, 07122 Palma, Spain;
| | - Antoni Miralles
- Laboratory of Neurophysiology, Biology Department, University of Balearic Islands (UIB), Ctra. Valldemossa Km 7.5, E-07122 Palma de Mallorca, Spain; (M.R.R.); (F.S.); (D.M.); (S.T.); (A.M.)
- Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma, Spain
| | - Susana Esteban
- Laboratory of Neurophysiology, Biology Department, University of Balearic Islands (UIB), Ctra. Valldemossa Km 7.5, E-07122 Palma de Mallorca, Spain; (M.R.R.); (F.S.); (D.M.); (S.T.); (A.M.)
- Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma, Spain
- Correspondence: ; Tel.: +34-971-173-145
| |
Collapse
|
31
|
Turner BD, Smith NK, Manz KM, Chang BT, Delpire E, Grueter CA, Grueter BA. Cannabinoid type 1 receptors in A2a neurons contribute to cocaine-environment association. Psychopharmacology (Berl) 2021; 238:1121-1131. [PMID: 33454843 PMCID: PMC8386588 DOI: 10.1007/s00213-021-05759-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 01/06/2021] [Indexed: 12/14/2022]
Abstract
RATIONALE Cannabinoid type 1 receptors (CB1Rs) are widely expressed within the brain's reward circuits and are implicated in regulating drug induced behavioral adaptations. Understanding how CB1R signaling in discrete circuits and cell types contributes to drug-related behavior provides further insight into the pathology of substance use disorders. OBJECTIVE AND METHODS We sought to determine how cell type-specific expression of CB1Rs within striatal circuits contributes to cocaine-induced behavioral plasticity, hypothesizing that CB1R function in distinct striatal neuron populations would differentially impact behavioral outcomes. We crossed conditional Cnr1fl/fl mice and striatal output pathway cre lines (Drd1a -cre; D1, Adora2a -cre; A2a) to generate cell type-specific CB1R knockout mice and assessed their performance in cocaine locomotor and associative behavioral assays. RESULTS Both knockout lines retained typical locomotor activity at baseline. D1-Cre x Cnr1fl/fl mice did not display hyperlocomotion in response to acute cocaine dosing, and both knockout lines exhibited blunted locomotor activity across repeated cocaine doses. A2a-cre Cnr1fl/fl, mice did not express a preference for cocaine paired environments in a two-choice place preference task. CONCLUSIONS This study aids in mapping CB1R-dependent cocaine-induced behavioral adaptations onto distinct striatal neuron subtypes. A reduction of cocaine-induced locomotor activation in the D1- and A2a-Cnr1 knockout mice supports a role for CB1R function in the motor circuit. Furthermore, a lack of preference for cocaine-associated context in A2a-Cnr1 mice suggests that CB1Rs on A2a-neuron inhibitory terminals are necessary for either reward perception, memory consolidation, or recall. These results direct future investigations into CB1R-dependent adaptations underlying the development and persistence of substance use disorders.
Collapse
MESH Headings
- Animals
- Cocaine-Related Disorders/psychology
- Conditioning, Operant/drug effects
- Corpus Striatum/drug effects
- Environment
- Male
- Mice
- Mice, Knockout
- Motor Activity/drug effects
- Neurons/drug effects
- Receptor, Adenosine A2A/drug effects
- Receptor, Adenosine A2A/genetics
- Receptor, Cannabinoid, CB1/drug effects
- Receptor, Cannabinoid, CB1/genetics
- Receptor, Cannabinoid, CB1/metabolism
- Reward
Collapse
Affiliation(s)
- Brandon D Turner
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, 37232, USA
| | - Nicholas K Smith
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, 37232, USA
| | - Kevin M Manz
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, 37232, USA
- Medical Scientist Training Program, Vanderbilt University, Nashville, TN, 37232, USA
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Betty T Chang
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, 37232, USA
| | - Eric Delpire
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, 37232, USA
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Carrie A Grueter
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, 37232, USA
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Brad A Grueter
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, 37232, USA.
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA.
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA.
- Anesthesiology Research Division, Vanderbilt University School of Medicine, 2213 Garland Avenue, P435H MRB IV, Nashville, TN, 37232-0413, USA.
| |
Collapse
|
32
|
Dempsey LM, Kavanagh JJ. Muscarinic receptor blockade causes postcontraction enhancement in corticospinal excitability following maximal contractions. J Neurophysiol 2021; 125:1269-1278. [PMID: 33625939 DOI: 10.1152/jn.00673.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Although synaptic transmission in motor pathways can be regulated by neuromodulators, such as acetylcholine, few studies have examined how cholinergic activity affects cortical and spinal motor circuits following muscle contractions of varying intensities. This was a human, double-blinded, placebo-controlled, crossover study. Participants attended two sessions where they were administered either a placebo or 25 mg of promethazine. Electromyography of the abductor digiti minimi (ADM) was measured for all conditions. Motor evoked potentials (MEPs) were obtained via motor cortical transcranial magnetic stimulation (TMS), and F waves were obtained via ulnar nerve electrical stimulation. MEPs and F waves were examined: 1) when the muscle was at rest; 2) after the muscle had been active; and 3) after the muscle had been fatigued. MEPs were unaffected by muscarinic receptor blockade when measurements were recorded from resting muscle or following a 50% isometric maximal voluntary contraction (MVC). However, muscarinic receptor blockade increased MEP area following a 10-s MVC (P = 0.019) and following a fatiguing 60-s MVC (P = 0.040). F wave area and persistence were not affected by promethazine for any muscle contraction condition. Corticospinal excitability was influenced by cholinergic effects when voluntary drive to the muscle was high. Given that spinal motoneurone excitability remained unaffected, it is likely that cholinergic effects are influential within the motor cortex during strong muscle contractions. Future research should evaluate how cholinergic effects alter the relationship between subcortical structures and the motor cortex, as well as brainstem neuromodulatory pathways and spinal motoneurons.NEW & NOTEWORTHY The relationship between motor function and cholinergic circuitry in the central nervous system is complex. Although many studies have approached this issue at the cellular level, few studies have examined cholinergic mechanisms in humans performing muscle contractions. This study demonstrates that blockade of muscarinic acetylcholine receptors enhances motor evoked potentials (elicited with transcranial magnetic stimulation) following strong muscle contractions, but not weak muscle contractions.
