1
|
Zhang Z, Bao C, Li Z, He C, Jin W, Li C, Chen Y. Integrated omics analysis reveals the alteration of gut microbiota and fecal metabolites in Cervus elaphus kansuensis. Appl Microbiol Biotechnol 2024; 108:125. [PMID: 38229330 DOI: 10.1007/s00253-023-12841-5] [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: 04/13/2023] [Revised: 10/05/2023] [Accepted: 10/17/2023] [Indexed: 01/18/2024]
Abstract
The gut microbiota is the largest and most complex microecosystem in animals. It is influenced by the host's dietary habits and living environment, and its composition and diversity play irreplaceable roles in animal nutrient metabolism, immunity, and adaptation to the environment. Although the gut microbiota of red deer has been studied, the composition and function of the gut microbiota in Gansu red deer (Cervus elaphus kansuensis), an endemic subspecies of red deer in China, has not been reported. In this study, the composition and diversity of the gut microbiome and fecal metabolomics of C. elaphus kansuensis were identified and compared for the first time by using 16S rDNA sequencing, metagenomic sequencing, and LC-MS/MS. There were significant differences in gut microbiota structure and diversity between wild and farmed C. elaphus kansuensis. The 16S rDNA sequencing results showed that the genus UCRD-005 was dominant in both captive red deer (CRD) and wild red deer (WRD). Metagenomic sequencing showed similar results to those of 16S rDNA sequencing for gut microbiota in CRD and WRD at the phylum and genus levels. 16S rDNA and metagenomics sequencing data suggested that Bacteroides and Bacillus might serve as marker genera for CRD and WRD, respectively. Fecal metabolomics results showed that 520 metabolites with significant differences were detected between CRD and WRD and most differential metabolites were involved in lipid metabolism. The results suggested that large differences in gut microbiota composition and fecal metabolites between CRD and WRD, indicating that different dietary habits and living environments over time have led to the development of stable gut microbiome characteristics for CRD and WRD to meet their respective survival and reproduction needs. KEY POINTS: • Environment and food affected the gut microbiota and fecal metabolites in red deer • Genera Bacteroides and Bacillus may play important roles in CRD and WRD, respectively • Flavonoids and ascorbic acid in fecal metabolites may influence health of red deer.
Collapse
Affiliation(s)
- Zhenxiang Zhang
- College of Eco-Environmental Engineering, Qinghai University, No. 251 Ningda Road, Xining, 810016, China
- Qinghai Provincial Key Laboratory of Adaptive Management on Alpine Grassland, Academy of Animal Science and Veterinary Medicine, Qinghai University, Xining, China
| | - Changhong Bao
- College of Eco-Environmental Engineering, Qinghai University, No. 251 Ningda Road, Xining, 810016, China
| | - Zhaonan Li
- College of Eco-Environmental Engineering, Qinghai University, No. 251 Ningda Road, Xining, 810016, China
| | - Caixia He
- College of Eco-Environmental Engineering, Qinghai University, No. 251 Ningda Road, Xining, 810016, China
| | - Wenjie Jin
- College of Eco-Environmental Engineering, Qinghai University, No. 251 Ningda Road, Xining, 810016, China
| | - Changzhong Li
- College of Eco-Environmental Engineering, Qinghai University, No. 251 Ningda Road, Xining, 810016, China.
| | - Yanxia Chen
- College of Eco-Environmental Engineering, Qinghai University, No. 251 Ningda Road, Xining, 810016, China.
| |
Collapse
|
2
|
Jia Q, Tan H, Li T, Duan X. Role of adenosine in the pathophysiology and treatment of attention deficit hyperactivity disorder. Purinergic Signal 2024:10.1007/s11302-024-10059-2. [PMID: 39480600 DOI: 10.1007/s11302-024-10059-2] [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: 06/26/2024] [Accepted: 10/21/2024] [Indexed: 11/02/2024] Open
Abstract
Attention deficit hyperactivity disorder (ADHD) is a complex neurodevelopmental condition characterized by persistent inattention, hyperactivity, and impulsivity. Although its precise etiology remains unclear, current evidence suggests that dysregulation within the neurotransmitter system plays a key role in the pathogenesis of ADHD. Adenosine, an endogenous nucleoside widely distributed throughout the body, modulates various physiological processes, including neurotransmitter release, sleep regulation, and cognitive functions through its receptors. This review critically examines the role of the adenosine system in ADHD, focusing on the links between adenosine receptor function and ADHD-related symptoms. Additionally, it explores how adenosine interacts with dopamine and other neurotransmitter pathways, shedding light on its involvement in ADHD pathophysiology. This review aims to provide insights into the potential therapeutic implications of targeting the adenosine system for ADHD management.
Collapse
Affiliation(s)
- Qingxia Jia
- Department of Rehabilitation Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing, 400014, China
| | - Hongwan Tan
- People's Hospital of Tongliang District, Chongqing, 402560, Tongliang, China
| | - Tingsong Li
- Department of Rehabilitation Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing, 400014, China
| | - Xiaoling Duan
- Department of Rehabilitation Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing, 400014, China.
| |
Collapse
|
3
|
Mah A, Golden CEM, Constantinople CM. Dopamine transients encode reward prediction errors independent of learning rates. Cell Rep 2024; 43:114840. [PMID: 39395170 DOI: 10.1016/j.celrep.2024.114840] [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: 04/15/2024] [Revised: 08/19/2024] [Accepted: 09/20/2024] [Indexed: 10/14/2024] Open
Abstract
Biological accounts of reinforcement learning posit that dopamine encodes reward prediction errors (RPEs), which are multiplied by a learning rate to update state or action values. These values are thought to be represented by corticostriatal synaptic weights, which are updated by dopamine-dependent plasticity. This suggests that dopamine release reflects the product of the learning rate and RPE. Here, we characterize dopamine encoding of learning rates in the nucleus accumbens core (NAcc) in a volatile environment. Using a task with semi-observable states offering different rewards, we find that rats adjust how quickly they initiate trials across states using RPEs. Computational modeling and behavioral analyses show that learning rates are higher following state transitions and scale with trial-by-trial changes in beliefs about hidden states, approximating normative Bayesian strategies. Notably, dopamine release in the NAcc encodes RPEs independent of learning rates, suggesting that dopamine-independent mechanisms instantiate dynamic learning rates.
Collapse
Affiliation(s)
- Andrew Mah
- Center for Neural Science, New York University, New York, NY, USA
| | - Carla E M Golden
- Center for Neural Science, New York University, New York, NY, USA
| | | |
Collapse
|
4
|
Cai X, Liu C, Tsutsui-Kimura I, Lee JH, Guo C, Banerjee A, Lee J, Amo R, Xie Y, Patriarchi T, Li Y, Watabe-Uchida M, Uchida N, Kaeser PS. Dopamine dynamics are dispensable for movement but promote reward responses. Nature 2024:10.1038/s41586-024-08038-z. [PMID: 39415006 DOI: 10.1038/s41586-024-08038-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/11/2024] [Indexed: 10/18/2024]
Abstract
Dopamine signalling modes differ in kinetics and spatial patterns of receptor activation1,2. How these modes contribute to motor function, motivation and learning has long been debated3-21. Here we show that action-potential-induced dopamine release is dispensable for movement initiation but supports reward-oriented behaviour. We generated mice with dopamine-neuron-specific knockout of the release site organizer protein RIM to disrupt action-potential-induced dopamine release. In these mice, rapid in vivo dopamine dynamics were strongly impaired, but baseline dopamine persisted and fully supported spontaneous movement. Conversely, reserpine-mediated dopamine depletion or blockade of dopamine receptors disrupted movement initiation. The dopamine precursor L-DOPA reversed reserpine-induced bradykinesia without restoring fast dopamine dynamics, a result that substantiated the conclusion that these dynamics are dispensable for movement initiation. In contrast to spontaneous movement, reward-oriented behaviour was impaired in dopamine-neuron-specific RIM knockout mice. In conditioned place preference and two-odour discrimination tasks, the mice effectively learned to distinguish the cues, which indicates that reward-based learning persists after RIM ablation. However, the performance vigour was reduced. During probabilistic cue-reward association, dopamine dynamics and conditioned responses assessed through anticipatory licking were disrupted. These results demonstrate that action-potential-induced dopamine release is dispensable for motor function and subsecond precision of movement initiation but promotes motivation and performance during reward-guided behaviours.
Collapse
Affiliation(s)
- Xintong Cai
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Changliang Liu
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Iku Tsutsui-Kimura
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Joon-Hyuk Lee
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Chong Guo
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Aditi Banerjee
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Jinoh Lee
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Ryunosuke Amo
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Yudi Xie
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Tommaso Patriarchi
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, ETH and University of Zurich, Zurich, Switzerland
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
| | - Mitsuko Watabe-Uchida
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Naoshige Uchida
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
5
|
Pan-Vazquez A, Sanchez Araujo Y, McMannon B, Louka M, Bandi A, Haetzel L, Faulkner M, Pillow JW, Daw ND, Witten IB. Pre-existing visual responses in a projection-defined dopamine population explain individual learning trajectories. Curr Biol 2024:S0960-9822(24)01286-7. [PMID: 39413788 DOI: 10.1016/j.cub.2024.09.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 06/11/2024] [Accepted: 09/17/2024] [Indexed: 10/18/2024]
Abstract
A key challenge of learning a new task is that the environment is high dimensional-there are many different sensory features and possible actions, with typically only a small reward-relevant subset. Although animals can learn to perform complex tasks that involve arbitrary associations between stimuli, actions, and rewards,1,2,3,4,5,6 a consistent and striking result across varied experimental paradigms is that in initially acquiring such tasks, large differences between individuals are apparent in the learning process.7,8,9,10,11,12 What neural mechanisms contribute to initial task acquisition, and why do some individuals learn a new task much more quickly than others? To address these questions, we recorded longitudinally from dopaminergic (DA) axon terminals in mice learning a visual decision-making task.7 Across striatum, DA responses tracked idiosyncratic and side-specific learning trajectories, consistent with widespread reward prediction error coding across DA terminals. However, even before any rewards were delivered, contralateral-side-specific visual responses were present in DA terminals, primarily in the dorsomedial striatum (DMS). These pre-existing responses predicted the extent of learning for contralateral stimuli. Moreover, activation of these terminals improved contralateral performance. Thus, the initial conditions of a projection-specific and feature-specific DA signal help explain individual learning trajectories. More broadly, this work suggests that functional heterogeneity across DA projections may serve to bias target regions toward learning about different subsets of task features, providing a potential mechanism to address the dimensionality of the initial task learning problem.
Collapse
Affiliation(s)
- Alejandro Pan-Vazquez
- Princeton Neuroscience Institute, Princeton University, Washington Road, Princeton, NJ 08540, USA
| | - Yoel Sanchez Araujo
- Princeton Neuroscience Institute, Princeton University, Washington Road, Princeton, NJ 08540, USA
| | - Brenna McMannon
- Princeton Neuroscience Institute, Princeton University, Washington Road, Princeton, NJ 08540, USA
| | - Miranta Louka
- Princeton Neuroscience Institute, Princeton University, Washington Road, Princeton, NJ 08540, USA
| | - Akhil Bandi
- Princeton Neuroscience Institute, Princeton University, Washington Road, Princeton, NJ 08540, USA
| | - Laura Haetzel
- Princeton Neuroscience Institute, Princeton University, Washington Road, Princeton, NJ 08540, USA
| | - Mayo Faulkner
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Jonathan W Pillow
- Princeton Neuroscience Institute, Princeton University, Washington Road, Princeton, NJ 08540, USA
| | - Nathaniel D Daw
- Princeton Neuroscience Institute, Princeton University, Washington Road, Princeton, NJ 08540, USA; Department of Psychology, Princeton University, Washington Road, Princeton, NJ 08540, USA.
| | - Ilana B Witten
- Princeton Neuroscience Institute, Princeton University, Washington Road, Princeton, NJ 08540, USA; Howard Hughes Medical Institute, Princeton University, Washington Road, Princeton, NJ 08540, USA.
| |
Collapse
|
6
|
Kim MJ, Gibson DJ, Hu D, Yoshida T, Hueske E, Matsushima A, Mahar A, Schofield CJ, Sompolpong P, Tran KT, Tian L, Graybiel AM. Dopamine release plateau and outcome signals in dorsal striatum contrast with classic reinforcement learning formulations. Nat Commun 2024; 15:8856. [PMID: 39402067 PMCID: PMC11473536 DOI: 10.1038/s41467-024-53176-7] [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: 07/26/2023] [Accepted: 10/03/2024] [Indexed: 10/17/2024] Open
Abstract
We recorded dopamine release signals in centromedial and centrolateral sectors of the striatum as mice learned consecutive versions of visual cue-outcome conditioning tasks. Dopamine release responses differed for the centromedial and centrolateral sites. In neither sector could these be accounted for by classic reinforcement learning alone as classically applied to the activity of nigral dopamine-containing neurons. Medially, cue responses ranged from initial sharp peaks to modulated plateau responses; outcome (reward) responses during cue conditioning were minimal or, initially, negative. At centrolateral sites, by contrast, strong, transient dopamine release responses occurred at both cue and outcome. Prolonged, plateau release responses to cues emerged in both regions when discriminative behavioral responses became required. At most sites, we found no evidence for a transition from outcome signaling to cue signaling, a hallmark of temporal difference reinforcement learning as applied to midbrain dopaminergic neuronal activity. These findings delineate a reshaping of striatal dopamine release activity during learning and suggest that current views of reward prediction error encoding need review to accommodate distinct learning-related spatial and temporal patterns of striatal dopamine release in the dorsal striatum.
