1
|
Wei A, Zhao A, Zheng C, Dong N, Cheng X, Duan X, Zhong S, Liu X, Jian J, Qin Y, Yang Y, Gu Y, Wang B, Gooya N, Huo J, Yao J, Li W, Huang K, Liu H, Mao F, Wang R, Shao M, Wang B, Zhang Y, Chen Y, Song Q, Huang R, Qu Q, Zhang C, Kang X, Xu H, Wang C. Sexually dimorphic dopaminergic circuits determine sex preference. Science 2025; 387:eadq7001. [PMID: 39787240 DOI: 10.1126/science.adq7001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 11/08/2024] [Indexed: 01/12/2025]
Abstract
Sociosexual preference is critical for reproduction and survival. However, neural mechanisms encoding social decisions on sex preference remain unclear. In this study, we show that both male and female mice exhibit female preference but shift to male preference when facing survival threats; their preference is mediated by the dimorphic changes in the excitability of ventral tegmental area dopaminergic (VTADA) neurons. In males, VTADA projections to the nucleus accumbens (NAc) mediate female preference, and those to the medial preoptic area mediate male preference. In females, firing-pattern (phasic-like versus tonic-like) alteration of the VTADA-NAc projection determines sociosexual preferences. These findings define VTADA neurons as a key node for social decision-making and reveal the sexually dimorphic DA circuit mechanisms underlying sociosexual preference.
Collapse
Affiliation(s)
- Anqi Wei
- Department of Neurology, the First Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Anran Zhao
- Department of Neurology, the First Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Chaowen Zheng
- Department of Neurology, the First Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Nan Dong
- Department of Neurology, the First Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Xu Cheng
- Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, and the Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Xueting Duan
- Department of Neurology, the First Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Shuaijie Zhong
- Department of Neurology, the First Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Xiaoying Liu
- Department of Neurology, the First Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Jie Jian
- Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, and the Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Yuhao Qin
- Department of Neurology, the First Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Yuxin Yang
- Department of Neurology, the First Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
- College of Agriculture and Biology, Liaocheng University, Liaocheng, China
| | - Yuhao Gu
- Department of Neurology, the First Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Bianbian Wang
- Department of Neurology, the First Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Niki Gooya
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jingxiao Huo
- Department of Neurology, the First Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Jingyu Yao
- Department of Neurology, the First Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Weiwei Li
- Department of Neurology, the First Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
- Department of Acupuncture, Massage and Rehabilitation, Shaanxi Provincial Hospital of Chinese Medicine, Xi'an, China
| | - Kai Huang
- Department of Neurology, the First Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Haiyao Liu
- Department of Neurology, the First Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Fenghan Mao
- Department of Neurology, the First Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Ruolin Wang
- Department of Neurology, the First Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Mingjie Shao
- Department of Neurology, the First Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Botao Wang
- Department of Neurology, the First Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Yichi Zhang
- Department of Neurology, the First Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Yang Chen
- Department of Neurology, the First Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Qian Song
- Department of Neurology, the First Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Rong Huang
- Department of Neurology, the First Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Qiumin Qu
- Department of Neurology, the First Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Chunxiang Zhang
- Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, and the Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Xinjiang Kang
- Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, and the Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- College of Agriculture and Biology, Liaocheng University, Liaocheng, China
| | - Huadong Xu
- Department of Neurology, the First Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Changhe Wang
- Department of Neurology, the First Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, and the Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Neurology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
2
|
Vounatsos MV, Gittis AH. GPe Projections to the Retrorubral Field Give Rise to Diverging GABAergic and DAergic Circuits. J Neurosci 2025; 45:e1446242024. [PMID: 39510836 PMCID: PMC11714346 DOI: 10.1523/jneurosci.1446-24.2024] [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: 07/30/2024] [Revised: 10/25/2024] [Accepted: 10/30/2024] [Indexed: 11/15/2024] Open
Abstract
The external segment of the globus pallidus (GPe) is involved in the modulation of the motor output and limbic components of behavior. The circuitry by which the GPe contributes to limbic components of behavior remains unknown. While many investigations have focused on the local circuitry within the basal ganglia, numerous GPe projections to target regions outside of the basal ganglia including the midbrain, thalamus, and cortex exist and remain largely unexplored. Here, by anatomical and electrophysiological means, we investigate the projections of the GPe to the retrorubral field (RRF), a dopaminergic nucleus in the caudal midbrain, in both male and female mice. We find that the GPe targets both GABAergic and DAergic neurons in the RRF (RRFDA and RRFGABA). Viral-assisted circuit mapping to trace the cell-type-specific projection patterns of RRFDA and RRFGABA populations revealed diverging pathways. The connectivity of GPe-recipient RRFDA neurons followed the traditional medial forebrain bundle path, innervating the ventral striatum and to a lesser extent the dorsal striatum. GPe-recipient RRFGABA neurons, on the other hand, displayed broad projections targeting numerous brain regions involved in limbic function including the parabrachial nucleus, amygdala, and hypothalamus. These contrasting projection profiles reveal multiple nodes by which the GPe is disynaptically connected to the limbic system, representing novel access points for the GPe to modulate limbic components of behavior.