Collapse
Affiliation(s)
- Lisa M Dempsey
- Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
| | - Justin J Kavanagh
- Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
| |
Collapse
|
33
|
Gαo is a major determinant of cAMP signaling in the pathophysiology of movement disorders. Cell Rep 2021; 34:108718. [PMID: 33535037 PMCID: PMC7903328 DOI: 10.1016/j.celrep.2021.108718] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 12/07/2020] [Accepted: 01/11/2021] [Indexed: 01/20/2023] Open
Abstract
The G protein alpha subunit o (Gαo) is one of the most abundant proteins in the nervous system, and pathogenic mutations in its gene (GNAO1) cause movement disorder. However, the function of Gαo is ill defined mechanistically. Here, we show that Gαo dictates neuromodulatory responsiveness of striatal neurons and is required for movement control. Using in vivo optical sensors and enzymatic assays, we determine that Gαo provides a separate transduction channel that modulates coupling of both inhibitory and stimulatory dopamine receptors to the cyclic AMP (cAMP)-generating enzyme adenylyl cyclase. Through a combination of cell-based assays and rodent models, we demonstrate that GNAO1-associated mutations alter Gαo function in a neuron-type-specific fashion via a combination of a dominant-negative and loss-of-function mechanisms. Overall, our findings suggest that Gαo and its pathological variants function in specific circuits to regulate neuromodulatory signals essential for executing motor programs. Muntean et al. describe biochemical, cellular, and physiological mechanisms by which the heterotrimeric G protein subunit Gαo controls neuromodulatory signaling in the striatum and elucidate mechanisms by which Gαo mutations compromise movements in GNAO1 disorder.
Collapse
|
34
|
Single-Cell Technologies in Parkinson׳s Disease. SYSTEMS MEDICINE 2021. [DOI: 10.1016/b978-0-12-801238-3.11613-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
35
|
Prager EM, Dorman DB, Hobel ZB, Malgady JM, Blackwell KT, Plotkin JL. Dopamine Oppositely Modulates State Transitions in Striosome and Matrix Direct Pathway Striatal Spiny Neurons. Neuron 2020; 108:1091-1102.e5. [PMID: 33080228 PMCID: PMC7769890 DOI: 10.1016/j.neuron.2020.09.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 07/29/2020] [Accepted: 09/21/2020] [Indexed: 12/18/2022]
Abstract
Corticostriatal synaptic integration is partitioned among striosome (patch) and matrix compartments of the dorsal striatum, allowing compartmentalized control of discrete aspects of behavior. Despite the significance of such organization, it's unclear how compartment-specific striatal output is dynamically achieved, particularly considering new evidence that overlap of afferents is substantial. We show that dopamine oppositely shapes responses to convergent excitatory inputs in mouse striosome and matrix striatal spiny projection neurons (SPNs). Activation of postsynaptic D1 dopamine receptors promoted the generation of long-lasting synaptically evoked "up-states" in matrix SPNs but opposed it in striosomes, which were more excitable under basal conditions. Differences in dopaminergic modulation were mediated, in part, by dendritic voltage-gated calcium channels (VGCCs): pharmacological manipulation of L-type VGCCs reversed compartment-specific responses to D1 receptor activation. These results support a novel mechanism for the selection of striatal circuit components, where fluctuating levels of dopamine shift the balance of compartment-specific striatal output.
Collapse
Affiliation(s)
- Eric M Prager
- Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University School of Medicine, Stony Brook, NY 11794, USA
| | - Daniel B Dorman
- Interdisciplinary Program in Neuroscience, George Mason University, Fairfax, VA 22030, USA
| | - Zachary B Hobel
- Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University School of Medicine, Stony Brook, NY 11794, USA
| | - Jeffrey M Malgady
- Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University School of Medicine, Stony Brook, NY 11794, USA
| | - Kim T Blackwell
- Interdisciplinary Program in Neuroscience, George Mason University, Fairfax, VA 22030, USA; Bioengineering Department, Volgenau School of Engineering, George Mason University, Fairfax, VA 22030, USA
| | - Joshua L Plotkin
- Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University School of Medicine, Stony Brook, NY 11794, USA.
| |
Collapse
|
36
|
Basal Ganglia Output Has a Permissive Non-Driving Role in a Signaled Locomotor Action Mediated by the Midbrain. J Neurosci 2020; 41:1529-1552. [PMID: 33328292 DOI: 10.1523/jneurosci.1067-20.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 11/25/2020] [Accepted: 12/02/2020] [Indexed: 01/11/2023] Open
Abstract
The basal ganglia are important for movement and reinforcement learning. Using mice of either sex, we found that the main basal ganglia GABAergic output in the midbrain, the substantia nigra pars reticulata (SNr), shows movement-related neural activity during the expression of a negatively reinforced signaled locomotor action known as signaled active avoidance; this action involves mice moving away during a warning signal to avoid a threat. In particular, many SNr neurons deactivate during active avoidance responses. However, whether SNr deactivation has an essential role driving or regulating active avoidance responses is unknown. We found that optogenetic excitation of SNr or striatal GABAergic fibers that project to an area in the pedunculopontine tegmentum (PPT) within the midbrain locomotor region abolishes signaled active avoidance responses, while optogenetic inhibition of SNr cells (mimicking the SNr deactivation observed during an active avoidance behavior) serves as an effective conditioned stimulus signal to drive avoidance responses by disinhibiting PPT neurons. However, preclusion of SNr deactivation, or direct inhibition of SNr fibers in the PPT, does not impair the expression of signaled active avoidance, indicating that SNr output does not drive the expression of a signaled locomotor action mediated by the midbrain. Consistent with a permissive regulatory role, SNr output provides information about the state of the ongoing action to downstream structures that mediate the action.SIGNIFICANCE STATEMENT During signaled active avoidance behavior, subjects move away to avoid a threat when directed by an innocuous sensory stimulus. Excitation of GABAergic cells in the substantia nigra pars reticulata (SNr), the main output of the basal ganglia, blocks signaled active avoidance, while inhibition of SNr cells is an effective stimulus to drive active avoidance. Interestingly, many SNr cells inhibit their firing during active avoidance responses, suggesting that SNr inhibition could be driving avoidance responses by disinhibiting downstream areas. However, interfering with the modulation of SNr cells does not impair the behavior. Thus, SNr may regulate the active avoidance movement in downstream areas that mediate the behavior, but does not drive it.