Collapse
Affiliation(s)
- Min Jung Kim
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, 43 Vassar St., Cambridge, MA, 02139, USA
- Advanced Imaging Research Center, University of Texas, Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Daniel J Gibson
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, 43 Vassar St., Cambridge, MA, 02139, USA
| | - Dan Hu
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, 43 Vassar St., Cambridge, MA, 02139, USA
| | - Tomoko Yoshida
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, 43 Vassar St., Cambridge, MA, 02139, USA
| | - Emily Hueske
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, 43 Vassar St., Cambridge, MA, 02139, USA
| | - Ayano Matsushima
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, 43 Vassar St., Cambridge, MA, 02139, USA
| | - Ara Mahar
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, 43 Vassar St., Cambridge, MA, 02139, USA
| | - Cynthia J Schofield
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, 43 Vassar St., Cambridge, MA, 02139, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Patlapa Sompolpong
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, 43 Vassar St., Cambridge, MA, 02139, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Kathy T Tran
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, 43 Vassar St., Cambridge, MA, 02139, USA
| | - Lin Tian
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, 33458, USA
| | - Ann M Graybiel
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, 43 Vassar St., Cambridge, MA, 02139, USA.
| |
Collapse
|
7
|
Long C, Lee K, Yang L, Dafalias T, Wu AK, Masmanidis SC. Constraints on the subsecond modulation of striatal dynamics by physiological dopamine signaling. Nat Neurosci 2024; 27:1977-1986. [PMID: 38961230 DOI: 10.1038/s41593-024-01699-z] [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: 06/27/2022] [Accepted: 06/07/2024] [Indexed: 07/05/2024]
Abstract
Dopaminergic neurons play a crucial role in associative learning, but their capacity to regulate behavior on subsecond timescales remains debated. It is thought that dopaminergic neurons drive certain behaviors by rapidly modulating striatal spiking activity; however, a view has emerged that only artificially high (that is, supra-physiological) dopamine signals alter behavior on fast timescales. This raises the possibility that moment-to-moment striatal spiking activity is not strongly shaped by dopamine signals in the physiological range. To test this, we transiently altered dopamine levels while monitoring spiking responses in the ventral striatum of behaving mice. These manipulations led to only weak changes in striatal activity, except when dopamine release exceeded reward-matched levels. These findings suggest that dopaminergic neurons normally play a minor role in the subsecond modulation of striatal dynamics in relation to other inputs and demonstrate the importance of discerning dopaminergic neuron contributions to brain function under physiological and potentially nonphysiological conditions.
Collapse
Affiliation(s)
- Charltien Long
- Department of Neurobiology, University of California, Los Angeles, CA, USA
- Medical Scientist Training Program, University of California, Los Angeles, CA, USA
| | - Kwang Lee
- Department of Neurobiology, University of California, Los Angeles, CA, USA
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Long Yang
- Department of Neurobiology, University of California, Los Angeles, CA, USA
| | - Theresia Dafalias
- Department of Neurobiology, University of California, Los Angeles, CA, USA
- Graduate Program for Neuroscience, Boston University, Boston, MA, USA
| | - Alexander K Wu
- Department of Neurobiology, University of California, Los Angeles, CA, USA
| | - Sotiris C Masmanidis
- Department of Neurobiology, University of California, Los Angeles, CA, USA.
- California Nanosystems Institute, University of California, Los Angeles, CA, USA.
| |
Collapse
|
8
|
Cazalé-Debat L, Scheunemann L, Day M, Fernandez-D V Alquicira T, Dimtsi A, Zhang Y, Blackburn LA, Ballardini C, Greenin-Whitehead K, Reynolds E, Lin AC, Owald D, Rezaval C. Mating proximity blinds threat perception. Nature 2024; 634:635-643. [PMID: 39198656 PMCID: PMC11485238 DOI: 10.1038/s41586-024-07890-3] [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: 08/18/2023] [Accepted: 07/31/2024] [Indexed: 09/01/2024]
Abstract
Romantic engagement can bias sensory perception. This 'love blindness' reflects a common behavioural principle across organisms: favouring pursuit of a coveted reward over potential risks1. In the case of animal courtship, such sensory biases may support reproductive success but can also expose individuals to danger, such as predation2,3. However, how neural networks balance the trade-off between risk and reward is unknown. Here we discover a dopamine-governed filter mechanism in male Drosophila that reduces threat perception as courtship progresses. We show that during early courtship stages, threat-activated visual neurons inhibit central courtship nodes via specific serotonergic neurons. This serotonergic inhibition prompts flies to abort courtship when they see imminent danger. However, as flies advance in the courtship process, the dopaminergic filter system reduces visual threat responses, shifting the balance from survival to mating. By recording neural activity from males as they approach mating, we demonstrate that progress in courtship is registered as dopaminergic activity levels ramping up. This dopamine signalling inhibits the visual threat detection pathway via Dop2R receptors, allowing male flies to focus on courtship when they are close to copulation. Thus, dopamine signalling biases sensory perception based on perceived goal proximity, to prioritize between competing behaviours.
Collapse
Affiliation(s)
- Laurie Cazalé-Debat
- School of Biosciences, University of Birmingham, Birmingham, UK
- Birmingham Centre for Neurogenetics, University of Birmingham, Birmingham, UK
| | - Lisa Scheunemann
- Freie Universität Berlin, Institute of Biology, Berlin, Germany
- Institut für Neurophysiologie and NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Megan Day
- School of Biosciences, University of Birmingham, Birmingham, UK
- Birmingham Centre for Neurogenetics, University of Birmingham, Birmingham, UK
| | - Tania Fernandez-D V Alquicira
- Institut für Neurophysiologie and NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Anna Dimtsi
- School of Biosciences, University of Birmingham, Birmingham, UK
- Birmingham Centre for Neurogenetics, University of Birmingham, Birmingham, UK
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Youchong Zhang
- School of Biosciences, University of Birmingham, Birmingham, UK
- Birmingham Centre for Neurogenetics, University of Birmingham, Birmingham, UK
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford, UK
| | - Lauren A Blackburn
- School of Biosciences, University of Birmingham, Birmingham, UK
- Birmingham Centre for Neurogenetics, University of Birmingham, Birmingham, UK
- School of Science and the Environment, University of Worcester, Worcester, UK
| | - Charles Ballardini
- School of Biosciences, University of Birmingham, Birmingham, UK
- Birmingham Centre for Neurogenetics, University of Birmingham, Birmingham, UK
| | - Katie Greenin-Whitehead
- School of Biosciences, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Eric Reynolds
- Institut für Neurophysiologie and NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Andrew C Lin
- School of Biosciences, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - David Owald
- Institut für Neurophysiologie and NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Carolina Rezaval
- School of Biosciences, University of Birmingham, Birmingham, UK.
- Birmingham Centre for Neurogenetics, University of Birmingham, Birmingham, UK.
| |
Collapse
|
9
|
Huang J, Crochet S, Sandi C, Petersen CC. Dopamine dynamics in nucleus accumbens across reward-based learning of goal-directed whisker-to-lick sensorimotor transformations in mice. Heliyon 2024; 10:e37831. [PMID: 39323852 PMCID: PMC11422591 DOI: 10.1016/j.heliyon.2024.e37831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/12/2024] [Accepted: 09/11/2024] [Indexed: 09/27/2024] Open
Abstract
The synaptic and neuronal circuit mechanisms underlying reward-based learning remain to be fully determined. In the mammalian brain, dopamine release in nucleus accumbens is thought to contribute importantly to reward signals for learning and promoting synaptic plasticity. Here, through longitudinal fiber photometry of a genetically-encoded fluorescent sensor, we investigated dopamine signals in the nucleus accumbens of thirsty head-restrained mice as they learned to lick a liquid reward spout in response to single deflections of the C2 whisker across varying reward contingencies. Reward delivery triggered by well-timed licking drove fast transient dopamine increases in nucleus accumbens. On the other hand, unrewarded licking was overall associated with reduced dopamine sensor fluorescence, especially in trials where reward was unexpectedly omitted. The dopamine reward signal upon liquid delivery decreased within individual sessions as mice became sated. As mice learned to lick the reward spout in response to whisker deflection, a fast transient sensory-evoked dopamine signal developed, correlating with the learning of the whisker detection task across consecutive training days, as well as within single learning sessions. The well-defined behavioral paradigm involving a unitary stimulus of a single whisker as a reward-predicting cue along with precisely quantified licking allows untangling of sensory, motor and reward-related dopamine signals and how they evolve across the learning of whisker-dependent goal-directed sensorimotor transformations. Our longitudinal measurements of dopamine dynamics across reward-based learning are overall consistent with the hypothesis that dopamine could play an important role as a reward signal for reinforcement learning.
Collapse
Affiliation(s)
- Jun Huang
- Laboratory of Sensory Processing, Brain Mind Institute, Faculty of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Laboratory of Behavioral Genetics, Brain Mind Institute, Faculty of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Sylvain Crochet
- Laboratory of Sensory Processing, Brain Mind Institute, Faculty of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Carmen Sandi
- Laboratory of Behavioral Genetics, Brain Mind Institute, Faculty of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Carl C.H. Petersen
- Laboratory of Sensory Processing, Brain Mind Institute, Faculty of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
10
|
Muir J, Anguiano M, Kim CK. Neuromodulator and neuropeptide sensors and probes for precise circuit interrogation in vivo. Science 2024; 385:eadn6671. [PMID: 39325905 PMCID: PMC11488521 DOI: 10.1126/science.adn6671] [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: 03/27/2024] [Accepted: 07/01/2024] [Indexed: 09/28/2024]
Abstract
To determine how neuronal circuits encode and drive behavior, it is often necessary to measure and manipulate different aspects of neurochemical signaling in awake animals. Optogenetics and calcium sensors have paved the way for these types of studies, allowing for the perturbation and readout of spiking activity within genetically defined cell types. However, these methods lack the ability to further disentangle the roles of individual neuromodulator and neuropeptides on circuits and behavior. We review recent advances in chemical biology tools that enable precise spatiotemporal monitoring and control over individual neuroeffectors and their receptors in vivo. We also highlight discoveries enabled by such tools, revealing how these molecules signal across different timescales to drive learning, orchestrate behavioral changes, and modulate circuit activity.
Collapse
Affiliation(s)
- J. Muir
- Center for Neuroscience, University of California, Davis, Davis, CA 95618, USA
- Department of Neurology, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - M. Anguiano
- Neuroscience Graduate Group, University of California, Davis, Davis, CA 95616, USA
| | - C. K. Kim
- Center for Neuroscience, University of California, Davis, Davis, CA 95618, USA
- Department of Neurology, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| |
Collapse
|
11
|
Gaertner Z, Oram C, Schneeweis A, Schonfeld E, Bolduc C, Chen C, Dombeck D, Parisiadou L, Poulin JF, Awatramani R. Molecular and spatial transcriptomic classification of midbrain dopamine neurons and their alterations in a LRRK2 G2019S model of Parkinson's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.06.597807. [PMID: 38895448 PMCID: PMC11185743 DOI: 10.1101/2024.06.06.597807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Several studies have revealed that midbrain dopamine (DA) neurons, even within a single neuroanatomical area, display heterogeneous properties. In parallel, studies using single cell profiling techniques have begun to cluster DA neurons into subtypes based on their molecular signatures. Recent work has shown that molecularly defined DA subtypes within the substantia nigra (SNc) display distinctive anatomic and functional properties, and differential vulnerability in Parkinson's disease (PD). Based on these provocative results, a granular understanding of these putative subtypes and their alterations in PD models, is imperative. We developed an optimized pipeline for single-nuclear RNA sequencing (snRNA-seq) and generated a high-resolution hierarchically organized map revealing 20 molecularly distinct DA neuron subtypes belonging to three main families. We integrated this data with spatial MERFISH technology to map, with high definition, the location of these subtypes in the mouse midbrain, revealing heterogeneity even within neuroanatomical sub-structures. Finally, we demonstrate that in the preclinical LRRK2G2019S knock-in mouse model of PD, subtype organization and proportions are preserved. Transcriptional alterations occur in many subtypes including those localized to the ventral tier SNc, where differential expression is observed in synaptic pathways, which might account for previously described DA release deficits in this model. Our work provides an advancement of current taxonomic schemes of the mouse midbrain DA neuron subtypes, a high-resolution view of their spatial locations, and their alterations in a prodromal mouse model of PD.
Collapse
Affiliation(s)
- Zachary Gaertner
- Northwestern University Feinberg School of Medicine, Dept of Neurology, Chicago, IL 60611
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Cameron Oram
- McGill University (Montreal Neurological Institute), Faculty of Medicine and Health Sciences, Dept of Neurology and Neurosurgery, Montreal (QC), Canada
| | - Amanda Schneeweis
- Northwestern University Feinberg School of Medicine, Dept of Neurology, Chicago, IL 60611
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Elan Schonfeld
- Northwestern University Feinberg School of Medicine, Dept of Neurology, Chicago, IL 60611
| | - Cyril Bolduc
- McGill University (Montreal Neurological Institute), Faculty of Medicine and Health Sciences, Dept of Neurology and Neurosurgery, Montreal (QC), Canada
| | - Chuyu Chen
- Northwestern University Feinberg School of Medicine, Dept of Pharmacology, Chicago, IL 60611
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Daniel Dombeck
- Northwestern University, Dept of Neurobiology, Evanston, IL 60201
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Loukia Parisiadou
- Northwestern University Feinberg School of Medicine, Dept of Pharmacology, Chicago, IL 60611
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Jean Francois Poulin
- McGill University (Montreal Neurological Institute), Faculty of Medicine and Health Sciences, Dept of Neurology and Neurosurgery, Montreal (QC), Canada
| | - Rajeshwar Awatramani
- Northwestern University Feinberg School of Medicine, Dept of Neurology, Chicago, IL 60611
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| |
Collapse
|
12
|
Howard JD, Edmonds D, Schoenbaum G, Kahnt T. Distributed midbrain responses signal the content of positive identity prediction errors. Curr Biol 2024; 34:4240-4247.e4. [PMID: 39197457 PMCID: PMC11421979 DOI: 10.1016/j.cub.2024.07.105] [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: 02/28/2024] [Revised: 06/12/2024] [Accepted: 07/31/2024] [Indexed: 09/01/2024]
Abstract
Recent work across species has shown that midbrain dopamine neurons signal not only errors in the prediction of reward value but also in the prediction of value-neutral sensory features. To support learning of associative structures in downstream areas, identity prediction errors (iPEs) should signal specific information about the mis-predicted outcome. Here, we used pattern-based analysis of functional magnetic resonance imaging (fMRI) data acquired during reversal learning to characterize the information content of iPE responses in the human midbrain. We find that fMRI responses to value-neutral identity errors contain information about the identity of the unexpectedly received reward (positive iPE+) but not about the identity of the omitted reward (negative iPE-). Exploratory analyses revealed representations of iPE- in the dorsomedial prefrontal cortex. These results demonstrate that ensemble midbrain responses to value-neutral identity errors convey information about the identity of unexpectedly received outcomes, which could shape the formation of novel stimulus-outcome associations that constitute cognitive maps.