Collapse
Affiliation(s)
- Maxime V Vounatsos
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213
- Center for the Neural Basis of Cognition, Pittsburgh, Pennsylvania 15213
| | - Aryn H Gittis
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213
- Center for the Neural Basis of Cognition, Pittsburgh, Pennsylvania 15213
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213
| |
Collapse
|
3
|
Hubbard E, Derdeyn P, Galinato VM, Wu A, Bartas K, Mahler SV, Beier KT. Neural basis of adolescent THC-induced potentiation of opioid responses later in life. Neuropsychopharmacology 2024:10.1038/s41386-024-02033-8. [PMID: 39658631 DOI: 10.1038/s41386-024-02033-8] [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: 05/03/2024] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 12/12/2024]
Abstract
Use of one addictive drug typically influences the behavioral response to other drugs, either administered at the same time or a subsequent time point. The nature of the drugs being used, as well as the timing and dosing, also influence how these drugs interact. Here, we tested the effects of adolescent THC exposure on the development of morphine-induced behavioral adaptations following repeated morphine exposure during adulthood. We found that adolescent THC administration paradoxically prevented the development of anxiety-related behaviors that emerge during a forced abstinence period following morphine administration but facilitated reinstatement of morphine CPP. Following forced abstinence, we then mapped the whole-brain response to a moderate dose of morphine and found that adolescent THC administration led to an overall increase in brain-wide neuronal activity and increased the functional connectivity between frontal cortical regions and the ventral tegmental area. Last, we show using rabies virus-based circuit mapping that adolescent THC exposure triggers a long-lasting elevation in connectivity from the frontal cortex regions onto ventral tegmental dopamine cells. Our study adds to the rich literature on the interaction between drugs, including THC and opioids, and provides potential neural substates by which adolescent THC exposure influences responses to morphine later in life.
Collapse
Affiliation(s)
- Elizabeth Hubbard
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
| | - Pieter Derdeyn
- Program in Mathematical, Computational, and Systems Biology, University of California, Irvine, CA, USA
| | | | - Andrew Wu
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
| | - Katrina Bartas
- Program in Mathematical, Computational, and Systems Biology, University of California, Irvine, CA, USA
| | - Stephen V Mahler
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Kevin T Beier
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA.
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA.
- Department of Biomedical Engineering, University of California, Irvine, CA, USA.
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, USA.
| |
Collapse
|
4
|
Tian G, Bartas K, Hui M, Chen L, Vasquez JJ, Azouz G, Derdeyn P, Manville RW, Ho EL, Fang AS, Li Y, Tyler I, Setola V, Aoto J, Abbott GW, Beier KT. Molecular and circuit determinants in the globus pallidus mediating control of cocaine-induced behavioral plasticity. Neuron 2024; 112:3470-3485.e12. [PMID: 39153478 PMCID: PMC11502257 DOI: 10.1016/j.neuron.2024.07.018] [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: 07/28/2023] [Revised: 04/12/2024] [Accepted: 07/22/2024] [Indexed: 08/19/2024]
Abstract
The globus pallidus externus (GPe) is a central component of the basal ganglia circuit that acts as a gatekeeper of cocaine-induced behavioral plasticity. However, the molecular and circuit mechanisms underlying this function are unknown. Here, we show that GPe parvalbumin-positive (GPePV) cells mediate cocaine responses by selectively modulating ventral tegmental area dopamine (VTADA) cells projecting to the dorsomedial striatum (DMS). Interestingly, GPePV cell activity in cocaine-naive mice is correlated with behavioral responses following cocaine, effectively predicting cocaine sensitivity. Expression of the voltage-gated potassium channels KCNQ3 and KCNQ5 that control intrinsic cellular excitability following cocaine was downregulated, contributing to the elevation in GPePV cell excitability. Acutely activating channels containing KCNQ3 and/or KCNQ5 using the small molecule carnosic acid, a key psychoactive component of Salvia rosmarinus (rosemary) extract, reduced GPePV cell excitability and impaired cocaine reward, sensitization, and volitional cocaine intake, indicating its therapeutic potential to counteract psychostimulant use disorder.
Collapse
Affiliation(s)
- Guilian Tian
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Katrina Bartas
- Program in Mathematical, Computational, and Systems Biology, University of California, Irvine, Irvine, CA, USA
| | - May Hui
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Lingxuan Chen
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Jose J Vasquez
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Ghalia Azouz
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Pieter Derdeyn
- Program in Mathematical, Computational, and Systems Biology, University of California, Irvine, Irvine, CA, USA
| | - Rían W Manville
- Bioelectricity Laboratory, Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Erick L Ho
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Amanda S Fang
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Yuan Li
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Isabella Tyler
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Vincent Setola
- Department of Neuroscience, West Virginia University, Morgantown, WV, USA; Department of Behavioral Medicine and Psychiatry, West Virginia University, Morgantown, WV, USA
| | - Jason Aoto
- University of Colorado Anschutz School of Medicine, Department of Pharmacology, Aurora, CO, USA
| | - Geoffrey W Abbott
- Bioelectricity Laboratory, Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Kevin T Beier
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA; Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA; Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA; Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
5
|
Wei J, Xiao C, Zhang GW, Shen L, Tao HW, Zhang LI. A distributed auditory network mediated by pontine central gray underlies ultra-fast awakening in response to alerting sounds. Curr Biol 2024; 34:4597-4611.e5. [PMID: 39265569 PMCID: PMC11521200 DOI: 10.1016/j.cub.2024.08.020] [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: 03/11/2024] [Revised: 07/12/2024] [Accepted: 08/13/2024] [Indexed: 09/14/2024]
Abstract
Sleeping animals can be woken up rapidly by external threat signals, which is an essential defense mechanism for survival. However, neuronal circuits underlying the fast transmission of sensory signals for this process remain unclear. Here, we report in mice that alerting sound can induce rapid awakening within hundreds of milliseconds and that glutamatergic neurons in the pontine central gray (PCG) play an important role in this process. These neurons exhibit higher sensitivity to auditory stimuli in sleep than wakefulness. Suppressing these neurons results in reduced sound-induced awakening and increased sleep in intrinsic sleep/wake cycles, whereas their activation induces ultra-fast awakening from sleep and accelerates awakening from anesthesia. Additionally, the sound-induced awakening can be attributed to the propagation of auditory signals from the PCG to multiple arousal-related regions, including the mediodorsal thalamus, lateral hypothalamus, and ventral tegmental area. Thus, the PCG serves as an essential distribution center to orchestrate a global auditory network to promote rapid awakening.