Collapse
|
37
|
Wang S, Deng Z, Wang J, Zhang W, Liu F, Xu J, Ma Y. Decreased GABAergic signaling, fewer parvalbumin-, somatostatin- and calretinin-positive neurons in brain of a rat model of simulated transport stress. Res Vet Sci 2020; 134:86-95. [PMID: 33360121 DOI: 10.1016/j.rvsc.2020.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 11/12/2020] [Accepted: 12/12/2020] [Indexed: 10/22/2022]
Abstract
Transport stress (TS) in animals lead to change in blood composition, brain structure, and the endocrine system as well as behavior. γ-aminobutyric acid (GABA), a major inhibitory neurotransmitter in the mammalian central nervous system (CNS), influences many physiological functions and plays a significant role in coping with stress. This study aimed to explore the effect of stress on behavior, HPA axis, GABA transmitters and the distribution of GABAergic interneurons in the prefrontal cortex (PFC) and striatum of the brain by a rat model of simulated transport stress (STS). Thirty-six male Sprague Dawley rats were randomly divided into a control group (n = 12, no stress), a TS1d group (n = 12, 2 h stress for 1 d) and a TS7d group (n = 12, 2 h stress each day for 7 d). After STS, the rats were subjected to open-field testing (OFT) followed by serologic analysis, colorimetry, Western blot and immunohistochemistry. The total score of the OFT showed the similar profile with serum concentrations of corticosterone (CORT) and norepinephrine (NE), which in the TS7d group were all higher than the TS1d group but lower than the control group. STS also reduced GABA, glutamate decarboxylase 67 (GAD67) and GABA transporter 1 (GAT1) expression in the TS1d and these markers were increased in the TS7d, suggesting that GABA was related to hypothalamic-pituitary-adrenal (HPA) axis activation under stress. The number of parvalbumin (PV)-, somatostatin (SOM)-, and calretinin (CR)- positive cells were decreased with stress increase. Our findings revealed that STS affected the behavior of rats, synthesis and release of GABA by altering the HPA axis.
Collapse
Affiliation(s)
- Shujing Wang
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Ziteng Deng
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Jia Wang
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Wenjun Zhang
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Fenghua Liu
- College of Animal Science and Technology, Beijing University of Agriculture, Beijing 102206, China
| | - Jianqin Xu
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Yunfei Ma
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
38
|
Andrews K, Josiah SS, Zhang J. The Therapeutic Potential of Neuronal K-Cl Co-Transporter KCC2 in Huntington's Disease and Its Comorbidities. Int J Mol Sci 2020; 21:E9142. [PMID: 33266310 PMCID: PMC7730145 DOI: 10.3390/ijms21239142] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/16/2020] [Accepted: 11/28/2020] [Indexed: 02/05/2023] Open
Abstract
Intracellular chloride levels in the brain are regulated primarily through the opposing effects of two cation-chloride co-transporters (CCCs), namely K+-Cl- co-transporter-2 (KCC2) and Na+-K+-Cl- co-transporter-1 (NKCC1). These CCCs are differentially expressed throughout the course of development, thereby determining the excitatory-to-inhibitory γ-aminobutyric acid (GABA) switch. GABAergic excitation (depolarisation) is important in controlling the healthy development of the nervous system; as the brain matures, GABAergic inhibition (hyperpolarisation) prevails. This developmental switch in excitability is important, as uncontrolled regulation of neuronal excitability can have implications for health. Huntington's disease (HD) is an example of a genetic disorder whereby the expression levels of KCC2 are abnormal due to mutant protein interactions. Although HD is primarily considered a motor disease, many other clinical manifestations exist; these often present in advance of any movement abnormalities. Cognitive change, in addition to sleep disorders, is prevalent in the HD population; the effect of uncontrolled KCC2 function on cognition and sleep has also been explored. Several mechanisms by which KCC2 expression is reduced have been proposed recently, thereby suggesting extensive investigation of KCC2 as a possible therapeutic target for the development of pharmacological compounds that can effectively treat HD co-morbidities. Hence, this review summarizes the role of KCC2 in the healthy and HD brain, and highlights recent advances that attest to KCC2 as a strong research and therapeutic target candidate.