Collapse
Affiliation(s)
- James D Howard
- Department of Psychology, Brandeis University, Waltham, MA 02453, USA.
| | - Donnisa Edmonds
- Department of Neurology, Northwestern University, Chicago, IL 60611, USA
| | - Geoffrey Schoenbaum
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224, USA
| | - Thorsten Kahnt
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224, USA.
| |
Collapse
|
13
|
Gershman SJ, Assad JA, Datta SR, Linderman SW, Sabatini BL, Uchida N, Wilbrecht L. Explaining dopamine through prediction errors and beyond. Nat Neurosci 2024; 27:1645-1655. [PMID: 39054370 DOI: 10.1038/s41593-024-01705-4] [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: 04/24/2023] [Accepted: 06/13/2024] [Indexed: 07/27/2024]
Abstract
The most influential account of phasic dopamine holds that it reports reward prediction errors (RPEs). The RPE-based interpretation of dopamine signaling is, in its original form, probably too simple and fails to explain all the properties of phasic dopamine observed in behaving animals. This Perspective helps to resolve some of the conflicting interpretations of dopamine that currently exist in the literature. We focus on the following three empirical challenges to the RPE theory of dopamine: why does dopamine (1) ramp up as animals approach rewards, (2) respond to sensory and motor features and (3) influence action selection? We argue that the prediction error concept, once it has been suitably modified and generalized based on an analysis of each computational problem, answers each challenge. Nonetheless, there are a number of additional empirical findings that appear to demand fundamentally different theoretical explanations beyond encoding RPE. Therefore, looking forward, we discuss the prospects for a unifying theory that respects the diversity of dopamine signaling and function as well as the complex circuitry that both underlies and responds to dopaminergic transmission.
Collapse
Affiliation(s)
- Samuel J Gershman
- Department of Psychology and Center for Brain Science, Harvard University, Cambridge, MA, USA.
- Kempner Institute for the Study of Natural and Artificial Intelligence, Harvard University, Cambridge, MA, USA.
| | - John A Assad
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | | | - Scott W Linderman
- Department of Statistics and Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Bernardo L Sabatini
- Kempner Institute for the Study of Natural and Artificial Intelligence, Harvard University, Cambridge, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Naoshige Uchida
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Linda Wilbrecht
- Department of Psychology and Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
| |
Collapse
|
14
|
Abstract
Neuroscience has convinced people that much of their behavior is determined by causes unknown to them and beyond their control. However, are advances in neuroscience truly a prerequisite for such beliefs? Robert Kane's theory of ultimate responsibility is libertarian theory. Its innovative nature makes it possible to discuss the neurophysiological basis of its postulates. Using the functions of the midbrain dopaminergic system as an example, this article provides an overview of this neurophysiological basis. According to Kane, if we are to be ultimately responsible for our wills as well as for our actions, some actions in our lives must lack sufficient motives and causes. These are self-forming actions. Dopamine is hypothesized to mediate self-forming action execution. Dopamine not only mediates action but also ensures synaptic plasticity in the brain, that is, learning from action; hence, dopamine changes the acting individual and provides the formation of our own wills. The basal ganglia, which are the main target of dopamine in the brain, act through parallel pathways and are involved in decision-making processes. Dopamine is also involved in the regulation of the neurodynamical properties of prefrontal cortex networks with random spiking noise. It can be assumed that the activity of the dopaminergic system is closely related to the physiological basis of free will.
Collapse
Affiliation(s)
- Natalia Ivlieva
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Butlerova Str., 5a, Moscow, 117485, Russia.
| |
Collapse
|
15
|
Jiang Z, He M, Young C, Cai J, Xu Y, Jiang Y, Li H, Yang M, Tong Q. Dopaminergic Neurons in Zona Incerta Drives Appetitive Self-Grooming. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308974. [PMID: 39099402 PMCID: PMC11422805 DOI: 10.1002/advs.202308974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 07/07/2024] [Indexed: 08/06/2024]
Abstract
Dopaminergic (DA) neurons are known to play a key role in controlling behaviors. While DA neurons in other brain regions are extensively characterized, those in zona incerta (ZITH or A13) receive much less attention and their function remains to be defined. Here it is shown that optogenetic stimulation of these neurons elicited intensive self-grooming behaviors and promoted place preference, which can be enhanced by training but cannot be converted into contextual memory. Interestingly, the same stimulation increased DA release to periaqueductal grey (PAG) neurons and local PAG antagonism of DA action reduced the elicited self-grooming. In addition, A13 neurons increased their activity in response to various external stimuli and during natural self-grooming episodes. Finally, monosynaptic retrograde tracing showed that the paraventricular hypothalamus represents one of the major upstream brain regions to A13 neurons. Taken together, these results reveal that A13 neurons are one of the brain sites that promote appetitive self-grooming involving DA release to the PAG.
Collapse
Affiliation(s)
- Zhiying Jiang
- The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Michelle He
- The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
- Summer Undergraduate Research Program, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
- Sargent College of Health and Rehabilitation Sciences, Boston University, Boston, MA, 02215, USA
| | - Claire Young
- The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Jing Cai
- The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
- MD Anderson Cancer Center & UTHealth Graduate School for Biomedical Sciences, University of Texas Health Science at Houston, Houston, TX, 77030, USA
| | - Yuanzhong Xu
- The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Yanyan Jiang
- The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Hongli Li
- The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Maojie Yang
- The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Qingchun Tong
- The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
- MD Anderson Cancer Center & UTHealth Graduate School for Biomedical Sciences, University of Texas Health Science at Houston, Houston, TX, 77030, USA
| |
Collapse
|
16
|
Wang Y, Xu H, Chen S, Chen J, Zheng Q, Ma Y, Zhao X, Shi Y, Xiao L. Oxytocin Protects Nigrostriatal Dopamine Signal via Activating GABAergic Circuit in the MPTP-Induced Parkinson's Disease Model. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2310244. [PMID: 39099429 PMCID: PMC11423065 DOI: 10.1002/advs.202310244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 07/23/2024] [Indexed: 08/06/2024]
Abstract
The most pronounced neuropathological feature of Parkinson's disease (PD) is the loss of dopamine (DA) neurons in the substantia nigra compacta (SNc), which depletes striatal DA. Hypothalamic oxytocin is found to be reduced in PD patients and closely interacts with the DA system, but the role of oxytocin in PD remains unclear. Here, the disturbances of endogenous oxytocin level and the substantia nigra (SN) oxytocin receptor expression in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mouse model is observed, correlated with the striatal tyrosine hydroxylase (TH) expression reduction. Killing/silencing hypothalamic oxytocin neurons aggravates the vulnerability of nigrostriatal DA signal to MPTP, whereas elevating oxytocin level by intranasal delivery or microinjecting into the SN promotes the resistance. In addition, knocking out SN oxytocin receptors induces the time-dependent reductions of SNc DA neurons, striatal TH expression, and striatal DA level by increasing neuronal excitotoxicity. These results further uncover that oxytocin dampens the excitatory synaptic inputs onto DA neurons via activating oxytocin receptor-expressed SN GABA neurons, which target GABA(B) receptors expressed in SNc DA neuron-projecting glutamatergic axons, to reduce excitotoxicity. Thus, besides the well-known prosocial effect, oxytocin acts as a key endogenous factor in protecting the nigrostriatal DA system.
Collapse
Affiliation(s)
- Yurong Wang
- Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Hao Xu
- Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Saiyong Chen
- Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Junhao Chen
- Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Qimeng Zheng
- Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Yuanyuan Ma
- Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Xinru Zhao
- Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Ying Shi
- Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Lei Xiao
- Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| |
Collapse
|
17
|
Beauséjour PA, Veilleux JC, Condamine S, Zielinski BS, Dubuc R. Olfactory Projections to Locomotor Control Centers in the Sea Lamprey. Int J Mol Sci 2024; 25:9370. [PMID: 39273317 PMCID: PMC11395479 DOI: 10.3390/ijms25179370] [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: 07/11/2024] [Revised: 08/20/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
Although olfaction is well known to guide animal behavior, the neural circuits underlying the motor responses elicited by olfactory inputs are poorly understood. In the sea lamprey, anatomical evidence shows that olfactory inputs project to the posterior tuberculum (PT), a structure containing dopaminergic (DA) neurons homologous to the mammalian ventral tegmental area and the substantia nigra pars compacta. Olfactory inputs travel directly from the medial olfactory bulb (medOB) or indirectly through the main olfactory bulb and the lateral pallium (LPal). Here, we characterized the transmission of olfactory inputs to the PT in the sea lamprey, Petromyzon marinus. Abundant projections from the medOB were observed close to DA neurons of the PT. Moreover, electrophysiological experiments revealed that PT neurons are activated by both the medOB and LPal, and calcium imaging indicated that the olfactory signal is then relayed to the mesencephalic locomotor region to initiate locomotion. In semi-intact preparations, stimulation of the medOB and LPal induced locomotion that was tightly associated with neural activity in the PT. Moreover, PT neurons were active throughout spontaneously occurring locomotor bouts. Altogether, our observations suggest that the medOB and LPal convey olfactory inputs to DA neurons of the PT, which in turn activate the brainstem motor command system to elicit locomotion.
Collapse
Affiliation(s)
| | - Jean-Christophe Veilleux
- Research Group in Adapted Physical Activity, Department of Exercise Sciences, Faculty of Sciences, University of Quebec in Montreal, Montreal, QC H2X 1Y4, Canada
| | - Steven Condamine
- Department of Neurosciences, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Barbara S Zielinski
- Department of Integrative Biology, Faculty of Science, University of Windsor, Windsor, ON N9B 3P4, Canada
| | - Réjean Dubuc
- Department of Neurosciences, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
- Research Group in Adapted Physical Activity, Department of Exercise Sciences, Faculty of Sciences, University of Quebec in Montreal, Montreal, QC H2X 1Y4, Canada
| |
Collapse
|
18
|
Callahan JW, Morales JC, Atherton JF, Wang D, Kostic S, Bevan MD. Movement-related increases in subthalamic activity optimize locomotion. Cell Rep 2024; 43:114495. [PMID: 39068661 PMCID: PMC11407793 DOI: 10.1016/j.celrep.2024.114495] [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: 11/17/2023] [Revised: 05/27/2024] [Accepted: 06/27/2024] [Indexed: 07/30/2024] Open
Abstract
The subthalamic nucleus (STN) is traditionally thought to restrict movement. Lesion or prolonged STN inhibition increases movement vigor and propensity, while optogenetic excitation has opposing effects. However, STN neurons often exhibit movement-related increases in firing. To address this paradox, STN activity was recorded and manipulated in head-fixed mice at rest and during self-initiated and self-paced treadmill locomotion. We found that (1) most STN neurons (type 1) exhibit locomotion-dependent increases in activity, with half firing preferentially during the propulsive phase of the contralateral locomotor cycle; (2) a minority of STN neurons exhibit dips in activity or are uncorrelated with movement; (3) brief optogenetic inhibition of the lateral STN (where type 1 neurons are concentrated) slows and prematurely terminates locomotion; and (4) in Q175 Huntington's disease mice, abnormally brief, low-velocity locomotion is associated with type 1 hypoactivity. Together, these data argue that movement-related increases in STN activity contribute to optimal locomotor performance.
Collapse
Affiliation(s)
- Joshua W Callahan
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Juan Carlos Morales
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Jeremy F Atherton
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Dorothy Wang
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Selena Kostic
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Mark D Bevan
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
19
|
Brown E, Zi Y, Vu MA, Bouabid S, Lindsey J, Godfrey-Nwachukwu C, Attarwala A, Litwin-Kumar A, DePasquale B, Howe M. Spatially organized striatal neuromodulator release encodes trajectory errors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.13.607797. [PMID: 39185163 PMCID: PMC11343099 DOI: 10.1101/2024.08.13.607797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Goal-directed navigation requires animals to continuously evaluate their current direction and speed of travel relative to landmarks to discern whether they are approaching or deviating from their goal. Striatal dopamine and acetylcholine are powerful modulators of goal-directed behavior, but it is unclear whether and how neuromodulator dynamics at landmarks incorporate relative motion for effective behavioral guidance. Using optical measurements in mice, we demonstrate that cue-evoked striatal dopamine release encodes bi-directional 'trajectory errors' reflecting relationships between ongoing speed and direction of locomotion and visual flow relative to optimal goal trajectories. Striatum-wide micro-fiber array recordings resolved an anatomical gradient of trajectory error signaling across the anterior-posterior axis, distinct from trajectory error independent cue signals. Dynamic regression modeling revealed that positive and negative trajectory error encoding emerges early and late respectively during learning and over different time courses in the medial and lateral striatum, enabling region specific contributions to learning. Striatal acetylcholine release also encodes trajectory errors, but encoding is more spatially restricted, opposite polarity, and delayed relative to dopamine, supporting distinct roles in modulating striatal output and behavior. Dopamine trajectory error signaling and task performance were reproduced in a reinforcement learning model incorporating a conjunctive state space representation, suggesting a potential neural substrate for trajectory error generation. Our results establish region specific neuromodulator signals positioned to guide the speed and direction of locomotion to reach goals based on environmental landmarks during navigation.