Collapse
Affiliation(s)
- Jinxing Wei
- Zilkha Neurogenetic Institute, Center for Neural Circuits and Sensory Processing Disorders, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Cuiyu Xiao
- Zilkha Neurogenetic Institute, Center for Neural Circuits and Sensory Processing Disorders, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Guang-Wei Zhang
- Zilkha Neurogenetic Institute, Center for Neural Circuits and Sensory Processing Disorders, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Li Shen
- Zilkha Neurogenetic Institute, Center for Neural Circuits and Sensory Processing Disorders, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Huizhong W Tao
- Zilkha Neurogenetic Institute, Center for Neural Circuits and Sensory Processing Disorders, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Li I Zhang
- Zilkha Neurogenetic Institute, Center for Neural Circuits and Sensory Processing Disorders, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
6
|
Mohammadkhani A, Qiao M, Borgland SL. Distinct Neuromodulatory Effects of Endogenous Orexin and Dynorphin Corelease on Projection-Defined Ventral Tegmental Dopamine Neurons. J Neurosci 2024; 44:e0682242024. [PMID: 39187377 PMCID: PMC11426376 DOI: 10.1523/jneurosci.0682-24.2024] [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: 04/09/2024] [Revised: 08/11/2024] [Accepted: 08/16/2024] [Indexed: 08/28/2024] Open
Abstract
Dopamine (DA) neurons in the ventral tegmental area (VTA) respond to motivationally relevant cues, and circuit-specific signaling drives different aspects of motivated behavior. Orexin (ox; also known as hypocretin) and dynorphin (dyn) are coexpressed lateral hypothalamic (LH) neuropeptides that project to the VTA. These peptides have opposing effects on the firing activity of VTADA neurons via orexin 1 (Ox1R) or kappa opioid (KOR) receptors. Given that Ox1R activation increases VTADA firing, and KOR decreases firing, it is unclear how the coreleased peptides contribute to the net activity of DA neurons. We tested if optical stimulation of LHox/dyn neuromodulates VTADA neuronal activity via peptide release and if the effects of optically driven LHox/dyn release segregate based on VTADA projection targets including the basolateral amygdala (BLA) or the lateral or medial shell of the nucleus accumbens (lAcbSh, mAchSh). Using a combination of circuit tracing, optogenetics, and patch-clamp electrophysiology in male and female orexincre mice, we showed a diverse response of LHox/dyn optical stimulation on VTADA neuronal firing, which is not mediated by fast transmitter release and is blocked by antagonists to KOR and Ox1R signaling. Additionally, where optical stimulation of LHox/dyn inputs in the VTA inhibited firing of the majority of BLA-projecting VTADA neurons, optical stimulation of LHox/dyn inputs in the VTA bidirectionally affects firing of either lAcbSh- or mAchSh-projecting VTADA neurons. These findings indicate that LHox/dyn corelease may influence the output of the VTA by balancing ensembles of neurons within each population which contribute to different aspects of reward seeking.
Collapse
Affiliation(s)
- Aida Mohammadkhani
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, The University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Min Qiao
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, The University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Stephanie L Borgland
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, The University of Calgary, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
7
|
Hernandez Silva JC, Pausic N, Marroquin Rivera A, Labonté B, Proulx CD. Chronic Social Defeat Stress Induces Pathway-Specific Adaptations at Lateral Habenula Neuronal Outputs. J Neurosci 2024; 44:e2082232024. [PMID: 39164106 PMCID: PMC11426382 DOI: 10.1523/jneurosci.2082-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 07/15/2024] [Accepted: 08/10/2024] [Indexed: 08/22/2024] Open
Abstract
The lateral habenula (LHb) has emerged as a pivotal brain region implicated in depression, displaying hyperactivity in human and animal models of depression. While the role of LHb efferents in depressive disorders has been acknowledged, the specific synaptic alterations remain elusive. Here, employing optogenetics, retrograde tracing, and ex vivo whole-cell patch-clamp techniques, we investigated synaptic transmission in male mice subjected to chronic social defeat stress (CSDS) at three major LHb neuronal outputs: the dorsal raphe nucleus (DRN), the ventral tegmental area (VTA), and the rostromedial tegmental nucleus (RMTg). Our findings uncovered distinct synaptic adaptations in LHb efferent circuits in response to CSDS. Specifically, CSDS induced in susceptible mice postsynaptic potentiation and postsynaptic depression at the DRN and VTA neurons, respectively, receiving excitatory inputs from the LHb, while CSDS altered presynaptic transmission at the LHb terminals in RMTg in both susceptible and resilient mice. Moreover, whole-cell recordings at projection-defined LHb neurons indicate decreased spontaneous activity in VTA-projecting LHb neurons, accompanied by an imbalance in excitatory-inhibitory inputs at the RMTg-projecting LHb neurons. Collectively, these novel findings underscore the circuit-specific alterations in LHb efferents following chronic social stress, shedding light on potential synaptic adaptations underlying stress-induced depressive-like states.