Collapse
Affiliation(s)
| | | | - Jinwei Zhang
- Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, Hatherly Laboratories, Exeter EX4 4PS, UK; (K.A.); (S.S.J.)
| |
Collapse
|
39
|
Augustin SM, Loewinger GC, O'Neal TJ, Kravitz AV, Lovinger DM. Dopamine D2 receptor signaling on iMSNs is required for initiation and vigor of learned actions. Neuropsychopharmacology 2020; 45:2087-2097. [PMID: 32811899 PMCID: PMC7547091 DOI: 10.1038/s41386-020-00799-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/30/2020] [Accepted: 08/05/2020] [Indexed: 12/15/2022]
Abstract
Striatal dopamine D2 receptors (D2Rs) are important for motor output. Selective deletion of D2Rs from indirect pathway-projecting medium spiny neurons (iMSNs) impairs locomotor activities in a task-specific manner. However, the role of D2Rs in the initiation of motor actions in reward seeking and taking is not fully understood, and there is little information about how receptors contribute under different task demands and with different outcome types. The iMSN-D2Rs modulate neuronal activity and synaptic transmission, exerting control on circuit functions that may play distinct roles in action learning and performance. Selective deletion of D2Rs on iMSNs resulted in slower action initiation and response rate in an instrumental conditioning task, but only when performance demand was increased. The iMSN-Drd2KO mice were also slower to initiate swimming in a T-maze procedural learning task but were unimpaired in cognitive function and behavioral flexibility. In contrast, in a Pavlovian discrimination learning task, iMSN-Drd2KO mice exhibited normal acquisition and extinction of rewarded responding. The iMSN-Drd2KO mice showed performance deficits at all phases of rotarod skill learning. These findings reveal that dopamine modulation through iMSN-D2Rs influences the ability to self-initiate actions, as well as the willingness and/or vigor with which these responses are performed. However, these receptors seem to have little influence on simple associative learning or on stimulus-driven responding. The loss of normal D2R roles may contribute to disorders in which impaired dopamine signaling leads to hypokinesia or impaired initiation of specific voluntary actions.
Collapse
Affiliation(s)
- Shana M Augustin
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD, 20852, USA
| | - Gabriel C Loewinger
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD, 20852, USA
- Department of Biostatistics, Harvard TH Chan School of Public Health, Boston, MA, 02115, USA
| | - Timothy J O'Neal
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
- Graduate Program in Neuroscience and Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, Washington, 98195, USA
| | - Alexxai V Kravitz
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
- Departments of Psychiatry, Anesthesiology, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri, 63110, USA
| | - David M Lovinger
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD, 20852, USA.
| |
Collapse
|
40
|
Spatial organization of functional clusters representing reward and movement information in the striatal direct and indirect pathways. Proc Natl Acad Sci U S A 2020; 117:27004-27015. [PMID: 33055217 DOI: 10.1073/pnas.2010361117] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
To obtain insights into striatal neural processes underlying reward-based learning and movement control, we examined spatial organizations of striatal neurons related to movement and reward-based learning. For this, we recorded the activity of direct- and indirect-pathway neurons (D1 and A2a receptor-expressing neurons, respectively) in mice engaged in probabilistic classical conditioning and open-field free exploration. We found broadly organized functional clusters of striatal neurons in the direct as well as indirect pathways for both movement- and reward-related variables. Functional clusters for different variables were partially overlapping in both pathways, but the overlap between outcome- and value-related functional clusters was greater in the indirect than direct pathway. Also, value-related spatial clusters were progressively refined during classical conditioning. Our study shows the broad and learning-dependent spatial organization of functional clusters of dorsal striatal neurons in the direct and indirect pathways. These findings further argue against the classic model of the basal ganglia and support the importance of spatiotemporal patterns of striatal neuronal ensemble activity in the control of behavior.
Collapse
|
41
|
Paolone G. From the Gut to the Brain and Back: Therapeutic Approaches for the Treatment of Network Dysfunction in Parkinson's Disease. Front Neurol 2020; 11:557928. [PMID: 33117258 PMCID: PMC7575743 DOI: 10.3389/fneur.2020.557928] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 09/04/2020] [Indexed: 12/16/2022] Open
Abstract
Parkinson's disease (PD) is a complex, multisystem, progressive, degenerative disorder characterized by severe, debilitating motor dysfunction, cognitive impairments, and mood disorders. Although preclinical research has traditionally focused on the motor deficits resulting from the loss of nigrostriatal dopaminergic neurons, up to two thirds of PD patients present separate and distinct behavioral changes. Loss of basal forebrain cholinergic neurons occurs as early as the loss of dopaminergic cells and contributes to the cognitive decline in PD. In addition, attentional deficits can limit posture control and movement efficacy caused by dopaminergic cell loss. Complicating the picture further is intracellular α-synuclein accumulation beginning in the enteric nervous system and diffusing to the substantia nigra through the dorsal motor neurons of the vagus nerve. It seems that α-synuclein's role is that of mediating dopamine synthesis, storage, and release, and its function has not been completely understood. Treating a complex, multistage network disorder, such as PD, likely requires a multipronged approach. Here, we describe a few approaches that could be used alone or perhaps in combination to achieve a greater mosaic of behavioral benefit. These include (1) using encapsulated, genetically modified cells as delivery vehicles for administering neuroprotective trophic factors, such as GDNF, in a direct and sustained means to the brain; (2) immunotherapeutic interventions, such as vaccination or the use of monoclonal antibodies against aggregated, pathological α-synuclein; (3) the continuous infusion of levodopa-carbidopa through an intestinal gel pad to attenuate the loss of dopaminergic function and manage the motor and non-motor complications in PD patients; and (4) specific rehabilitation treatment programs for drug-refractory motor complications.