Collapse
Affiliation(s)
- Eleanor Brown
- Department of Psychological & Brain Sciences, Boston University, Boston, MA, USA
| | - Yihan Zi
- Department of Psychological & Brain Sciences, Boston University, Boston, MA, USA
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Mai-Anh Vu
- Department of Psychological & Brain Sciences, Boston University, Boston, MA, USA
| | - Safa Bouabid
- Department of Psychological & Brain Sciences, Boston University, Boston, MA, USA
| | - Jack Lindsey
- Department of Neuroscience, Columbia University, New York, NY, USA
| | | | - Aaquib Attarwala
- Department of Psychological & Brain Sciences, Boston University, Boston, MA, USA
| | | | - Brian DePasquale
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Mark Howe
- Department of Psychological & Brain Sciences, Boston University, Boston, MA, USA
| |
Collapse
|
20
|
Roth RH, Ding JB. Cortico-basal ganglia plasticity in motor learning. Neuron 2024; 112:2486-2502. [PMID: 39002543 PMCID: PMC11309896 DOI: 10.1016/j.neuron.2024.06.014] [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: 02/29/2024] [Revised: 05/29/2024] [Accepted: 06/17/2024] [Indexed: 07/15/2024]
Abstract
One key function of the brain is to control our body's movements, allowing us to interact with the world around us. Yet, many motor behaviors are not innate but require learning through repeated practice. Among the brain's motor regions, the cortico-basal ganglia circuit is particularly crucial for acquiring and executing motor skills, and neuronal activity in these regions is directly linked to movement parameters. Cell-type-specific adaptations of activity patterns and synaptic connectivity support the learning of new motor skills. Functionally, neuronal activity sequences become structured and associated with learned movements. On the synaptic level, specific connections become potentiated during learning through mechanisms such as long-term synaptic plasticity and dendritic spine dynamics, which are thought to mediate functional circuit plasticity. These synaptic and circuit adaptations within the cortico-basal ganglia circuitry are thus critical for motor skill acquisition, and disruptions in this plasticity can contribute to movement disorders.
Collapse
Affiliation(s)
- Richard H Roth
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA.
| | - Jun B Ding
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA; Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA; The Phil & Penny Knight Initiative for Brain Resilience at the Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
21
|
Halfhide C, Cammarano TL, Anderson KA, Galik SM, Rossignol J, Dunbar GL, Sandstrom MI. Using microdialysis to monitor dopaminergic support of limb-use control following mesencephalic neurosphere transplantation in a rodent model of Parkinson's Disease. Behav Brain Res 2024; 471:115121. [PMID: 38945302 DOI: 10.1016/j.bbr.2024.115121] [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: 09/05/2023] [Revised: 05/31/2024] [Accepted: 06/19/2024] [Indexed: 07/02/2024]
Abstract
Controlled nigrostriatal dopamine release supports effective limb use during locomotion coordination that becomes compromised after this pathway deteriorates in Parkinson's Disease (PD). How dopamine release relates to active ongoing behavior control remains unknown. Restoring proper release strategy appears important to successful PD treatment with transplanted dopamine-producing stem cells. This is suggested by apparently distinct behavioral support from tonic or phasic release and corresponding requirements of requisite afferent control exhibited by intact nigrostriatal neurons. Our laboratory previously demonstrated that transplanted dopaminergic cells can elicit skilled movement recovery known to depend on phasic dopamine release. However, efforts to measure this movement-related dopamine release yielded seemingly paradoxical, incongruent results. In response, here we explored whether those previous observations derived from rapid reuptake transport into either transplanted cells or residual, lesion-surviving terminals. We confirmed this using minimal reuptake blockade during intrastriatal microdialysis. After unilateral dopamine depletion, rats received transplants and were subjected to our swimming protocol. Among dopamine-depleted and transplanted rats, treatment supported restoration of limb movement symmetry. Interestingly, subsequent reuptake-restricted microdialysis confirmed distinct swimming-induced dopamine increases clearly occurred among these lesioned/transplanted subjects. Thus, phasic firing control appears to contribute to transplant-derived recovery in Parkinsonian animals.
Collapse
Affiliation(s)
- Chloé Halfhide
- Central Michigan University, Program in Neuroscience, Mt. Pleasant, MI, United States
| | - Tommie L Cammarano
- Central Michigan University, Program in Neuroscience, Mt. Pleasant, MI, United States
| | - Kevin A Anderson
- Central Michigan University, Experimental Psychology Program, Mt. Pleasant, MI, United States
| | - Stefani M Galik
- Central Michigan University, Experimental Psychology Program, Mt. Pleasant, MI, United States
| | - Julien Rossignol
- Central Michigan University, Program in Neuroscience, Mt. Pleasant, MI, United States; Central Michigan University, College of Medicine, Mt. Pleasant, MI, United States
| | - Gary L Dunbar
- Central Michigan University, Program in Neuroscience, Mt. Pleasant, MI, United States; Central Michigan University, Experimental Psychology Program, Mt. Pleasant, MI, United States
| | - Michael I Sandstrom
- Central Michigan University, Program in Neuroscience, Mt. Pleasant, MI, United States; Central Michigan University, Experimental Psychology Program, Mt. Pleasant, MI, United States.
| |
Collapse
|
22
|
Li Z, Mukherjee D, Duric B, Austin-Zimmerman I, Trotta G, Spinazzola E, Quattrone D, Murray RM, Di Forti M. Systematic review and meta-analysis on the effects of chronic peri-adolescent cannabinoid exposure on schizophrenia-like behaviour in rodents. Mol Psychiatry 2024:10.1038/s41380-024-02668-5. [PMID: 39090371 DOI: 10.1038/s41380-024-02668-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 07/02/2024] [Accepted: 07/05/2024] [Indexed: 08/04/2024]
Abstract
BACKGROUND The link between cannabis use and schizophrenia is well-established in epidemiological studies, especially among adolescents with early-onset use. However, this association in rodent models is less clear. This meta-analysis examined the effects of adolescent cannabinoid exposure on distinct schizophrenia-like behaviours in rodents and how experimental variations influence outcomes. METHODS Following a pre-registered protocol (CRD42022338761), we searched PubMed, Ovid Medline, Embse and APA PsychInfo for English-language original studies until May 2024. We synthesised data from experiments on schizophrenia-like behaviour in rats and mice after repeated peri-pubertal (onset between P23-P45) cannabinoid exposure. Risk of bias was assessed using the SYRCLE's tool. RESULTS We included 359 experiments from 108 articles across 9 behavioural tests. We found meta-analytic evidence supporting that CB1R agonists, both natural and synthetic, elicited broad schizophrenia-like behavioural alterations, including impaired working memory [g = -0.56; (CI: -0.93, -0.18)], novel object recognition [g = -0.66; (CI: -0.97, -0.35)], novel object location recognition [g = -0.70; (CI: -1.07, -0.33]), social novelty preference [g = -0.52; (CI: -0.93, -0.11)], social motivation [g = -0.21; (CI: -0.42, -0.00)], pre-pulse inhibition [g = -0.43; (CI: -0.76, -0.10)], and sucrose preference [g = -0.87; (CI: -1.46, -0.27)]. By contrast, effects on novelty-induced locomotion were negligible. Subgroup analyses revealed similar effects across sexes and species. Substantial variance in the protocols and moderate-to-high heterogeneity in behavioural outcomes were observed. We found CBD may enhance fear memory recall, but data was limited. DISCUSSION This is the first meta-analysis to comprehensively assess the link between cannabinoids and schizophrenia-like behaviours in rodents. Our results support epidemiological links between early cannabis use and schizophrenia-like phenotypes, confirming the utility of animal models. Standardising protocols will optimise models to strengthen reproducibility and comparisons, our work provides a framework for refining rodent models to elucidate biological pathways linking cannabis and schizophrenia.
Collapse
Affiliation(s)
- Zhikun Li
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 8AF, UK
| | - Diptendu Mukherjee
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, London, SE1 1UL, UK
| | - Bea Duric
- GKT School of Medical Education, King's College London, London, SE1 1UL, UK
| | - Isabelle Austin-Zimmerman
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 8AF, UK
| | - Giulia Trotta
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 8AF, UK
- South London and Maudsley NHS Mental Health Foundation Trust, London, UK
| | - Edoardo Spinazzola
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 8AF, UK
- South London and Maudsley NHS Mental Health Foundation Trust, London, UK
| | - Diego Quattrone
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 8AF, UK
- South London and Maudsley NHS Mental Health Foundation Trust, London, UK
| | - Robin M Murray
- South London and Maudsley NHS Mental Health Foundation Trust, London, UK
- Department of Psychosis Studies, Institute of Psychiatry, King's College London, De Crespigny Park, Denmark Hill, London, SE5 8AF, UK
| | - Marta Di Forti
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 8AF, UK.
- South London and Maudsley NHS Mental Health Foundation Trust, London, UK.
- National Institute for Health Research (NIHR) Mental Health Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London, London, UK.
| |
Collapse
|
23
|
Vakilipour P, Fekrvand S. Brain-to-brain interface technology: A brief history, current state, and future goals. Int J Dev Neurosci 2024; 84:351-367. [PMID: 38711277 DOI: 10.1002/jdn.10334] [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: 11/29/2023] [Revised: 04/05/2024] [Accepted: 04/18/2024] [Indexed: 05/08/2024] Open
Abstract
A brain-to-brain interface (BBI), defined as a combination of neuroimaging and neurostimulation methods to extract and deliver information between brains directly without the need for the peripheral nervous system, is a budding communication technique. A BBI system is made up of two parts known as the brain-computer interface part, which reads a sender's brain activity and digitalizes it, and the computer-brain interface part, which writes the delivered brain activity to a receiving brain. As with other technologies, BBI systems have gone through an evolutionary process since they first appeared. The BBI systems have been employed for numerous purposes, including rehabilitation for post-stroke patients, communicating with patients suffering from amyotrophic lateral sclerosis, locked-in syndrome and speech problems following stroke. Also, it has been proposed that a BBI system could play an important role on future battlefields. This technology was not only employed for communicating between two human brains but also for making a direct communication path among different species through which motor or sensory commands could be sent and received. However, the application of BBI systems has provoked significant challenges to human rights principles due to their ability to access and manipulate human brain information. In this study, we aimed to review the brain-computer interface and computer-brain interface technologies as components of BBI systems, the development of BBI systems, applications of this technology, arising ethical issues and expectations for future use.
Collapse
Affiliation(s)
- Pouya Vakilipour
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Saba Fekrvand
- Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
Lee RS, Sagiv Y, Engelhard B, Witten IB, Daw ND. A feature-specific prediction error model explains dopaminergic heterogeneity. Nat Neurosci 2024; 27:1574-1586. [PMID: 38961229 DOI: 10.1038/s41593-024-01689-1] [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: 02/08/2022] [Accepted: 05/22/2024] [Indexed: 07/05/2024]
Abstract
The hypothesis that midbrain dopamine (DA) neurons broadcast a reward prediction error (RPE) is among the great successes of computational neuroscience. However, recent results contradict a core aspect of this theory: specifically that the neurons convey a scalar, homogeneous signal. While the predominant family of extensions to the RPE model replicates the classic model in multiple parallel circuits, we argue that these models are ill suited to explain reports of heterogeneity in task variable encoding across DA neurons. Instead, we introduce a complementary 'feature-specific RPE' model, positing that individual ventral tegmental area DA neurons report RPEs for different aspects of an animal's moment-to-moment situation. Further, we show how our framework can be extended to explain patterns of heterogeneity in action responses reported among substantia nigra pars compacta DA neurons. This theory reconciles new observations of DA heterogeneity with classic ideas about RPE coding while also providing a new perspective of how the brain performs reinforcement learning in high-dimensional environments.
Collapse
Affiliation(s)
- Rachel S Lee
- Princeton Neuroscience Institute, Princeton, NJ, USA
| | - Yotam Sagiv
- Princeton Neuroscience Institute, Princeton, NJ, USA
| | - Ben Engelhard
- Princeton Neuroscience Institute, Princeton, NJ, USA
| | | | - Nathaniel D Daw
- Princeton Neuroscience Institute, Princeton, NJ, USA.
- Department of Psychology, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
25
|
Young PA, Waller O, Ball K, Williams CC, Nashmi R. Phasic Stimulation of Dopaminergic Neurons of the Lateral Substantia Nigra Increases Open Field Exploratory Behaviour and Reduces Habituation Over Time. Neuroscience 2024; 551:276-289. [PMID: 38838978 DOI: 10.1016/j.neuroscience.2024.05.025] [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: 10/05/2023] [Revised: 04/09/2024] [Accepted: 05/20/2024] [Indexed: 06/07/2024]
Abstract
Transient nigrostriatal dopaminergic signalling is well known for its role in reinforcement learning and increasingly so for its role in the initiation of voluntary movement. However, how transient bursts of dopamine modulate voluntary movement remains unclear, likely due to the heterogeneity of the nigrostriatal system, the focus of optogenetic studies on locomotion at sub-sec time intervals, and the overlapping roles of phasic dopamine in behaviour and novelty signalling. In this study we investigated how phasic activity in the lateral substantia nigra pars compacta (lateral SNc) over time affects voluntary behaviours during exploration. Using a transgenic mouse model of both sexes expressing channelrhodopsin (ChR2) in dopamine transporter-expressing cells, we stimulated the lateral SNc while mice explored an open field over two consecutive days. We found that phasic activation of the lateral SNc induced an increase in exploratory behaviours including horizontal movement activity, locomotion initiation, and rearing specifically on the first open field exposure, but not on the second day. In addition, stimulated animals did not habituate to the same extent as their ChR2-negative counterparts, as indicated by a lack of decrease in baseline activity. These findings suggest that rather than prompting voluntary movement in general, phasic nigrostriatal dopamine prompts context-appropriate behaviours. In addition, dopamine signalling that modulates movement acts over longer timescales than the transient signal, affecting behaviour even after the signal has ended.