Collapse
Affiliation(s)
- Jose Cesar Hernandez Silva
- CERVO Brain Research Center, Department of Psychiatry and Neuroscience, Faculty of medicine, Université Laval, Quebec City, QC G1J 2G3, Canada
| | - Nikola Pausic
- CERVO Brain Research Center, Department of Psychiatry and Neuroscience, Faculty of medicine, Université Laval, Quebec City, QC G1J 2G3, Canada
| | - Arturo Marroquin Rivera
- CERVO Brain Research Center, Department of Psychiatry and Neuroscience, Faculty of medicine, Université Laval, Quebec City, QC G1J 2G3, Canada
| | - Benoît Labonté
- CERVO Brain Research Center, Department of Psychiatry and Neuroscience, Faculty of medicine, Université Laval, Quebec City, QC G1J 2G3, Canada
| | - Christophe D Proulx
- CERVO Brain Research Center, Department of Psychiatry and Neuroscience, Faculty of medicine, Université Laval, Quebec City, QC G1J 2G3, Canada
| |
Collapse
|
8
|
Park G, Park Y, Yang S, Cho Y, Serikov A, Jung D, Seo DC, Lee SE, Nam MH, Kim D, Kim J. Neurotensin-specific corticothalamic circuit regulates innate response conflict. Curr Biol 2024; 34:3473-3487.e6. [PMID: 39067450 DOI: 10.1016/j.cub.2024.06.068] [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: 01/14/2024] [Revised: 04/22/2024] [Accepted: 06/26/2024] [Indexed: 07/30/2024]
Abstract
Animals must simultaneously select and balance multiple action contingencies in ambiguous situations: for instance, evading danger during feeding. This has rarely been examined in the context of information selection; despite corticothalamic pathways that mediate sensory attention being relatively well characterized, neural mechanisms filtering conflicting actions remain unclear. Here, we develop a new loom/feed test to observe conflict between naturally induced fear and feeding and identify a novel anterior cingulate cortex (ACC) output to the ventral anterior and ventral lateral thalamus (VA/VL) that adjusts selectivity between these innate actions. Using micro-endoscopy and fiber photometry, we reveal that activity in corticofugal outputs was lowered during unbalanced/singularly occupied periods, as were the resulting decreased thalamic initiation-related signals for less-favored actions, suggesting that the integration of ACC-thalamic firing may directly regulate the output of behavior choices. Accordingly, the optoinhibition of ACC-VA/VL circuits induced high bias toward feeding at the expense of defense. To identify upstream "commander" cortical cells gating this output, we established dual-order tracing (DOT)-translating ribosome affinity purification (TRAP)-a scheme to label upstream neurons with transcriptome analysis-and found a novel population of neurotensin-positive interneurons (ACCNts). The photoexcitation of ACCNts cells indeed caused similarly hyper-selective behaviors. Collectively, this new "corticofugal action filter" scheme suggests that communication in multi-step cingulate circuits may critically influence the summation of motor signals in thalamic outputs, regulating bias between innate action types.
Collapse
Affiliation(s)
- Geunhong Park
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Yongjun Park
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, Seoul 02792, Republic of Korea
| | - Seulkee Yang
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Yoonjeong Cho
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Almas Serikov
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, Seoul 02792, Republic of Korea
| | - Dajung Jung
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Dong-Chan Seo
- Research Animal Resources Center, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Seung Eun Lee
- Research Animal Resources Center, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Min-Ho Nam
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, Seoul 02792, Republic of Korea
| | - Daesoo Kim
- Department of Brain and Cognitive Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Jeongjin Kim
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, Seoul 02792, Republic of Korea.