Collapse
Affiliation(s)
- Giovanna Paolone
- Department of Diagnostic and Public Health - Section of Pharmacology, University of Verona, Verona, Italy
| |
Collapse
|
42
|
Kumar SP, Babu PP. Aberrant Dopamine Receptor Signaling Plays Critical Role in the Impairment of Striatal Neurons in Experimental Cerebral Malaria. Mol Neurobiol 2020; 57:5069-5083. [PMID: 32833186 DOI: 10.1007/s12035-020-02076-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 08/14/2020] [Indexed: 01/19/2023]
Abstract
One-fourth survivors of cerebral malaria (CM) retain long-term cognitive and behavioral deficits. Structural abnormalities in striatum are reported in 80% of children with CM. Dopamine receptors (D1 and D2) are widely expressed in striatal medium spiny neurons (MSNs) that regulate critical physiological functions related to behavior and cognition. Dysregulation of dopamine receptors alters the expression of downstream proteins such as dopamine- and cAMP-regulated phosphoprotein (DARPP), Ca2+/calmodulin-dependent protein kinase II alpha (CaMKIIα), and p25/cyclin-dependent kinase 5 (cdk5). However, the role of dopamine receptor signaling dysfunction on the outcome of striatal neuron degeneration is unknown underlying the pathophysiology of CM. Using experimental CM (ECM), the present study attempted to understand the role of aberrant dopamine receptor signaling and its possible relation in causing MSNs morphological impairment. The effect of antimalarial drug artemether (ARM) rescue therapy was also assessed after ECM on the outcome of dopamine receptors downstream signaling. ECM was induced in C57BL/6 mice (male and female) infecting with Plasmodium berghei ANKA (PbA) parasite that reiterates the clinical setting of CM. We demonstrated that ECM caused a significant increase in the expression of D1, D2 receptors, phosphorylated DARPP, p25, cdk5, CaMKIIα, and D1-D2 heteromers. A substantial increase in neuronal damage observed in the dorsolateral striatum region of ECM brains (particularly in MSNs) as revealed by increased Fluoro-Jade C staining, reduced dendritic spine density, and impaired dendritic arborization with varicosities. While the ARM rescue therapy significantly altered the effects of ECM induced dopamine receptor signaling dysfunction and neurodegeneration. Overall, our data suggest that dysregulation of dopamine receptor signaling plays an important role in the degeneration of MSNs, and the ARM rescue therapy might provide better insights to develop effective therapeutic strategies for CM.
Collapse
Affiliation(s)
- Simhadri Praveen Kumar
- Neuroscience Laboratory (F-23/71), Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, 500046, India
| | - Phanithi Prakash Babu
- Neuroscience Laboratory (F-23/71), Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, 500046, India.
| |
Collapse
|
43
|
Farombi EO, Awogbindin IO, Owoeye O, Abah VO, Izomoh ER, Ezekiel IO. Kolaviron ameliorates behavioural deficit and injury to striatal dopaminergic terminals via modulation of oxidative burden, DJ-1 depletion and CD45R + cells infiltration in MPTP-model of Parkinson's disease. Metab Brain Dis 2020; 35:933-946. [PMID: 32430695 DOI: 10.1007/s11011-020-00578-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 04/30/2020] [Indexed: 10/24/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease. Currently, the precise pathogenic detail of PD is not entirely clear and first line therapeutics fail to attenuate the progress of the disease. In this study, we examined the neuroprotective effect of kolaviron, a natural antioxidant and anti-inflammatory biflavonoid from Garcinia kola seed, on behavioural impairment, neurodegeneration, oxidative stress and neuroinflammation in an acute MPTP-induced PD model. Kolaviron mitigated the frequently interrupted MPTP-associated hyperkinesia, inefficient gait, immobility, inability to pay attention to sizable holes on walking path, habitual clockwise rotations characterized with minimal diversion of movements and impaired balance. Also, kolaviron suppressed MPTP-mediated striatal oxidative stress, depletion as well as degeneration of dopaminergic terminals, reduced DJ-1 secretion and upregulated expression of caspase-3. Kolaviron facilitated cytoprotective antioxidant response and prevented MPTP-mediated neuroinflammation by blocking striatal infiltration of peripheral CD45R positive cells. Additionally, kolaviron reversed MPTP-induced inhibition of acetylcholinesterase activity. Together, our study provides evidence that the neuroprotective capacity of kolaviron to modulate striatal degeneration, behavioural impairment, antioxidant/redox imbalance and neuroinflammation implicated in the pathogenesis of PD may involve upregulation of DJ-1 secretion and inhibition of CD45R cells infiltration. Our data recommend kolaviron as a possible neuroprotective strategy in the management of Parkinson's disease and the associated behavioural complications, albeit the identity of MPTP-associated striatal CD45R infiltrate needs to be further characterized.
Collapse
Affiliation(s)
- Ebenezer O Farombi
- Drug Metabolism and Molecular Toxicology Research Laboratories, Department of Biochemistry, University of Ibadan, Ibadan, Nigeria.
| | - Ifeoluwa O Awogbindin
- Drug Metabolism and Molecular Toxicology Research Laboratories, Department of Biochemistry, University of Ibadan, Ibadan, Nigeria
| | - Olatunde Owoeye
- Department of Anatomy, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Victoria O Abah
- Drug Metabolism and Molecular Toxicology Research Laboratories, Department of Biochemistry, University of Ibadan, Ibadan, Nigeria
| | - Edirin R Izomoh
- Drug Metabolism and Molecular Toxicology Research Laboratories, Department of Biochemistry, University of Ibadan, Ibadan, Nigeria
| | - Ibukunoluwa O Ezekiel
- Drug Metabolism and Molecular Toxicology Research Laboratories, Department of Biochemistry, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
44
|
Wu M, Di Y, Diao Z, Yao L, Qian Z, Wei C, Zheng Q, Liu Y, Han J, Liu Z, Fan J, Tian Y, Ren W. Abnormal reinforcement learning in a mice model of autism induced by prenatal exposure to valproic acid. Behav Brain Res 2020; 395:112836. [PMID: 32745663 DOI: 10.1016/j.bbr.2020.112836] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 07/03/2020] [Accepted: 07/24/2020] [Indexed: 10/23/2022]
Abstract
Individuals with autism spectrum disorder (ASD) display dysfunction in learning from environmental stimulus that have positive or negative emotional values, posing obstacles to their everyday life. Unfortunately, mechanisms of the dysfunction are still unclear. Although early intervention for ASD victims based on reinforcement learning are commonly used, the mechanisms and characteristics of the improvement are also unknown. By using a mice model of ASD produced by prenatal exposure to valproic acid (VPA), the present work discovered a delayed response-reinforcer forming, and an impaired habit forming in a negative reinforcement learning paradigm in VPA exposure male offspring. But the extinction of the learned skills was found to become faster than normal male animals. Since escape action of nosepoking and the motility remain unchanged in the VPA male offspring, the impaired learning and the accelerated extinction are caused by deficits in higher brain functions underlying association between the animals' behavioral responses and the outcomes of such responses. The results further suggest that the rodent ASD model produced by prenatal exposure to VPA reproduces the deficits in reasoning or building the contingency between one's own behaviors and the consequent outcomes of the behavior seen in ASD patients.