Collapse
Affiliation(s)
- Penelope A Young
- Department of Biology, University of Victoria, British Columbia V8W 2Y2, Canada; Division of Medical Sciences, University of Victoria, British Columbia V8W 2Y2, Canada
| | - Olivia Waller
- Department of Biology, University of Victoria, British Columbia V8W 2Y2, Canada
| | - Katherine Ball
- Department of Biology, University of Victoria, British Columbia V8W 2Y2, Canada
| | - Chad C Williams
- Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA
| | - Raad Nashmi
- Department of Biology, University of Victoria, British Columbia V8W 2Y2, Canada; Division of Medical Sciences, University of Victoria, British Columbia V8W 2Y2, Canada.
| |
Collapse
|
26
|
Engel L, Wolff AR, Blake M, Collins VL, Sinha S, Saunders BT. Dopamine neurons drive spatiotemporally heterogeneous striatal dopamine signals during learning. Curr Biol 2024; 34:3086-3101.e4. [PMID: 38925117 PMCID: PMC11279555 DOI: 10.1016/j.cub.2024.05.069] [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: 03/25/2024] [Revised: 04/25/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024]
Abstract
Environmental cues, through Pavlovian learning, become conditioned stimuli that invigorate and guide animals toward rewards. Dopamine (DA) neurons in the ventral tegmental area (VTA) and substantia nigra (SNc) are crucial for this process, via engagement of a reciprocally connected network with their striatal targets. Critically, it remains unknown how dopamine neuron activity itself engages dopamine signals throughout the striatum, across learning. Here, we investigated how optogenetic Pavlovian cue conditioning of VTA or SNc dopamine neurons directs cue-evoked behavior and shapes subregion-specific striatal dopamine dynamics. We used a fluorescent biosensor to monitor dopamine in the nucleus accumbens (NAc) core and shell, dorsomedial striatum (DMS), and dorsolateral striatum (DLS). We demonstrate spatially heterogeneous, learning-dependent dopamine changes across striatal regions. Although VTA stimulation-evoked robust dopamine release in NAc core, shell, and DMS, predictive cues preferentially recruited dopamine release in NAc core, starting early in training, and DMS, late in training. Negative prediction error signals, reflecting a violation in the expectation of dopamine neuron activation, only emerged in the NAc core and DMS. Despite the development of vigorous movement late in training, conditioned dopamine signals did not emerge in the DLS, even during Pavlovian conditioning with SNc dopamine neuron activation, which elicited robust DLS dopamine release. Together, our studies show a broad dissociation in the fundamental prediction and reward-related information generated by VTA and SNc dopamine neuron populations and signaled by dopamine across the striatum. Further, they offer new insight into how larger-scale adaptations across the striatal network emerge during learning to coordinate behavior.
Collapse
Affiliation(s)
- Liv Engel
- Department of Neuroscience, University of Minnesota, 2001 6th St SE, Minneapolis, MN 55455, USA; Medical Discovery Team on Addiction, University of Minnesota, 2001 6th St SE, Minneapolis, MN 55455, USA
| | - Amy R Wolff
- Department of Neuroscience, University of Minnesota, 2001 6th St SE, Minneapolis, MN 55455, USA; Medical Discovery Team on Addiction, University of Minnesota, 2001 6th St SE, Minneapolis, MN 55455, USA
| | - Madelyn Blake
- Department of Neuroscience, University of Minnesota, 2001 6th St SE, Minneapolis, MN 55455, USA
| | - Val L Collins
- Department of Neuroscience, University of Minnesota, 2001 6th St SE, Minneapolis, MN 55455, USA; Medical Discovery Team on Addiction, University of Minnesota, 2001 6th St SE, Minneapolis, MN 55455, USA
| | - Sonal Sinha
- Krieger School of Arts & Sciences, Johns Hopkins University, 3400 N. Charles St, Baltimore, MD 21218, USA
| | - Benjamin T Saunders
- Department of Neuroscience, University of Minnesota, 2001 6th St SE, Minneapolis, MN 55455, USA; Medical Discovery Team on Addiction, University of Minnesota, 2001 6th St SE, Minneapolis, MN 55455, USA.
| |
Collapse
|
27
|
Ohtake M, Abe K, Hasegawa M, Itokazu T, Selvakumar V, Matunis A, Stacy E, Froebrich E, Huynh N, Lee H, Kambe Y, Yamamoto T, Sato TK, Sato TR. Encoding of self-initiated actions in axon terminals of the mesocortical pathway. NEUROPHOTONICS 2024; 11:033408. [PMID: 38726349 PMCID: PMC11080647 DOI: 10.1117/1.nph.11.3.033408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 03/01/2024] [Accepted: 03/05/2024] [Indexed: 05/12/2024]
Abstract
Significance The initiation of goal-directed actions is a complex process involving the medial prefrontal cortex and dopaminergic inputs through the mesocortical pathway. However, it is unclear what information the mesocortical pathway conveys and how it impacts action initiation. In this study, we unveiled the indispensable role of mesocortical axon terminals in encoding the execution of movements in self-initiated actions. Aim To investigate the role of mesocortical axon terminals in encoding the execution of movements in self-initiated actions. Approach We designed a lever-press task in which mice internally determine the timing of the press, receiving a larger reward for longer waiting periods. Results Our study revealed that self-initiated actions depend on dopaminergic signaling mediated by D2 receptors, whereas sensory-triggered lever-press actions do not involve D2 signaling. Microprism-mediated two-photon calcium imaging further demonstrated ramping activity in mesocortical axon terminals approximately 0.5 s before the self-initiated lever press. Remarkably, the ramping patterns remained consistent whether the mice responded to cues immediately for a smaller reward or held their response for a larger reward. Conclusions We conclude that mesocortical dopamine axon terminals encode the timing of self-initiated actions, shedding light on a crucial aspect of the intricate neural mechanisms governing goal-directed behavior.
Collapse
Affiliation(s)
- Makoto Ohtake
- Medical University of South Carolina, Department of Neuroscience, Charleston, South Carolina, United States
- Yokohama City University, Department of Neurosurgery, Yokohama, Japan
| | - Kenta Abe
- Medical University of South Carolina, Department of Neuroscience, Charleston, South Carolina, United States
| | - Masashi Hasegawa
- Rutgers, The State University of New Jersey, Robert Wood Johnson Medical School, Center for Advanced Biotechnology and Medicine, Department of Neuroscience and Cell Biology, Piscataway, New Jersey, United States
| | - Takahide Itokazu
- Osaka University, Department of Neuro-Medical Science, Osaka, Japan
| | - Vihashini Selvakumar
- Medical University of South Carolina, Department of Neuroscience, Charleston, South Carolina, United States
| | - Ashley Matunis
- Medical University of South Carolina, Department of Neuroscience, Charleston, South Carolina, United States
- College of Charleston, Department of Biology, Charleston, South Carolina, United States
| | - Emma Stacy
- Medical University of South Carolina, Department of Neuroscience, Charleston, South Carolina, United States
- College of Charleston, Department of Biology, Charleston, South Carolina, United States
| | - Emily Froebrich
- Medical University of South Carolina, Department of Neuroscience, Charleston, South Carolina, United States
- College of Charleston, Department of Biology, Charleston, South Carolina, United States
| | - Nathan Huynh
- Kagoshima University, Department of Pharmacology, Kagoshima, Japan
| | - Haesuk Lee
- Kagoshima University, Department of Pharmacology, Kagoshima, Japan
| | - Yuki Kambe
- Kagoshima University, Department of Pharmacology, Kagoshima, Japan
| | - Tetsuya Yamamoto
- Yokohama City University, Department of Neurosurgery, Yokohama, Japan
| | - Tatsuo K. Sato
- Kagoshima University, Department of Pharmacology, Kagoshima, Japan
- FOREST, Japan Science and Technology Agency, Saitama, Japan
| | - Takashi R. Sato
- Medical University of South Carolina, Department of Neuroscience, Charleston, South Carolina, United States
| |
Collapse
|
28
|
Song MR, Lee SW. Rethinking dopamine-guided action sequence learning. Eur J Neurosci 2024; 60:3447-3465. [PMID: 38798086 DOI: 10.1111/ejn.16426] [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: 08/17/2023] [Revised: 04/21/2024] [Accepted: 05/08/2024] [Indexed: 05/29/2024]
Abstract
As opposed to those requiring a single action for reward acquisition, tasks necessitating action sequences demand that animals learn action elements and their sequential order and sustain the behaviour until the sequence is completed. With repeated learning, animals not only exhibit precise execution of these sequences but also demonstrate enhanced smoothness and efficiency. Previous research has demonstrated that midbrain dopamine and its major projection target, the striatum, play crucial roles in these processes. Recent studies have shown that dopamine from the substantia nigra pars compacta (SNc) and the ventral tegmental area (VTA) serve distinct functions in action sequence learning. The distinct contributions of dopamine also depend on the striatal subregions, namely the ventral, dorsomedial and dorsolateral striatum. Here, we have reviewed recent findings on the role of striatal dopamine in action sequence learning, with a focus on recent rodent studies.
Collapse
Affiliation(s)
- Minryung R Song
- Department of Brain and Cognitive Sciences, KAIST, Daejeon, South Korea
| | - Sang Wan Lee
- Department of Brain and Cognitive Sciences, KAIST, Daejeon, South Korea
- Kim Jaechul Graduate School of AI, KAIST, Daejeon, South Korea
- KI for Health Science and Technology, KAIST, Daejeon, South Korea
- Center for Neuroscience-inspired AI, KAIST, Daejeon, South Korea
| |
Collapse
|
29
|
Phillips CD, Hodge AT, Myers CC, Leventhal DK, Burgess CR. Striatal Dopamine Contributions to Skilled Motor Learning. J Neurosci 2024; 44:e0240242024. [PMID: 38806248 PMCID: PMC11211718 DOI: 10.1523/jneurosci.0240-24.2024] [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: 02/04/2024] [Revised: 05/15/2024] [Accepted: 05/21/2024] [Indexed: 05/30/2024] Open
Abstract
Coordinated multijoint limb and digit movements-"manual dexterity"-underlie both specialized skills (e.g., playing the piano) and more mundane tasks (e.g., tying shoelaces). Impairments in dexterous skill cause significant disability, as occurs with motor cortical injury, Parkinson's disease, and a range of other pathologies. Clinical observations, as well as basic investigations, suggest that corticostriatal circuits play a critical role in learning and performing dexterous skills. Furthermore, dopaminergic signaling in these regions is implicated in synaptic plasticity and motor learning. Nonetheless, the role of striatal dopamine signaling in skilled motor learning remains poorly understood. Here, we use fiber photometry paired with a genetically encoded dopamine sensor to investigate striatal dopamine release in both male and female mice as they learn and perform a skilled reaching task. Dopamine rapidly increases during a skilled reach and peaks near pellet consumption. In the dorsolateral striatum, dopamine dynamics are faster than in the dorsomedial and ventral striatum. Across training, as reaching performance improves, dopamine signaling shifts from pellet consumption to cues that predict pellet availability, particularly in medial and ventral areas of the striatum. Furthermore, performance prediction errors are present across the striatum, with reduced dopamine release after an unsuccessful reach. These findings show that dopamine dynamics during skilled motor behaviors change with learning and are differentially regulated across striatal subregions.
Collapse
Affiliation(s)
- Chris D Phillips
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, Michigan 48109
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109
- Department of Neuroscience, University of Texas at Dallas, Richardson, Texas 75080
| | - Alexander T Hodge
- Department of Neurology, University of Michigan, Ann Arbor, Michigan 48109
| | - Courtney C Myers
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, Michigan 48109
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, Michigan 48109
| | - Daniel K Leventhal
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, Michigan 48109
- Department of Neurology, University of Michigan, Ann Arbor, Michigan 48109
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, Michigan 48109
- Parkinson's Disease Foundation Research Center of Excellence, University of Michigan, Ann Arbor, Michigan 48109
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan 48109
- Department of Neurology, VA Ann Arbor Health System, Ann Arbor, Michigan 48109
| | - Christian R Burgess
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, Michigan 48109
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, Michigan 48109
| |
Collapse
|
30
|
Brill-Weil SG, Kramer PF, Yanez A, Clever FH, Zhang R, Khaliq ZM. Presynaptic GABA A receptors control integration of nicotinic input onto dopaminergic axons in the striatum. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.25.600616. [PMID: 39372741 PMCID: PMC11451734 DOI: 10.1101/2024.06.25.600616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Axons of dopaminergic neurons express gamma-aminobutyric acid type-A receptors (GABAARs) and nicotinic acetylcholine receptors (nAChRs) which are both independently positioned to shape striatal dopamine release. Using electrophysiology and calcium imaging, we investigated how interactions between GABAARs and nAChRs influence dopaminergic axon excitability. Direct axonal recordings showed that benzodiazepine application suppresses subthreshold axonal input from cholinergic interneurons (CINs). In imaging experiments, we used the first temporal derivative of presynaptic calcium signals to distinguish between direct- and nAChR-evoked activity in dopaminergic axons. We found that GABAAR antagonism with gabazine selectively enhanced nAChR-evoked axonal signals. Acetylcholine release was unchanged in gabazine suggesting that GABAARs located on dopaminergic axons, but not CINs, mediated this enhancement. Unexpectedly, we found that a widely used GABAAR antagonist, picrotoxin, inhibits axonal nAChRs and should be used cautiously for striatal circuit analysis. Overall, we demonstrate that GABAARs on dopaminergic axons regulate integration of nicotinic input to shape presynaptic excitability.
Collapse
Affiliation(s)
- Samuel G. Brill-Weil
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Paul F. Kramer
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Anthony Yanez
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Faye H. Clever
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Renshu Zhang
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Zayd M. Khaliq
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| |
Collapse
|
31
|
Zhang L, Sun Y, Wang J, Zhang M, Wang Q, Xie B, Yu F, Wen D, Ma C. Dopaminergic dominance in the ventral medial hypothalamus: A pivotal regulator for methamphetamine-induced pathological aggression. Prog Neuropsychopharmacol Biol Psychiatry 2024; 132:110971. [PMID: 38365104 DOI: 10.1016/j.pnpbp.2024.110971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/05/2024] [Accepted: 02/13/2024] [Indexed: 02/18/2024]
Abstract
Methamphetamine (METH) abuse is associated with a spectrum of behavioral consequences, among which heightened aggression presents a significant challenge. However, the causal role of METH's impact in aggression and its target circuit mechanisms remains largely unknown. We established an acute METH exposure-aggression mouse model to investigate the role of ventral tegmental area (VTA) dopaminergic neurons and ventral medial hypothalamus VMH glutamatergic neuron. Our findings revealed that METH-induced VTA dopamine excitability activates the ventromedial hypothalamus (VMH) glutamatergic neurons, contributing to pathological aggression. Notably, we uncovered a dopaminergic transmission within the VTA-VMH circuit that exclusively functioned under METH influence. This dopaminergic pathway emerged as a potential key player in enabling dopamine-related pathological aggression, with heightened dopaminergic excitability implicated in various psychiatric symptoms. Also, the modulatory function of this pathway opens new possibilities for targeted therapeutic strategies for intervention to improve treatment in METH abuse and may have broader implications for addressing pathological aggression syndromes.