| |
Collapse
|
9
|
Marinescu AM, Labouesse MA. The nucleus accumbens shell: a neural hub at the interface of homeostatic and hedonic feeding. Front Neurosci 2024; 18:1437210. [PMID: 39139500 PMCID: PMC11319282 DOI: 10.3389/fnins.2024.1437210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/16/2024] [Indexed: 08/15/2024] Open
Abstract
Feeding behavior is a complex physiological process regulated by the interplay between homeostatic and hedonic feeding circuits. Among the neural structures involved, the nucleus accumbens (NAc) has emerged as a pivotal region at the interface of these two circuits. The NAc comprises distinct subregions and in this review, we focus mainly on the NAc shell (NAcSh). Homeostatic feeding circuits, primarily found in the hypothalamus, ensure the organism's balance in energy and nutrient requirements. These circuits monitor peripheral signals, such as insulin, leptin, and ghrelin, and modulate satiety and hunger states. The NAcSh receives input from these homeostatic circuits, integrating information regarding the organism's metabolic needs. Conversely, so-called hedonic feeding circuits involve all other non-hunger and -satiety processes, i.e., the sensory information, associative learning, reward, motivation and pleasure associated with food consumption. The NAcSh is interconnected with hedonics-related structures like the ventral tegmental area and prefrontal cortex and plays a key role in encoding hedonic information related to palatable food seeking or consumption. In sum, the NAcSh acts as a crucial hub in feeding behavior, integrating signals from both homeostatic and hedonic circuits, to facilitate behavioral output via its downstream projections. Moreover, the NAcSh's involvement extends beyond simple integration, as it directly impacts actions related to food consumption. In this review, we first focus on delineating the inputs targeting the NAcSh; we then present NAcSh output projections to downstream structures. Finally we discuss how the NAcSh regulates feeding behavior and can be seen as a neural hub integrating homeostatic and hedonic feeding signals, via a functionally diverse set of projection neuron subpopulations.
Collapse
Affiliation(s)
- Alina-Măriuca Marinescu
- Brain, Wire and Behavior Group, Translational Nutritional Biology Laboratory, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Marie A. Labouesse
- Brain, Wire and Behavior Group, Translational Nutritional Biology Laboratory, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
10
|
Higgs MH, Beckstead MJ. Impact of Unitary Synaptic Inhibition on Spike Timing in Ventral Tegmental Area Dopamine Neurons. eNeuro 2024; 11:ENEURO.0203-24.2024. [PMID: 38969500 PMCID: PMC11287791 DOI: 10.1523/eneuro.0203-24.2024] [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: 05/11/2024] [Revised: 06/25/2024] [Accepted: 06/28/2024] [Indexed: 07/07/2024] Open
Abstract
Midbrain dopamine neurons receive convergent synaptic input from multiple brain areas, which perturbs rhythmic pacemaking to produce the complex firing patterns observed in vivo. This study investigated the impact of single and multiple inhibitory inputs on ventral tegmental area (VTA) dopamine neuron firing in mice of both sexes using novel experimental measurements and modeling. We first measured unitary inhibitory postsynaptic currents produced by single axons using both minimal electrical stimulation and minimal optical stimulation of rostromedial tegmental nucleus and ventral pallidum afferents. We next determined the phase resetting curve, the reversal potential for GABAA receptor-mediated inhibitory postsynaptic currents (IPSCs), and the average interspike membrane potential trajectory during pacemaking. We combined these data in a phase oscillator model of a VTA dopamine neuron, simulating the effects of unitary inhibitory postsynaptic conductances (uIPSGs) on spike timing and rate. The effect of a uIPSG on spike timing was predicted to vary according to its timing within the interspike interval or phase. Simulations were performed to predict the pause duration resulting from the synchronous arrival of multiple uIPSGs and the changes in firing rate and regularity produced by asynchronous uIPSGs. The model data suggest that asynchronous inhibition is more effective than synchronous inhibition, because it tends to hold the neuron at membrane potentials well positive to the IPSC reversal potential. Our results indicate that small fluctuations in the inhibitory synaptic input arriving from the many afferents to each dopamine neuron are sufficient to produce highly variable firing patterns, including pauses that have been implicated in reinforcement.
Collapse
Affiliation(s)
- Matthew H Higgs
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
| | - Michael J Beckstead
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
| |
Collapse
|
11
|
Tian G, Bartas K, Hui M, Chen L, Vasquez JJ, Azouz G, Derdeyn P, Manville RW, Ho EL, Fang AS, Li Y, Tyler I, Setola V, Aoto J, Abbott GW, Beier KT. Molecular and circuit determinants in the globus pallidus mediating control of cocaine-induced behavioral plasticity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.29.596557. [PMID: 38853899 PMCID: PMC11160764 DOI: 10.1101/2024.05.29.596557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
The globus pallidus externus (GPe) is a central component of the basal ganglia circuit, receiving strong input from the indirect pathway and regulating a variety of functions, including locomotor output and habit formation. We recently showed that it also acts as a gatekeeper of cocaine-induced behavioral plasticity, as inhibition of parvalbumin-positive cells in the GPe (GPe PV ) prevents the development of cocaine-induced reward and sensitization. However, the molecular and circuit mechanisms underlying this function are unknown. Here we show that GPe PV cells control cocaine reward and sensitization by inhibiting GABAergic neurons in the substantia nigra pars reticulata (SNr GABA ), and ultimately, selectively modulating the activity of ventral tegmental area dopamine (VTA DA ) cells projecting to the lateral shell of the nucleus accumbens (NAcLat). A major input to GPe PV cells is the indirect pathway of the dorsomedial striatum (DMS D 2 ), which receives DAergic innervation from collaterals of VTA DA →NAcLat cells, making this a closed-loop circuit. Cocaine likely facilitates reward and sensitization not directly through actions in the GPe, but rather in the upstream DMS, where the cocaine-induced elevation of DA triggers a depression in DMS D 2 cell activity. This cocaine-induced elevation in DA levels can be blocked by inhibition of GPe PV cells, closing the loop. Interestingly, the level of GPe PV cell activity prior to cocaine administration is correlated with the extent of reward and sensitization that animals experience in response to future administration of cocaine, indicating that GPe PV cell activity is a key predictor of future behavioral responses to cocaine. Single nucleus RNA-sequencing of GPe cells indicated that genes encoding voltage-gated potassium channels KCNQ3 and KCNQ5 that control intrinsic cellular excitability are downregulated in GPe PV cells following a single cocaine exposure, contributing to the elevation in GPe PV cell excitability. Acutely activating channels containing KCNQ3 and/or KCNQ5 using the small molecule carnosic acid, a key psychoactive component of Salvia rosmarinus (rosemary) extract, reduced GPe PV cell excitability and also impaired cocaine reward, sensitization, and volitional cocaine intake, indicating its potential as a therapeutic to counteract psychostimulant use disorder. Our findings illuminate the molecular and circuit mechanisms by which the GPe orchestrates brain-wide changes in response to cocaine that are required for reward, sensitization, and self-administration behaviors.