Collapse
Affiliation(s)
- Meilin Wu
- MOE Key Laboratory of Modern Teaching Technology, Center for Teacher Professional Ability Development, Shaanxi Normal University, Xi'an, 710062, China
| | - Yuanyuan Di
- MOE Key Laboratory of Modern Teaching Technology, Center for Teacher Professional Ability Development, Shaanxi Normal University, Xi'an, 710062, China
| | - Zhijun Diao
- MOE Key Laboratory of Modern Teaching Technology, Center for Teacher Professional Ability Development, Shaanxi Normal University, Xi'an, 710062, China
| | - Li Yao
- MOE Key Laboratory of Modern Teaching Technology, Center for Teacher Professional Ability Development, Shaanxi Normal University, Xi'an, 710062, China
| | - Zhaoqiang Qian
- MOE Key Laboratory of Modern Teaching Technology, Center for Teacher Professional Ability Development, Shaanxi Normal University, Xi'an, 710062, China
| | - Chunling Wei
- MOE Key Laboratory of Modern Teaching Technology, Center for Teacher Professional Ability Development, Shaanxi Normal University, Xi'an, 710062, China
| | - Qiaohua Zheng
- MOE Key Laboratory of Modern Teaching Technology, Center for Teacher Professional Ability Development, Shaanxi Normal University, Xi'an, 710062, China
| | - Yihui Liu
- MOE Key Laboratory of Modern Teaching Technology, Center for Teacher Professional Ability Development, Shaanxi Normal University, Xi'an, 710062, China
| | - Jing Han
- MOE Key Laboratory of Modern Teaching Technology, Center for Teacher Professional Ability Development, Shaanxi Normal University, Xi'an, 710062, China
| | - Zhiqiang Liu
- MOE Key Laboratory of Modern Teaching Technology, Center for Teacher Professional Ability Development, Shaanxi Normal University, Xi'an, 710062, China
| | - Juan Fan
- College of Life Sciences, Shaanxi Normal University, Xi'an, 710062, China
| | - Yingfang Tian
- College of Life Sciences, Shaanxi Normal University, Xi'an, 710062, China.
| | - Wei Ren
- MOE Key Laboratory of Modern Teaching Technology, Center for Teacher Professional Ability Development, Shaanxi Normal University, Xi'an, 710062, China.
| |
Collapse
|
45
|
Claustral Neurons Projecting to Frontal Cortex Mediate Contextual Association of Reward. Curr Biol 2020; 30:3522-3532.e6. [PMID: 32707061 DOI: 10.1016/j.cub.2020.06.064] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 06/01/2020] [Accepted: 06/19/2020] [Indexed: 12/14/2022]
Abstract
The claustrum is a small nucleus, exhibiting vast reciprocal connectivity with cortical, subcortical, and midbrain regions. Recent studies, including ours, implicate the claustrum in salience detection and attention. In the current study, we develop an iterative functional investigation of the claustrum, guided by quantitative spatial transcriptional analysis. Using this approach, we identify a circuit involving dopamine-receptor expressing claustral neurons projecting to frontal cortex necessary for context association of reward. We describe the recruitment of claustral neurons by cocaine and their role in drug sensitization. In order to characterize the circuit within which these neurons are embedded, we apply chemo- and opto-genetic manipulation of increasingly specified claustral subpopulations. This strategy resolves the role of a defined network of claustrum neurons expressing dopamine D1 receptors and projecting to frontal cortex in the acquisition of cocaine conditioned-place preference and real-time optogenetic conditioned-place preference. In sum, our results suggest a role for a claustrum-to-frontal cortex circuit in the attribution of incentive salience, allocating attention to reward-related contextual cues.
Collapse
|
46
|
Porcu A, Vaughan M, Nilsson A, Arimoto N, Lamia K, Welsh DK. Vulnerability to helpless behavior is regulated by the circadian clock component CRYPTOCHROME in the mouse nucleus accumbens. Proc Natl Acad Sci U S A 2020; 117:13771-13782. [PMID: 32487727 PMCID: PMC7306774 DOI: 10.1073/pnas.2000258117] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The nucleus accumbens (NAc), a central component of the midbrain dopamine reward circuit, exhibits disturbed circadian rhythms in the postmortem brains of depressed patients. We hypothesized that normal mood regulation requires proper circadian timing in the NAc, and that mood disorders are associated with dysfunctions of the NAc cellular circadian clock. In mice exhibiting stress-induced depression-like behavior (helplessness), we found altered circadian clock function and high nighttime expression of the core circadian clock component CRYPTOCHROME (CRY) in the NAc. In the NAc of helpless mice, we found that higher expression of CRY is associated with decreased activation of dopamine 1 receptor-expressing medium spiny neurons (D1R-MSNs). Furthermore, D1R-MSN-specific CRY-knockdown in the NAc reduced susceptibility to stress-induced helplessness and increased NAc neuronal activation at night. Finally, we show that CRY inhibits D1R-induced G protein activation, likely by interacting with the Gs protein. Altered circadian rhythms and CRY expression were also observed in human fibroblasts from major depressive disorder patients. Our data reveal a causal role for CRY in regulating the midbrain dopamine reward system, and provide a mechanistic link between the NAc circadian clock and vulnerability to depression.