Collapse
Affiliation(s)
- Ludi Zhang
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, 050017 Shijiazhuang, Hebei, PR China; Identification Center of Forensic Medicine, Hebei Medical University, 050017 Shijiazhuang, Hebei, PR China; Key Laboratory of Neural and Vascular Biology, Ministry of Education, 050017 Shijiazhuang, Hebei, PR China; Hebei Medical University Postdoctoral Research Station, 050017, Shijiazhuang, Hebei, PR China
| | - Yufei Sun
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, 050017 Shijiazhuang, Hebei, PR China
| | - Jian Wang
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, 050017 Shijiazhuang, Hebei, PR China; Identification Center of Forensic Medicine, Hebei Medical University, 050017 Shijiazhuang, Hebei, PR China
| | - Minglong Zhang
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, 050017 Shijiazhuang, Hebei, PR China
| | - Qingwu Wang
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, 050017 Shijiazhuang, Hebei, PR China; Identification Center of Forensic Medicine, Hebei Medical University, 050017 Shijiazhuang, Hebei, PR China
| | - Bing Xie
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, 050017 Shijiazhuang, Hebei, PR China; Identification Center of Forensic Medicine, Hebei Medical University, 050017 Shijiazhuang, Hebei, PR China
| | - Feng Yu
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, 050017 Shijiazhuang, Hebei, PR China; Identification Center of Forensic Medicine, Hebei Medical University, 050017 Shijiazhuang, Hebei, PR China
| | - Di Wen
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, 050017 Shijiazhuang, Hebei, PR China; Identification Center of Forensic Medicine, Hebei Medical University, 050017 Shijiazhuang, Hebei, PR China; Key Laboratory of Neural and Vascular Biology, Ministry of Education, 050017 Shijiazhuang, Hebei, PR China.
| | - Chunling Ma
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, 050017 Shijiazhuang, Hebei, PR China; Identification Center of Forensic Medicine, Hebei Medical University, 050017 Shijiazhuang, Hebei, PR China; Key Laboratory of Neural and Vascular Biology, Ministry of Education, 050017 Shijiazhuang, Hebei, PR China.
| |
Collapse
|
32
|
Ryczko D. The Mesencephalic Locomotor Region: Multiple Cell Types, Multiple Behavioral Roles, and Multiple Implications for Disease. Neuroscientist 2024; 30:347-366. [PMID: 36575956 PMCID: PMC11107129 DOI: 10.1177/10738584221139136] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The mesencephalic locomotor region (MLR) controls locomotion in vertebrates. In humans with Parkinson disease, locomotor deficits are increasingly associated with decreased activity in the MLR. This brainstem region, commonly considered to include the cuneiform and pedunculopontine nuclei, has been explored as a target for deep brain stimulation to improve locomotor function, but the results are variable, from modest to promising. However, the MLR is a heterogeneous structure, and identification of the best cell type to target is only beginning. Here, I review the studies that uncovered the role of genetically defined MLR cell types, and I highlight the cells whose activation improves locomotor function in animal models of Parkinson disease. The promising cell types to activate comprise some glutamatergic neurons in the cuneiform and caudal pedunculopontine nuclei, as well as some cholinergic neurons of the pedunculopontine nucleus. Activation of MLR GABAergic neurons should be avoided, since they stop locomotion or evoke bouts flanked with numerous stops. MLR is also considered a potential target in spinal cord injury, supranuclear palsy, primary progressive freezing of gait, or stroke. Better targeting of the MLR cell types should be achieved through optimized deep brain stimulation protocols, pharmacotherapy, or the development of optogenetics for human use.
Collapse
Affiliation(s)
- Dimitri Ryczko
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Canada
- Centre de recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Canada
- Neurosciences Sherbrooke, Sherbrooke, Canada
- Institut de Pharmacologie de Sherbrooke, Sherbrooke, Canada
| |
Collapse
|
33
|
Li X, Du Y, Huang JF, Li WW, Song W, Fan RN, Zhou H, Jiang T, Lu CG, Guan Z, Wang XF, Gong H, Li XN, Li A, Fu L, Sun YG. Link Brain-Wide Projectome to Neuronal Dynamics in the Mouse Brain. Neurosci Bull 2024:10.1007/s12264-024-01232-z. [PMID: 38819707 DOI: 10.1007/s12264-024-01232-z] [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: 01/17/2024] [Accepted: 01/28/2024] [Indexed: 06/01/2024] Open
Abstract
Knowledge about the neuronal dynamics and the projectome are both essential for understanding how the neuronal network functions in concert. However, it remains challenging to obtain the neural activity and the brain-wide projectome for the same neurons, especially for neurons in subcortical brain regions. Here, by combining in vivo microscopy and high-definition fluorescence micro-optical sectioning tomography, we have developed strategies for mapping the brain-wide projectome of functionally relevant neurons in the somatosensory cortex, the dorsal hippocampus, and the substantia nigra pars compacta. More importantly, we also developed a strategy to achieve acquiring the neural dynamic and brain-wide projectome of the molecularly defined neuronal subtype. The strategies developed in this study solved the essential problem of linking brain-wide projectome to neuronal dynamics for neurons in subcortical structures and provided valuable approaches for understanding how the brain is functionally organized via intricate connectivity patterns.
Collapse
Affiliation(s)
- Xiang Li
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
- MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
- Advanced Biomedical Imaging Facility, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
| | - Yun Du
- Institute of Neuroscience, Key Laboratory of Brain Coginition and Brain-inspired Intelligence Technology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jiang-Feng Huang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
- MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
- Advanced Biomedical Imaging Facility, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
| | - Wen-Wei Li
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
- MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
| | - Wei Song
- Institute of Neuroscience, Key Laboratory of Brain Coginition and Brain-inspired Intelligence Technology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ruo-Nan Fan
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
- MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
- Advanced Biomedical Imaging Facility, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
| | - Hua Zhou
- Institute of Neuroscience, Key Laboratory of Brain Coginition and Brain-inspired Intelligence Technology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Tao Jiang
- HUST-Suzhou Institute for Brainsmatics, JITRI Institute for Brainsmatics, Suzhou, 215123, China
| | - Chang-Geng Lu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
- MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
- Advanced Biomedical Imaging Facility, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
| | - Zhuang Guan
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
- MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
| | - Xiao-Fei Wang
- Institute of Neuroscience, Key Laboratory of Brain Coginition and Brain-inspired Intelligence Technology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Hui Gong
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
- MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
- HUST-Suzhou Institute for Brainsmatics, JITRI Institute for Brainsmatics, Suzhou, 215123, China
| | - Xiang-Ning Li
- HUST-Suzhou Institute for Brainsmatics, JITRI Institute for Brainsmatics, Suzhou, 215123, China
- School of Biomedical Engineering, Hainan University, Haikou, 570228, China
| | - Anan Li
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
- MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
- HUST-Suzhou Institute for Brainsmatics, JITRI Institute for Brainsmatics, Suzhou, 215123, China
| | - Ling Fu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China.
- MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China.
- Advanced Biomedical Imaging Facility, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China.
- School of Biomedical Engineering, Hainan University, Haikou, 570228, China.
- School of Physics and Optoelectronics Engineering, Hainan University, Haikou, 570228, Hainan, China.
| | - Yan-Gang Sun
- Institute of Neuroscience, Key Laboratory of Brain Coginition and Brain-inspired Intelligence Technology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
34
|
Callahan JW, Morales JC, Atherton JF, Wang D, Kostic S, Bevan MD. Movement-related increases in subthalamic activity optimize locomotion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.07.570617. [PMID: 38105984 PMCID: PMC10723456 DOI: 10.1101/2023.12.07.570617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
The subthalamic nucleus (STN) is traditionally thought to restrict movement. Lesion or prolonged STN inhibition increases movement vigor and propensity, while ontogenetic excitation typically has opposing effects. Subthalamic and motor activity are also inversely correlated in movement disorders. However, most STN neurons exhibit movement-related increases in firing. To address this paradox, STN activity was recorded and manipulated in head-fixed mice at rest and during self-initiated treadmill locomotion. The majority of STN neurons (type 1) exhibited locomotion-dependent increases in activity, with half encoding the locomotor cycle. A minority of neurons exhibited dips in activity or were uncorrelated with movement. Brief optogenetic inhibition of the dorsolateral STN (where type 1 neurons are concentrated) slowed and prematurely terminated locomotion. In Q175 Huntington's disease mice abnormally brief, low-velocity locomotion was specifically associated with type 1 hyperactivity. Together these data argue that movement-related increases in STN activity contribute to optimal locomotor performance.
Collapse
|
35
|
Juárez Tello A, van der Zouwen CI, Dejas L, Duque-Yate J, Boutin J, Medina-Ortiz K, Suresh JS, Swiegers J, Sarret P, Ryczko D. Dopamine-sensitive neurons in the mesencephalic locomotor region control locomotion initiation, stop, and turns. Cell Rep 2024; 43:114187. [PMID: 38722743 PMCID: PMC11157412 DOI: 10.1016/j.celrep.2024.114187] [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: 10/20/2023] [Revised: 03/31/2024] [Accepted: 04/17/2024] [Indexed: 06/01/2024] Open
Abstract
The locomotor role of dopaminergic neurons is traditionally attributed to their ascending projections to the basal ganglia, which project to the mesencephalic locomotor region (MLR). In addition, descending dopaminergic projections to the MLR are present from basal vertebrates to mammals. However, the neurons targeted in the MLR and their behavioral role are unknown in mammals. Here, we identify genetically defined MLR cells that express D1 or D2 receptors and control different motor behaviors in mice. In the cuneiform nucleus, D1-expressing neurons promote locomotion, while D2-expressing neurons stop locomotion. In the pedunculopontine nucleus, D1-expressing neurons promote locomotion, while D2-expressing neurons evoke ipsilateral turns. Using RNAscope, we show that MLR dopamine-sensitive neurons comprise a combination of glutamatergic, GABAergic, and cholinergic neurons, suggesting that different neurotransmitter-based cell types work together to control distinct behavioral modules. Altogether, our study uncovers behaviorally relevant cell types in the mammalian MLR based on the expression of dopaminergic receptors.
Collapse
Affiliation(s)
- Andrea Juárez Tello
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Cornelis Immanuel van der Zouwen
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Léonie Dejas
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Juan Duque-Yate
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Joël Boutin
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Katherine Medina-Ortiz
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Jacinthlyn Sylvia Suresh
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Jordan Swiegers
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Philippe Sarret
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada; Centre de recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada; Neurosciences Sherbrooke, Institut de Pharmacologie de Sherbrooke, Sherbrooke, QC, Canada
| | - Dimitri Ryczko
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada; Centre de recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada; Neurosciences Sherbrooke, Institut de Pharmacologie de Sherbrooke, Sherbrooke, QC, Canada.
| |
Collapse
|
36
|
Forderhase AG, Ligons LA, Norwood E, McCarty GS, Sombers LA. Optimized Fabrication of Carbon-Fiber Microbiosensors for Codetection of Glucose and Dopamine in Brain Tissue. ACS Sens 2024; 9:2662-2672. [PMID: 38689483 DOI: 10.1021/acssensors.4c00527] [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] [Indexed: 05/02/2024]
Abstract
Dopamine (DA) signaling is critically important in striatal function, and this metabolically demanding process is fueled largely by glucose. However, DA and glucose are typically studied independently and, as such, the precise relationship between DA release and glucose availability remains unclear. Fast-scan cyclic voltammetry (FSCV) is commonly coupled with carbon-fiber microelectrodes to study DA transients. These microelectrodes can be modified with glucose oxidase (GOx) to generate microbiosensors capable of simultaneously quantifying real-time and physiologically relevant fluctuations of glucose, a nonelectrochemically active substrate, and DA, which is readily oxidized and reduced at the electrode surface. A chitosan hydrogel can be electrodeposited to entrap the oxidase enzyme on the sensor surface for stable, sensitive, and selective codetection of glucose and DA using FSCV. This strategy can also be used to entrap lactate oxidase on the carbon-fiber surface for codetection of lactate and DA. However, these custom probes are individually fabricated by hand, and performance is variable. This study characterizes the physical nature of the hydrogel and its effects on the acquired electrochemical data in the detection of glucose (2.6 mM) and DA (1 μM). The results demonstrate that the electrodeposition of the hydrogel membrane is improved using a linear potential sweep rather than a direct step to the target potential. Electrochemical impedance spectroscopy data relate information on the physical nature of the electrode/solution interface to the electrochemical performance of bare and enzyme-modified carbon-fiber microelectrodes. The electrodeposition waveform and scan rate were characterized for optimal membrane formation and performance. Finally, codetection of both DA/glucose and DA/lactate was demonstrated in intact rat striatum using probes fabricated according to the optimized protocol. Overall, this work improves the reliable fabrication of carbon-fiber microbiosensors for codetection of DA and important energetic substrates that are locally delivered to the recording site to meet metabolic demand.
Collapse
Affiliation(s)
- Alexandra G Forderhase
- Department of Chemistry, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Lailah A Ligons
- Department of Chemistry, North Carolina State University, Raleigh, North Carolina 27695, United States
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, North Carolina 27695, United States
| | - Emilie Norwood
- Department of Chemistry, North Carolina State University, Raleigh, North Carolina 27695, United States
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, North Carolina 27695, United States
| | - Gregory S McCarty
- Department of Chemistry, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Leslie A Sombers
- Department of Chemistry, North Carolina State University, Raleigh, North Carolina 27695, United States
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, North Carolina 27695, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27695, United States
| |
Collapse
|
37
|
Bornhoft KN, Prohofsky J, O'Neal TJ, Wolff AR, Saunders BT. Valence ambiguity dynamically shapes striatal dopamine heterogeneity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.17.594692. [PMID: 38798567 PMCID: PMC11118546 DOI: 10.1101/2024.05.17.594692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Adaptive decision making relies on dynamic updating of learned associations where environmental cues come to predict positive and negatively valenced stimuli, such as food or threat. Flexible cue-guided behaviors depend on a network of brain systems, including dopamine signaling in the striatum, which is critical for learning and maintenance of conditioned behaviors. Critically, it remains unclear how dopamine signaling encodes multi-valent, dynamic learning contexts, where positive and negative associations must be rapidly disambiguated. To understand this, we employed a Pavlovian discrimination paradigm, where cues predicting positive and negative outcomes were intermingled during conditioning sessions, and their meaning was serially reversed across training. We found that rats readily distinguished these cues, and updated their behavior rapidly upon valence reversal. Using fiber photometry, we recorded dopamine signaling in three major striatal subregions -,the dorsolateral striatum (DLS), the nucleus accumbens core, and the nucleus accumbens medial shell - and found heterogeneous responses to positive and negative conditioned cues and their predicted outcomes. Valence ambiguity introduced by cue reversal reshaped striatal dopamine on different timelines: nucleus accumbens core and shell signals updated more readily than those in the DLS. Together, these results suggest that striatal dopamine flexibly encodes multi-valent learning contexts, and these signals are dynamically modulated by changing contingencies to resolve ambiguity about the meaning of environmental cues.