Collapse
|
12
|
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
|
13
|
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
|
14
|
Song R, Soler-Cedeño O, Xi ZX. Optical Intracranial Self-Stimulation (oICSS): A New Behavioral Model for Studying Drug Reward and Aversion in Rodents. Int J Mol Sci 2024; 25:3455. [PMID: 38542425 PMCID: PMC10970671 DOI: 10.3390/ijms25063455] [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: 01/18/2024] [Revised: 03/10/2024] [Accepted: 03/17/2024] [Indexed: 11/03/2024] Open
Abstract
Brain-stimulation reward, also known as intracranial self-stimulation (ICSS), is a commonly used procedure for studying brain reward function and drug reward. In electrical ICSS (eICSS), an electrode is surgically implanted into the medial forebrain bundle (MFB) in the lateral hypothalamus or the ventral tegmental area (VTA) in the midbrain. Operant lever responding leads to the delivery of electrical pulse stimulation. The alteration in the stimulation frequency-lever response curve is used to evaluate the impact of pharmacological agents on brain reward function. If a test drug induces a leftward or upward shift in the eICSS response curve, it implies a reward-enhancing or abuse-like effect. Conversely, if a drug causes a rightward or downward shift in the functional response curve, it suggests a reward-attenuating or aversive effect. A significant drawback of eICSS is the lack of cellular selectivity in understanding the neural substrates underlying this behavior. Excitingly, recent advancements in optical ICSS (oICSS) have facilitated the development of at least three cell type-specific oICSS models-dopamine-, glutamate-, and GABA-dependent oICSS. In these new models, a comparable stimulation frequency-lever response curve has been established and employed to study the substrate-specific mechanisms underlying brain reward function and a drug's rewarding versus aversive effects. In this review article, we summarize recent progress in this exciting research area. The findings in oICSS have not only increased our understanding of the neural mechanisms underlying drug reward and addiction but have also introduced a novel behavioral model in preclinical medication development for treating substance use disorders.
Collapse
Affiliation(s)
- Rui Song
- Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology (BIPT), 27th Taiping Road, Beijing 100850, China
| | - Omar Soler-Cedeño
- Addiction Biology Unit, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse (NIDA), Intramural Research Program (IRP), Baltimore, MD 21224, USA;
| | - Zheng-Xiong Xi
- Addiction Biology Unit, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse (NIDA), Intramural Research Program (IRP), Baltimore, MD 21224, USA;
| |
Collapse
|
15
|
Washburn S, Oñate M, Yoshida J, Vera J, Bhuvanasundaram R, Khatami L, Nadim F, Khodakhah K. The cerebellum directly modulates the substantia nigra dopaminergic activity. Nat Neurosci 2024; 27:497-513. [PMID: 38272967 PMCID: PMC11441724 DOI: 10.1038/s41593-023-01560-9] [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: 05/05/2022] [Accepted: 12/13/2023] [Indexed: 01/27/2024]
Abstract
Evidence of direct reciprocal connections between the cerebellum and basal ganglia has challenged the long-held notion that these structures function independently. While anatomical studies have suggested the presence of cerebellar projections to the substantia nigra pars compacta (SNc), the nature and function of these connections (Cb-SNc) is unknown. Here we show, in mice, that Cb-SNc projections form monosynaptic glutamatergic synapses with dopaminergic and non-dopaminergic neurons in the SNc. Optogenetic activation of Cb-SNc axons in the SNc is associated with increased SNc activity, elevated striatal dopamine levels and increased locomotion. During behavior, Cb-SNc projections are bilaterally activated before ambulation and unilateral lever manipulation. Cb-SNc projections show prominent activation for water reward and higher activation for sweet water, suggesting that the pathway also encodes reward value. Thus, the cerebellum directly, rapidly and effectively modulates basal ganglia dopamine levels and conveys information related to movement initiation, vigor and reward processing.
Collapse
Affiliation(s)
- Samantha Washburn
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Maritza Oñate
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Junichi Yoshida
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jorge Vera
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | - Leila Khatami
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Farzan Nadim
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Biological Sciences, New Jersey Institute of Technology, Newark, NJ, USA
| | - Kamran Khodakhah
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA.
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY, USA.
- Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
16
|
Clarke-Williams CJ, Lopes-Dos-Santos V, Lefèvre L, Brizee D, Causse AA, Rothaermel R, Hartwich K, Perestenko PV, Toth R, McNamara CG, Sharott A, Dupret D. Coordinating brain-distributed network activities in memory resistant to extinction. Cell 2024; 187:409-427.e19. [PMID: 38242086 PMCID: PMC7615560 DOI: 10.1016/j.cell.2023.12.018] [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: 10/12/2022] [Revised: 07/13/2023] [Accepted: 12/13/2023] [Indexed: 01/21/2024]
Abstract
Certain memories resist extinction to continue invigorating maladaptive actions. The robustness of these memories could depend on their widely distributed implementation across populations of neurons in multiple brain regions. However, how dispersed neuronal activities are collectively organized to underpin a persistent memory-guided behavior remains unknown. To investigate this, we simultaneously monitored the prefrontal cortex, nucleus accumbens, amygdala, hippocampus, and ventral tegmental area (VTA) of the mouse brain from initial recall to post-extinction renewal of a memory involving cocaine experience. We uncover a higher-order pattern of short-lived beta-frequency (15-25 Hz) activities that are transiently coordinated across these networks during memory retrieval. The output of a divergent pathway from upstream VTA glutamatergic neurons, paced by a slower (4-Hz) oscillation, actuates this multi-network beta-band coactivation; its closed-loop phase-informed suppression prevents renewal of cocaine-biased behavior. Binding brain-distributed neural activities in this temporally structured manner may constitute an organizational principle of robust memory expression.
Collapse
Affiliation(s)
- Charlie J Clarke-Williams
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK.
| | - Vítor Lopes-Dos-Santos
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK
| | - Laura Lefèvre
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK
| | - Demi Brizee
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK
| | - Adrien A Causse
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK
| | - Roman Rothaermel
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK
| | - Katja Hartwich
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK
| | - Pavel V Perestenko
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK
| | - Robert Toth
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK
| | - Colin G McNamara
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK
| | - Andrew Sharott
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK
| | - David Dupret
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK.
| |
Collapse
|
17
|
Fudge JL, Kelly EA, Love TM. Amygdalo-nigral inputs target dopaminergic and GABAergic neurons in the primate: a view from dendrites and soma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.16.575910. [PMID: 38293165 PMCID: PMC10827221 DOI: 10.1101/2024.01.16.575910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
The central nucleus (CeN) of the amygdala is an important afferent to the DA system that mediates motivated learning. We previously found that CeN terminals in nonhuman primates primarily overlap the elongated lateral VTA (parabrachial pigmented nucleus, PBP, A10), and retrorubral field(A8) subregion. Here, we examined CeN afferent contacts on cell somata and proximal dendrites of DA and GABA neurons, and distal dendrites of each, using confocal and electron microscopy (EM) methods, respectively. At the soma/proximal dendrites, the proportion of TH+ and GAD1+ cells receiving at least one CeN afferent contact was surprisingly similar (TH = 0.55: GAD1=0.55 in PBP; TH = 0.56; GAD1 =0.51 in A8), with the vast majority of contacted TH+ and GAD1+ soma/proximal dendrites received 1-2 contacts. Similar numbers of tracer-labeled terminals also contacted TH-positive and GAD1-positive small dendrites and/or spines (39% of all contacted dendrites were either TH- or GAD1-labeled). Overall, axon terminals had more symmetric (putative inhibitory) axonal contacts with no difference in the relative distribution in the PBP versus A8, or onto TH+ versus GAD1+ dendrites/spines in either region. The striking uniformity in the amygdalonigral projection across the PBP-A8 terminal field suggests that neither neurotransmitter phenotype nor midbrain location dictates likelihood of a terminal contact. We discuss how this afferent uniformity can play out in recently discovered differences in DA:GABA cell densities between the PBP and A8, and affect specific outputs. Significance statement The amygdala's central nucleus (CeN) channels salient cues to influence both appetitive and aversive responses via DA outputs. In higher species, the broad CeN terminal field overlaps the parabrachial pigmented nucleus ('lateral A10') and the retrorubral field (A8). We quantified terminal contacts in each region on DA and GABAergic soma/proximal dendrites and small distal dendrites. There was striking uniformity in contacts on DA and GABAergic cells, regardless of soma and dendritic compartment, in both regions. Most contacts were symmetric (putative inhibitory) with little change in the ratio of inhibitory to excitatory contacts by region.We conclude that post-synaptic shifts in DA-GABA ratios are key to understanding how these relatively uniform inputs can produce diverse effects on outputs.