Collapse
Affiliation(s)
- Alessandra Porcu
- Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA 92161;
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92037
- Center for Circadian Biology, University of California San Diego, La Jolla, CA 92037
| | - Megan Vaughan
- Department of Molecular Medicine, Scripps Research, La Jolla, CA 92037
| | - Anna Nilsson
- Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA 92161
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92037
- Center for Circadian Biology, University of California San Diego, La Jolla, CA 92037
| | - Natsuko Arimoto
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92037
- Center for Circadian Biology, University of California San Diego, La Jolla, CA 92037
| | - Katja Lamia
- Department of Molecular Medicine, Scripps Research, La Jolla, CA 92037
| | - David K Welsh
- Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA 92161
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92037
- Center for Circadian Biology, University of California San Diego, La Jolla, CA 92037
| |
Collapse
|
47
|
Moshitzky G, Shoham S, Madrer N, Husain AM, Greenberg DS, Yirmiya R, Ben-Shaul Y, Soreq H. Cholinergic Stress Signals Accompany MicroRNA-Associated Stereotypic Behavior and Glutamatergic Neuromodulation in the Prefrontal Cortex. Biomolecules 2020; 10:E848. [PMID: 32503154 PMCID: PMC7355890 DOI: 10.3390/biom10060848] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 05/24/2020] [Accepted: 05/28/2020] [Indexed: 12/13/2022] Open
Abstract
Stereotypic behavior (SB) is common in emotional stress-involved psychiatric disorders and is often attributed to glutamatergic impairments, but the underlying molecular mechanisms are unknown. Given the neuro-modulatory role of acetylcholine, we sought behavioral-transcriptomic links in SB using TgR transgenic mice with impaired cholinergic transmission due to over-expression of the stress-inducible soluble 'readthrough' acetylcholinesterase-R splice variant AChE-R. TgR mice showed impaired organization of behavior, performance errors in a serial maze test, escape-like locomotion, intensified reaction to pilocarpine and reduced rearing in unfamiliar situations. Small-RNA sequencing revealed 36 differentially expressed (DE) microRNAs in TgR mice hippocampi, 8 of which target more than 5 cholinergic transcripts. Moreover, compared to FVB/N mice, TgR prefrontal cortices displayed individually variable changes in over 400 DE mRNA transcripts, primarily acetylcholine and glutamate-related. Furthermore, TgR brains presented c-fos over-expression in motor behavior-regulating brain regions and immune-labeled AChE-R excess in the basal ganglia, limbic brain nuclei and the brain stem, indicating a link with the observed behavioral phenotypes. Our findings demonstrate association of stress-induced SB to previously unknown microRNA-mediated perturbations of cholinergic/glutamatergic networks and underscore new therapeutic strategies for correcting stereotypic behaviors.
Collapse
Affiliation(s)
- Gilli Moshitzky
- The Institute of Life Sciences and The Edmond and Lily Safra Center of Brain Science, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel; (G.M.); (N.M.); (A.M.H.); (D.S.G.)
| | - Shai Shoham
- Herzog Medical Center, Givat Shaul, P.O. Box 3900, Jerusalem 9103702, Israel;
| | - Nimrod Madrer
- The Institute of Life Sciences and The Edmond and Lily Safra Center of Brain Science, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel; (G.M.); (N.M.); (A.M.H.); (D.S.G.)
| | - Amir Mouhammed Husain
- The Institute of Life Sciences and The Edmond and Lily Safra Center of Brain Science, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel; (G.M.); (N.M.); (A.M.H.); (D.S.G.)
| | - David S. Greenberg
- The Institute of Life Sciences and The Edmond and Lily Safra Center of Brain Science, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel; (G.M.); (N.M.); (A.M.H.); (D.S.G.)
| | - Raz Yirmiya
- Department of Psychology, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel;
| | - Yoram Ben-Shaul
- Department of Medical Neurobiology, The Institute of Medical Research Israel-Canada, Jerusalem 9112102, Israel;
| | - Hermona Soreq
- The Institute of Life Sciences and The Edmond and Lily Safra Center of Brain Science, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel; (G.M.); (N.M.); (A.M.H.); (D.S.G.)
| |
Collapse
|
48
|
Moses TE, Burmeister M, Greenwald MK. Heroin delay discounting and impulsivity: Modulation by DRD1 genetic variation. Addict Biol 2020; 25:e12777. [PMID: 31192519 DOI: 10.1111/adb.12777] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 04/11/2019] [Accepted: 04/27/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Dopamine D1 receptors (encoded by DRD1) are implicated in drug addiction and high-risk behaviors. Delay discounting (DD) procedures measure decisional balance between choosing smaller/sooner rewards vs larger/later rewards. Individuals with higher DD (rapid discounting) are prone to maladaptive behaviors that provide immediate reinforcement (eg, substance use). DRD1 variants have been linked with increased DD (in healthy volunteers) and opioid abuse. This study determined whether four dopaminergic functional variants modulated heroin DD and impulsivity. METHODS Substance use, DD, and genotype data (DRD1 rs686 and rs5326, DRD3 rs6280, COMT rs4680) were obtained from 106 current heroin users. Subjects completed an array of DD choices during two imagined conditions: heroin satiation and withdrawal. Rewards were expressed as $10 heroin bag units, with maximum delayed amount of 30 bags. Delays progressively increased from 3 to 96 hours. RESULTS DRD1 rs686 (A/A, n = 25; G/A, n = 56; G/G, n = 25) was linearly related to the difference in heroin DD (area under the curve; AUC) between the heroin satiation and withdrawal conditions; specifically, G/G homozygotes had a significantly smaller (satiation minus withdrawal) AUC difference score had higher drug-use impulsivity questionnaire scores, relative to A/A homozygotes, with G/A intermediate. DRD3 and COMT variants were not associated with these DD and impulsivity outcomes. CONCLUSION DRD1 rs686 modulated the difference in heroin DD score between pharmacological states and was associated with drug-use impulsivity. These data support a role of DRD1 in opioid DD and impulsive behaviors.