Collapse
|
38
|
Hueske E, Stine C, Yoshida T, Crittenden JR, Gupta A, Johnson JC, Achanta AS, Loftus J, Mahar A, Hul D, Azocar J, Gray RJ, Bruchas MR, Graybiel AM. Developmental and adult striatal patterning of nociceptin ligand marks striosomal population with direct dopamine projections. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.15.594426. [PMID: 38798373 PMCID: PMC11118414 DOI: 10.1101/2024.05.15.594426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Circuit influences on the midbrain dopamine system are crucial to adaptive behavior and cognition. Recent developments in the study of neuropeptide systems have enabled high-resolution investigations of the intersection of neuromodulatory signals with basal ganglia circuitry, identifying the nociceptin/orphanin FQ (N/OFQ) endogenous opioid peptide system as a prospective regulator of striatal dopamine signaling. Using a prepronociceptin-Cre reporter mouse line, we characterized highly selective striosomal patterning of Pnoc mRNA expression in mouse dorsal striatum, reflecting early developmental expression of Pnoc . In the ventral striatum, Pnoc expression was was clustered across the nucleus accumbens core and medial shell, including in adult striatum. We found that Pnoc tdTomato reporter cells largely comprise a population of dopamine receptor D1 ( Drd1 ) expressing medium spiny projection neurons localized in dorsal striosomes, known to be unique among striatal projections neurons for their direct innervation of midbrain dopamine neurons. These findings provide new understanding of the intersection of the N/OFQ system among basal ganglia circuits with particular implications for developmental regulation or wiring of striatal-nigral circuits.
Collapse
|
39
|
Taniguchi J, Melani R, Chantranupong L, Wen MJ, Mohebi A, Berke JD, Sabatini BL, Tritsch NX. Comment on 'Accumbens cholinergic interneurons dynamically promote dopamine release and enable motivation'. eLife 2024; 13:e95694. [PMID: 38748470 PMCID: PMC11095934 DOI: 10.7554/elife.95694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 05/02/2024] [Indexed: 05/18/2024] Open
Abstract
Acetylcholine is widely believed to modulate the release of dopamine in the striatum of mammals. Experiments in brain slices clearly show that synchronous activation of striatal cholinergic interneurons is sufficient to drive dopamine release via axo-axonal stimulation of nicotinic acetylcholine receptors. However, evidence for this mechanism in vivo has been less forthcoming. Mohebi, Collins and Berke recently reported that, in awake behaving rats, optogenetic activation of striatal cholinergic interneurons with blue light readily evokes dopamine release measured with the red fluorescent sensor RdLight1 (Mohebi et al., 2023). Here, we show that blue light alone alters the fluorescent properties of RdLight1 in a manner that may be misconstrued as phasic dopamine release, and that this artefactual photoactivation can account for the effects attributed to cholinergic interneurons. Our findings indicate that measurements of dopamine using the red-shifted fluorescent sensor RdLight1 should be interpreted with caution when combined with optogenetics. In light of this and other publications that did not observe large acetylcholine-evoked dopamine transients in vivo, the conditions under which such release occurs in behaving animals remain unknown.
Collapse
Affiliation(s)
- James Taniguchi
- Neuroscience Institute and Fresco Institute for Parkinson's and Movement Disorders, University Grossman School of MedicineNew YorkUnited States
| | - Riccardo Melani
- Neuroscience Institute and Fresco Institute for Parkinson's and Movement Disorders, University Grossman School of MedicineNew YorkUnited States
| | - Lynne Chantranupong
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical SchoolBostonUnited States
| | - Michelle J Wen
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical SchoolBostonUnited States
| | - Ali Mohebi
- Department of Neurology, University of California, San FranciscoSan FranciscoUnited States
| | - Joshua D Berke
- Department of Neurology, University of California, San FranciscoSan FranciscoUnited States
| | - Bernardo L Sabatini
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical SchoolBostonUnited States
| | - Nicolas X Tritsch
- Neuroscience Institute and Fresco Institute for Parkinson's and Movement Disorders, University Grossman School of MedicineNew YorkUnited States
| |
Collapse
|
40
|
Abe K, Kambe Y, Majima K, Hu Z, Ohtake M, Momennezhad A, Izumi H, Tanaka T, Matunis A, Stacy E, Itokazu T, Sato TR, Sato T. Functional diversity of dopamine axons in prefrontal cortex during classical conditioning. eLife 2024; 12:RP91136. [PMID: 38747563 PMCID: PMC11095940 DOI: 10.7554/elife.91136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024] Open
Abstract
Midbrain dopamine neurons impact neural processing in the prefrontal cortex (PFC) through mesocortical projections. However, the signals conveyed by dopamine projections to the PFC remain unclear, particularly at the single-axon level. Here, we investigated dopaminergic axonal activity in the medial PFC (mPFC) during reward and aversive processing. By optimizing microprism-mediated two-photon calcium imaging of dopamine axon terminals, we found diverse activity in dopamine axons responsive to both reward and aversive stimuli. Some axons exhibited a preference for reward, while others favored aversive stimuli, and there was a strong bias for the latter at the population level. Long-term longitudinal imaging revealed that the preference was maintained in reward- and aversive-preferring axons throughout classical conditioning in which rewarding and aversive stimuli were paired with preceding auditory cues. However, as mice learned to discriminate reward or aversive cues, a cue activity preference gradually developed only in aversive-preferring axons. We inferred the trial-by-trial cue discrimination based on machine learning using anticipatory licking or facial expressions, and found that successful discrimination was accompanied by sharper selectivity for the aversive cue in aversive-preferring axons. Our findings indicate that a group of mesocortical dopamine axons encodes aversive-related signals, which are modulated by both classical conditioning across days and trial-by-trial discrimination within a day.
Collapse
Affiliation(s)
- Kenta Abe
- Department of Neuroscience, Medical University of South CarolinaCharlestonUnited States
| | - Yuki Kambe
- Department of Pharmacology, Kagoshima UniversityKagoshimaJapan
| | - Kei Majima
- Institute for Quantum Life Science, National Institutes for Quantum Science and TechnologyChibaJapan
- Japan Science and Technology PRESTOSaitamaJapan
| | - Zijing Hu
- Department of Physiology, Monash UniversityClaytonAustralia
- Neuroscience Program, Biomedicine Discovery Institute, Monash UniversityClaytonAustralia
| | - Makoto Ohtake
- Department of Neuroscience, Medical University of South CarolinaCharlestonUnited States
| | - Ali Momennezhad
- Department of Pharmacology, Kagoshima UniversityKagoshimaJapan
| | - Hideki Izumi
- Faculty of Data Science, Shiga UniversityShigaJapan
| | | | - Ashley Matunis
- Department of Neuroscience, Medical University of South CarolinaCharlestonUnited States
- Department of Biology, College of CharlestonCharlestonUnited States
- Department of Neuro-Medical Science, Osaka UniversityOsakaJapan
| | - Emma Stacy
- Department of Neuroscience, Medical University of South CarolinaCharlestonUnited States
- Department of Biology, College of CharlestonCharlestonUnited States
| | | | - Takashi R Sato
- Department of Neuroscience, Medical University of South CarolinaCharlestonUnited States
| | - Tatsuo Sato
- Department of Pharmacology, Kagoshima UniversityKagoshimaJapan
- Japan Science and Technology PRESTOSaitamaJapan
- Department of Physiology, Monash UniversityClaytonAustralia
- Neuroscience Program, Biomedicine Discovery Institute, Monash UniversityClaytonAustralia
- Japan Science and Technology FORESTSaitamaJapan
| |
Collapse
|
41
|
Hart G, Burton TJ, Balleine BW. What Role Does Striatal Dopamine Play in Goal-directed Action? Neuroscience 2024; 546:20-32. [PMID: 38521480 DOI: 10.1016/j.neuroscience.2024.03.020] [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: 01/23/2023] [Revised: 02/15/2024] [Accepted: 03/18/2024] [Indexed: 03/25/2024]
Abstract
Evidence suggests that dopamine activity provides a US-related prediction error for Pavlovian conditioning and the reinforcement signal supporting the acquisition of habits. However, its role in goal-directed action is less clear. There are currently few studies that have assessed dopamine release as animals acquire and perform self-paced instrumental actions. Here we briefly review the literature documenting the psychological, behavioral and neural bases of goal-directed actions in rats and mice, before turning to describe recent studies investigating the role of dopamine in instrumental learning and performance. Plasticity in dorsomedial striatum, a central node in the network supporting goal-directed action, clearly requires dopamine release, the timing of which, relative to cortical and thalamic inputs, determines the degree and form of that plasticity. Beyond this, bilateral release appears to reflect reward prediction errors as animals experience the consequences of an action. Such signals feedforward to update the value of the specific action associated with that outcome during subsequent performance, with dopamine release at the time of action reflecting the updated predicted action value. More recently, evidence has also emerged for a hemispherically lateralised signal associated with the action; dopamine release is greater in the hemisphere contralateral to the spatial target of the action. This effect emerges over the course of acquisition and appears to reflect the strength of the action-outcome association. Thus, during goal-directed action, dopamine release signals the action, the outcome and their association to shape the learning and performance processes necessary to support this form of behavioral control.
Collapse
Affiliation(s)
- Genevra Hart
- Decision Neuroscience Lab, UNSW Sydney, Australia
| | | | | |
Collapse
|
42
|
Jang HJ, Ward RM, Golden CEM, Constantinople CM. Acetylcholine demixes heterogeneous dopamine signals for learning and moving. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.03.592444. [PMID: 38746300 PMCID: PMC11092744 DOI: 10.1101/2024.05.03.592444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Midbrain dopamine neurons promote reinforcement learning and movement vigor. A major outstanding question is how dopamine-recipient neurons in the striatum parse these heterogeneous signals. Here we characterized dopamine and acetylcholine release in the dorsomedial striatum (DMS) of rats performing a decision-making task. We found that dopamine acted as a reward prediction error (RPE), modulating behavior and DMS spiking on subsequent trials when coincident with pauses in cholinergic release. In contrast, at task events that elicited coincident bursts of acetylcholine and dopamine, dopamine preceded contralateral movements and predicted movement vigor without inducing plastic changes in DMS firing rates. Our findings provide a circuit-level mechanism by which cholinergic modulation allows the same dopamine signals to be used for either movement or learning depending on instantaneous behavioral context.
Collapse
|
43
|
Avvisati R, Kaufmann AK, Young CJ, Portlock GE, Cancemi S, Costa RP, Magill PJ, Dodson PD. Distributional coding of associative learning in discrete populations of midbrain dopamine neurons. Cell Rep 2024; 43:114080. [PMID: 38581677 PMCID: PMC7616095 DOI: 10.1016/j.celrep.2024.114080] [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: 10/31/2023] [Revised: 02/12/2024] [Accepted: 03/24/2024] [Indexed: 04/08/2024] Open
Abstract
Midbrain dopamine neurons are thought to play key roles in learning by conveying the difference between expected and actual outcomes. Recent evidence suggests diversity in dopamine signaling, yet it remains poorly understood how heterogeneous signals might be organized to facilitate the role of downstream circuits mediating distinct aspects of behavior. Here, we investigated the organizational logic of dopaminergic signaling by recording and labeling individual midbrain dopamine neurons during associative behavior. Our findings show that reward information and behavioral parameters are not only heterogeneously encoded but also differentially distributed across populations of dopamine neurons. Retrograde tracing and fiber photometry suggest that populations of dopamine neurons projecting to different striatal regions convey distinct signals. These data, supported by computational modeling, indicate that such distributional coding can maximize dynamic range and tailor dopamine signals to facilitate specialized roles of different striatal regions.
Collapse
Affiliation(s)
- Riccardo Avvisati
- School of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol BS8 1TD, UK; Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK
| | - Anna-Kristin Kaufmann
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK
| | - Callum J Young
- School of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol BS8 1TD, UK; Computational Neuroscience Unit, Department of Computer Science, SCEEM, Faculty of Engineering, University of Bristol, Bristol BS8 1UB, UK
| | - Gabriella E Portlock
- School of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol BS8 1TD, UK
| | - Sophie Cancemi
- School of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol BS8 1TD, UK
| | - Rui Ponte Costa
- Computational Neuroscience Unit, Department of Computer Science, SCEEM, Faculty of Engineering, University of Bristol, Bristol BS8 1UB, UK
| | - Peter J Magill
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK
| | - Paul D Dodson
- School of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol BS8 1TD, UK; Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK.
| |
Collapse
|
44
|
Mohebi A, Wei W, Pelattini L, Kim K, Berke JD. Dopamine transients follow a striatal gradient of reward time horizons. Nat Neurosci 2024; 27:737-746. [PMID: 38321294 PMCID: PMC11001583 DOI: 10.1038/s41593-023-01566-3] [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: 05/12/2022] [Accepted: 12/21/2023] [Indexed: 02/08/2024]
Abstract
Animals make predictions to guide their behavior and update those predictions through experience. Transient increases in dopamine (DA) are thought to be critical signals for updating predictions. However, it is unclear how this mechanism handles a wide range of behavioral timescales-from seconds or less (for example, if singing a song) to potentially hours or more (for example, if hunting for food). Here we report that DA transients in distinct rat striatal subregions convey prediction errors based on distinct time horizons. DA dynamics systematically accelerated from ventral to dorsomedial to dorsolateral striatum, in the tempo of spontaneous fluctuations, the temporal integration of prior rewards and the discounting of future rewards. This spectrum of timescales for evaluative computations can help achieve efficient learning and adaptive motivation for a broad range of behaviors.