Collapse
|
49
|
Sheynin J, Baetu I, Collins-Praino LE, Myers CE, Winwood-Smith R, Moustafa AA. Maladaptive avoidance patterns in Parkinson's disease are exacerbated by symptoms of depression. Behav Brain Res 2020; 382:112473. [PMID: 31935419 DOI: 10.1016/j.bbr.2020.112473] [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: 10/22/2019] [Revised: 01/05/2020] [Accepted: 01/05/2020] [Indexed: 11/26/2022]
Abstract
Parkinson's disease (PD) is a chronic, progressive neurodegenerative disorder, characterized by a loss of dopaminergic neurons in the substantia nigra pars compacta. Given that dopamine is critically involved in learning and other cognitive processes, such as working memory, dopamine loss in PD has been linked both to learning abnormalities and to cognitive dysfunction more generally in the disease. It is unclear, however, whether avoidance behavior is impacted in PD. This is significant, as this type of instrumental behavior plays an important role in both decision-making and emotional (dys) function. Consequently, the aim of the present study was to examine avoidance learning and operant extinction in PD using a computer-based task. On this task, participants control a spaceship and attempt to shoot an enemy spaceship to gain points. They also learn to hide in safe areas to protect from (i.e., avoid) aversive events (on-screen explosions and point loss). The results showed that patients with PD (N = 25) acquired an avoidance response during aversive periods to the same extent as healthy age-matched controls (N = 19); however, patients demonstrated greater hiding during safe periods not associated with aversive events, which could represent maladaptive generalization of the avoidance response. Furthermore, this impairment was more pronounced during the extinction phase, and in patients who reported higher levels of depression. These results demonstrate for the first time that PD is associated with maladaptive avoidance patterns, which could possibly contribute to the emergence of depression in the disease.
Collapse
Affiliation(s)
- Jony Sheynin
- Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI, USA; Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA.
| | - Irina Baetu
- School of Psychology, University of Adelaide, Adelaide, SA, Australia
| | - Lyndsey E Collins-Praino
- Department of Medical Sciences, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Catherine E Myers
- Department of Veterans Affairs, New Jersey Health Care System, East Orange, NJ, USA; Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Robyn Winwood-Smith
- School of Social Sciences and Psychology, Western Sydney University, Sydney, NSW, Australia
| | - Ahmed A Moustafa
- School of Social Sciences and Psychology, Western Sydney University, Sydney, NSW, Australia; The MARCS Institute for Brain, Behaviour and Development, Western Sydney University, Sydney, NSW, Australia
| |
Collapse
|
50
|
LeBlanc KH, London TD, Szczot I, Bocarsly ME, Friend DM, Nguyen KP, Mengesha MM, Rubinstein M, Alvarez VA, Kravitz AV. Striatopallidal neurons control avoidance behavior in exploratory tasks. Mol Psychiatry 2020; 25:491-505. [PMID: 29695836 PMCID: PMC6202282 DOI: 10.1038/s41380-018-0051-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 02/05/2018] [Accepted: 02/19/2018] [Indexed: 12/22/2022]
Abstract
The dorsal striatum has been linked to decision-making under conflict, but the mechanism by which striatal neurons contribute to approach-avoidance conflicts remains unclear. We hypothesized that striatopallidal dopamine D2 receptor (D2R)-expressing neurons promote avoidance, and tested this hypothesis in two exploratory approach-avoidance conflict paradigms in mice: the elevated zero maze and open field. Genetic elimination of D2Rs on striatopallidal neurons (iMSNs), but not other neural populations, increased avoidance of the open areas in both tasks, in a manner that was dissociable from global changes in movement. Population calcium activity of dorsomedial iMSNs was disrupted in mice lacking D2Rs on iMSNs, suggesting that disrupted output of iMSNs contributes to heightened avoidance behavior. Consistently, artificial disruption of iMSN output with optogenetic stimulation heightened avoidance of open areas of these tasks, while inhibition of iMSN output reduced avoidance. We conclude that dorsomedial striatal iMSNs control approach-avoidance conflicts in exploratory tasks, and highlight this neural population as a potential target for reducing avoidance in anxiety disorders.
Collapse
Affiliation(s)
- Kimberly H. LeBlanc
- grid.94365.3d0000 0001 2297 5165National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892 USA
| | - Tanisha D. London
- grid.94365.3d0000 0001 2297 5165National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892 USA
| | - Ilona Szczot
- grid.94365.3d0000 0001 2297 5165National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892 USA
| | - Miriam E. Bocarsly
- grid.94365.3d0000 0001 2297 5165National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892 USA
| | - Danielle M. Friend
- grid.94365.3d0000 0001 2297 5165National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892 USA
| | - Katrina P. Nguyen
- grid.94365.3d0000 0001 2297 5165National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892 USA
| | - Marda M. Mengesha
- grid.94365.3d0000 0001 2297 5165National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892 USA
| | - Marcelo Rubinstein
- grid.423606.50000 0001 1945 2152Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, CONICET, Buenos Aires, C1428ADN Argentina ,grid.7345.50000 0001 0056 1981FCEN, Universidad de Buenos Aires, Buenos Aires, C1428EGA Argentina ,grid.214458.e0000000086837370Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109 USA
| | - Veronica A. Alvarez
- grid.94365.3d0000 0001 2297 5165National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892 USA
| | - Alexxai V. Kravitz
- grid.94365.3d0000 0001 2297 5165National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892 USA ,grid.94365.3d0000 0001 2297 5165National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD 20892 USA
| |
Collapse
|