Collapse
Affiliation(s)
- Ali Mohebi
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Wei Wei
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Lilian Pelattini
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Kyoungjun Kim
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Joshua D Berke
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA.
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA, USA.
- Neuroscience Graduate Program, University of California San Francisco, San Francisco, CA, USA.
- Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, CA, USA.
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
45
|
Seiler JL, Zhuang X, Nelson AB, Lerner TN. Dopamine across timescales and cell types: Relevance for phenotypes in Parkinson's disease progression. Exp Neurol 2024; 374:114693. [PMID: 38242300 DOI: 10.1016/j.expneurol.2024.114693] [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: 09/12/2023] [Revised: 01/09/2024] [Accepted: 01/15/2024] [Indexed: 01/21/2024]
Abstract
Dopamine neurons in the substantia nigra pars compacta (SNc) synthesize and release dopamine, a critical neurotransmitter for movement and learning. SNc dopamine neurons degenerate in Parkinson's Disease (PD), causing a host of motor and non-motor symptoms. Here, we review recent conceptual advances in our basic understanding of the dopamine system - including our rapidly advancing knowledge of dopamine neuron heterogeneity - with special attention to their importance for understanding PD. In PD patients, dopamine neuron degeneration progresses from lateral SNc to medial SNc, suggesting clinically relevant heterogeneity in dopamine neurons. With technical advances in dopamine system interrogation, we can understand the relevance of this heterogeneity for PD progression and harness it to develop new treatments.
Collapse
Affiliation(s)
- Jillian L Seiler
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Xiaowen Zhuang
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA; Weill Institute for Neuroscience, University of California San Francisco, San Francisco, CA, USA; Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, CA, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Alexandra B Nelson
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA; Neuroscience Graduate Program, University of California San Francisco, San Francisco, CA, USA; Weill Institute for Neuroscience, University of California San Francisco, San Francisco, CA, USA; Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, CA, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| | - Talia N Lerner
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Northwestern University Interdepartmental Neuroscience Program (NUIN), Evanston, IL, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
46
|
Zhuo Y, Luo B, Yi X, Dong H, Miao X, Wan J, Williams JT, Campbell MG, Cai R, Qian T, Li F, Weber SJ, Wang L, Li B, Wei Y, Li G, Wang H, Zheng Y, Zhao Y, Wolf ME, Zhu Y, Watabe-Uchida M, Li Y. Improved green and red GRAB sensors for monitoring dopaminergic activity in vivo. Nat Methods 2024; 21:680-691. [PMID: 38036855 PMCID: PMC11009088 DOI: 10.1038/s41592-023-02100-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 10/23/2023] [Indexed: 12/02/2023]
Abstract
Dopamine (DA) plays multiple roles in a wide range of physiological and pathological processes via a large network of dopaminergic projections. To dissect the spatiotemporal dynamics of DA release in both dense and sparsely innervated brain regions, we developed a series of green and red fluorescent G-protein-coupled receptor activation-based DA (GRABDA) sensors using a variety of DA receptor subtypes. These sensors have high sensitivity, selectivity and signal-to-noise ratio with subsecond response kinetics and the ability to detect a wide range of DA concentrations. We then used these sensors in mice to measure both optogenetically evoked and behaviorally relevant DA release while measuring neurochemical signaling in the nucleus accumbens, amygdala and cortex. Using these sensors, we also detected spatially resolved heterogeneous cortical DA release in mice performing various behaviors. These next-generation GRABDA sensors provide a robust set of tools for imaging dopaminergic activity under a variety of physiological and pathological conditions.
Collapse
Affiliation(s)
- Yizhou Zhuo
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Bin Luo
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
- Peking-Tsinghua Center for Life Sciences, New Cornerstone Science Laboratory, Academy for Advanced Interdisciplinary Studies, Beijing, China
| | - Xinyang Yi
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Hui Dong
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
- Peking-Tsinghua Center for Life Sciences, New Cornerstone Science Laboratory, Academy for Advanced Interdisciplinary Studies, Beijing, China
| | - Xiaolei Miao
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
- Department of Anesthesiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Jinxia Wan
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
- Peking-Tsinghua Center for Life Sciences, New Cornerstone Science Laboratory, Academy for Advanced Interdisciplinary Studies, Beijing, China
| | - John T Williams
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Malcolm G Campbell
- Center for Brain Science, Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Ruyi Cai
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Tongrui Qian
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Fengling Li
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Sophia J Weber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - Lei Wang
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, Peking University, Beijing, China
| | - Bozhi Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
- Department of Neurology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yu Wei
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Guochuan Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Huan Wang
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Yu Zheng
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Yulin Zhao
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Marina E Wolf
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - Yingjie Zhu
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Mitsuko Watabe-Uchida
- Center for Brain Science, Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China.
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, New Cornerstone Science Laboratory, Academy for Advanced Interdisciplinary Studies, Beijing, China.
- Chinese Institute for Brain Research, Beijing, China.
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China.
- National Biomedical Imaging Center, Peking University, Beijing, China.
| |
Collapse
|
47
|
Engel L, Wolff AR, Blake M, Collins VL, Sinha S, Saunders BT. Dopamine neurons drive spatiotemporally heterogeneous striatal dopamine signals during learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.01.547331. [PMID: 38585717 PMCID: PMC10996462 DOI: 10.1101/2023.07.01.547331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Environmental cues, through Pavlovian learning, become conditioned stimuli that invigorate and guide animals toward acquisition of rewards. Dopamine neurons in the ventral tegmental area (VTA) and substantia nigra (SNC) are crucial for this process. Dopamine neurons are embedded in a reciprocally connected network with their striatal targets, the functional organization of which remains poorly understood. Here, we investigated how learning during optogenetic Pavlovian cue conditioning of VTA or SNC dopamine neurons directs cue-evoked behavior and shapes subregion-specific striatal dopamine dynamics. We used a fluorescent dopamine biosensor to monitor dopamine in the nucleus accumbens (NAc) core and shell, dorsomedial striatum (DMS), and dorsolateral striatum (DLS). We demonstrate spatially heterogeneous, learning-dependent dopamine changes across striatal regions. While VTA stimulation evoked robust dopamine release in NAc core, shell, and DMS, cues predictive of this activation preferentially recruited dopamine release in NAc core, starting early in training, and DMS, late in training. Corresponding negative prediction error signals, reflecting a violation in the expectation of dopamine neuron activation, only emerged in the NAc core and DMS, and not the shell. Despite development of vigorous movement late in training, conditioned dopamine signals did not similarly emerge in the DLS, even during Pavlovian conditioning with SNC dopamine neuron activation, which elicited robust DLS dopamine release. Together, our studies show broad dissociation in the fundamental prediction and reward-related information generated by different dopamine neuron populations and signaled by dopamine across the striatum. Further, they offer new insight into how larger-scale plasticity across the striatal network emerges during Pavlovian learning to coordinate behavior.
Collapse
Affiliation(s)
- Liv Engel
- Department of Neuroscience, University of Minnesota
- Medical Discovery Team on Addiction, University of Minnesota
- Current Address: Department of Psychology, University of Toronto
| | - Amy R Wolff
- Department of Neuroscience, University of Minnesota
- Medical Discovery Team on Addiction, University of Minnesota
| | - Madelyn Blake
- Department of Neuroscience, University of Minnesota
- Medical Discovery Team on Addiction, University of Minnesota
| | - Val L Collins
- Department of Neuroscience, University of Minnesota
- Medical Discovery Team on Addiction, University of Minnesota
| | | | - Benjamin T Saunders
- Department of Neuroscience, University of Minnesota
- Medical Discovery Team on Addiction, University of Minnesota
| |
Collapse
|
48
|
Hou G, Hao M, Duan J, Han MH. The Formation and Function of the VTA Dopamine System. Int J Mol Sci 2024; 25:3875. [PMID: 38612683 PMCID: PMC11011984 DOI: 10.3390/ijms25073875] [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: 10/20/2023] [Revised: 03/12/2024] [Accepted: 03/14/2024] [Indexed: 04/14/2024] Open
Abstract
The midbrain dopamine system is a sophisticated hub that integrates diverse inputs to control multiple physiological functions, including locomotion, motivation, cognition, reward, as well as maternal and reproductive behaviors. Dopamine is a neurotransmitter that binds to G-protein-coupled receptors. Dopamine also works together with other neurotransmitters and various neuropeptides to maintain the balance of synaptic functions. The dysfunction of the dopamine system leads to several conditions, including Parkinson's disease, Huntington's disease, major depression, schizophrenia, and drug addiction. The ventral tegmental area (VTA) has been identified as an important relay nucleus that modulates homeostatic plasticity in the midbrain dopamine system. Due to the complexity of synaptic transmissions and input-output connections in the VTA, the structure and function of this crucial brain region are still not fully understood. In this review article, we mainly focus on the cell types, neurotransmitters, neuropeptides, ion channels, receptors, and neural circuits of the VTA dopamine system, with the hope of obtaining new insight into the formation and function of this vital brain region.
Collapse
Affiliation(s)
- Guoqiang Hou
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China (M.H.); (J.D.)
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Mei Hao
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China (M.H.); (J.D.)
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jiawen Duan
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China (M.H.); (J.D.)
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Ming-Hu Han
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China (M.H.); (J.D.)
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
49
|
Hart G, Burton TJ, Nolan CR, Balleine BW. Striatal dopamine release tracks the relationship between actions and their consequences. Cell Rep 2024; 43:113828. [PMID: 38386550 DOI: 10.1016/j.celrep.2024.113828] [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: 09/06/2023] [Revised: 12/05/2023] [Accepted: 02/03/2024] [Indexed: 02/24/2024] Open
Abstract
The acquisition and performance of goal-directed actions has long been argued to depend on the integration of glutamatergic inputs to the posterior dorsomedial striatum (pDMS) under the modulatory influence of dopamine. Nevertheless, relatively little is known about the dynamics of striatal dopamine during goal-directed actions. To investigate this, we chronically recorded dopamine release in the pDMS as rats acquired two actions for distinct outcomes as these action-outcome associations were incremented and then subsequently degraded or reversed. We found that bilateral dopamine release scaled with action value, whereas the lateralized dopamine signal, i.e., the difference in dopamine release ipsilaterally and contralaterally to the direction of the goal-directed action, reflected the strength of the action-outcome association independently of changes in movement. Our results establish, therefore, that striatal dopamine activity during goal-directed action reflects both bilateral moment-to-moment changes in action value and the long-term action-outcome association.
Collapse
Affiliation(s)
- G Hart
- Decision Neuroscience Laboratory, School of Psychology, UNSW Sydney, Sydney, NSW, Australia
| | - T J Burton
- Decision Neuroscience Laboratory, School of Psychology, UNSW Sydney, Sydney, NSW, Australia
| | - C R Nolan
- Decision Neuroscience Laboratory, School of Psychology, UNSW Sydney, Sydney, NSW, Australia
| | - B W Balleine
- Decision Neuroscience Laboratory, School of Psychology, UNSW Sydney, Sydney, NSW, Australia.
| |
Collapse
|
50
|
Mocellin P, Barnstedt O, Luxem K, Kaneko H, Vieweg S, Henschke JU, Dalügge D, Fuhrmann F, Karpova A, Pakan JMP, Kreutz MR, Mikulovic S, Remy S. A septal-ventral tegmental area circuit drives exploratory behavior. Neuron 2024; 112:1020-1032.e7. [PMID: 38266645 DOI: 10.1016/j.neuron.2023.12.016] [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: 09/15/2022] [Revised: 11/10/2023] [Accepted: 12/20/2023] [Indexed: 01/26/2024]
Abstract
To survive, animals need to balance their exploratory drive with their need for safety. Subcortical circuits play an important role in initiating and modulating movement based on external demands and the internal state of the animal; however, how motivation and onset of locomotion are regulated remain largely unresolved. Here, we show that a glutamatergic pathway from the medial septum and diagonal band of Broca (MSDB) to the ventral tegmental area (VTA) controls exploratory locomotor behavior in mice. Using a self-supervised machine learning approach, we found an overrepresentation of exploratory actions, such as sniffing, whisking, and rearing, when this projection is optogenetically activated. Mechanistically, this role relies on glutamatergic MSDB projections that monosynaptically target a subset of both glutamatergic and dopaminergic VTA neurons. Taken together, we identified a glutamatergic basal forebrain to midbrain circuit that initiates locomotor activity and contributes to the expression of exploration-associated behavior.
Collapse
Affiliation(s)
- Petra Mocellin
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany; International Max Planck Research School for Brain and Behavior (IMPRS), Bonn 53175, Germany.
| | - Oliver Barnstedt
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany
| | - Kevin Luxem
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany
| | - Hiroshi Kaneko
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany
| | - Silvia Vieweg
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany
| | - Julia U Henschke
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany
| | - Dennis Dalügge
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany; International Max Planck Research School for Brain and Behavior (IMPRS), Bonn 53175, Germany
| | - Falko Fuhrmann
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany
| | - Anna Karpova
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany; Center for Behavioral Brain Sciences (CBBS), Magdeburg 39106, Germany
| | - Janelle M P Pakan
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany; German Center for Neurodegenerative Diseases (DZNE), Magdeburg 39120, Germany; Center for Behavioral Brain Sciences (CBBS), Magdeburg 39106, Germany
| | - Michael R Kreutz
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany; German Center for Neurodegenerative Diseases (DZNE), Magdeburg 39120, Germany; Center for Behavioral Brain Sciences (CBBS), Magdeburg 39106, Germany; German Center for Mental Health (DZPG), Magdeburg 39106, Germany
| | - Sanja Mikulovic
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany; Center for Behavioral Brain Sciences (CBBS), Magdeburg 39106, Germany; German Center for Mental Health (DZPG), Magdeburg 39106, Germany
| | - Stefan Remy
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany; German Center for Neurodegenerative Diseases (DZNE), Magdeburg 39120, Germany; Center for Behavioral Brain Sciences (CBBS), Magdeburg 39106, Germany; German Center for Mental Health (DZPG), Magdeburg 39106, Germany.
| |
Collapse
|