Collapse
|
18
|
Tong Y, Cho S, Coenen VA, Döbrössy MD. Input-output relation of midbrain connectomics in a rodent model of depression. J Affect Disord 2024; 345:443-454. [PMID: 37890539 DOI: 10.1016/j.jad.2023.10.124] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/13/2023] [Accepted: 10/18/2023] [Indexed: 10/29/2023]
Abstract
BACKGROUND The symptoms associated with depression are believed to arise from disruptions in information processing across brain networks. The ventral tegmental area (VTA) influences reward-based behavior, motivation, addiction, and psychiatric disorders, including depression. Deep brain stimulation (DBS) of the medial forebrain bundle (MFB), is an emerging therapy for treatment-resistant depression. Understanding the depression associated anatomical networks crucial for comprehending its antidepressant effects. METHODS Flinders Sensitive Line (FSL), a rodent model of depression and Sprague-Dawley rats (n = 10 each) were used in this study. We used monosynaptic tracing to map inputs of VTA efferent neurons: VTA-to-NAc nucleus accumbens (NAc) (both core and shell) and VTA-to-prefrontal cortex (PFC). Quantitative analysis explored afferent diversity and strengths. RESULTS VTA efferent neurons receive a variety of afferents with varying input weights and predominant neuromodulatory representation. Notably, NAc-core projecting VTA neurons showed stronger afferents from dorsal raphe, while NAc shell-projecting VTA neurons displayed lower input strengths from cortex, thalamus, zona incerta and pretectal area in FSL rats. NAc shell-projecting VTA neurons showed the most difference in connectivity across the experimental groups. LIMITATIONS Lack of functional properties of the anatomical connections is the major limitation of this study. Incomplete labeling and the cytotoxicity of the rabies virus should be made aware of. CONCLUSIONS These findings provide the first characterization of inputs to different VTA ascending projection neurons, shedding light on critical differences in the connectome of the midbrain-forebrain system. Moreover, these differences support potential network effects of these circuits in the context of MFB DBS neuromodulation for depression.
Collapse
Affiliation(s)
- Y Tong
- Laboratory of Stereotaxy and Interventional Neurosciences, Department of Stereotactic and Functional Neurosurgery, University Hospital Freiburg, 79106 Freiburg, Germany; Department of Stereotactic and Functional Neurosurgery, University Hospital Freiburg, 79106 Freiburg, Germany
| | - S Cho
- Laboratory of Stereotaxy and Interventional Neurosciences, Department of Stereotactic and Functional Neurosurgery, University Hospital Freiburg, 79106 Freiburg, Germany; Department of Stereotactic and Functional Neurosurgery, University Hospital Freiburg, 79106 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - V A Coenen
- Laboratory of Stereotaxy and Interventional Neurosciences, Department of Stereotactic and Functional Neurosurgery, University Hospital Freiburg, 79106 Freiburg, Germany; Department of Stereotactic and Functional Neurosurgery, University Hospital Freiburg, 79106 Freiburg, Germany; Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; Center for Basics in Neuromodulation, University of Freiburg, 79106 Freiburg, Germany; IMBIT (Institute for Machine-Brain Interfacing Technology), University of Freiburg, 79110 Freiburg, Germany
| | - M D Döbrössy
- Laboratory of Stereotaxy and Interventional Neurosciences, Department of Stereotactic and Functional Neurosurgery, University Hospital Freiburg, 79106 Freiburg, Germany; Department of Stereotactic and Functional Neurosurgery, University Hospital Freiburg, 79106 Freiburg, Germany; Center for Basics in Neuromodulation, University of Freiburg, 79106 Freiburg, Germany.
| |
Collapse
|
19
|
Wang F, Liu CB, Wang Y, Wang XX, Yang YY, Jiang CY, Le QM, Liu X, Ma L, Wang FF. Morphine- and foot shock-responsive neuronal ensembles in the VTA possess different connectivity and biased GPCR signaling pathway. Theranostics 2024; 14:1126-1146. [PMID: 38250036 PMCID: PMC10797299 DOI: 10.7150/thno.90792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 01/02/2024] [Indexed: 01/23/2024] Open
Abstract
Background: Neurons in the ventral tegmental area (VTA) are sensitive to stress and their maladaptation have been implicated in the psychiatric disorders such as anxiety and addiction, etc. The cellular properties of the VTA neurons in response to different stressors related to different emotional processing remain to be investigated. Methods: By combining immediate early gene (IEG)-dependent labeling, rabies virus tracing, ensemble-specific transcriptomic analysis and fiber photometry recording in the VTA of male mice, the spatial distribution, brain-wide connectivity and cellular signaling pathways in the VTA neuronal ensembles in response to morphine (Mor-Ens) or foot shock (Shock-Ens) stimuli were investigated. Results: Optogenetic activation of the Mor-Ens drove approach behavior, whereas chemogenetic activation of the Shock-Ens increased the anxiety level in mice. Mor-Ens were clustered and enriched in the ventral VTA, contained a higher proportion of dopaminergic neurons, received more inputs from the dorsal medial striatum and the medial hypothalamic zone, and exhibited greater axonal arborization in the zona incerta and ventral pallidum. Whereas Shock-Ens were more dispersed, contained a higher proportion of GABAergic neurons, and received more inputs from the ventral pallidum and the lateral hypothalamic area. The downstream targets of the G protein and β-arrestin pathways, PLCβ3 and phosphorylated AKT1Thr308, were relatively enriched in the Mor-Ens and Shock-Ens, respectively. Cariprazine, the G-protein-biased agonist for the dopamine D2 receptor, increased the response of Mor-Ens to sucrose water and decreased the anxiety-like behavior during morphine withdrawal, whereas the β-arrestin-biased agonist UNC9994 decreased the response of Shock-Ens to tail suspension. Conclusions: Taken together, these findings reveal the heterogeneous connectivity and signaling pathways of the VTA neurons in response to morphine and foot shock, providing new insights for development of specific interventions for psychiatric disorders caused by various stressors associated with different VTA neuronal functions.
Collapse
Affiliation(s)
- Fan Wang
- School of Basic Medical Sciences, MOE Frontiers Center for Brain Science, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Pharmacology Research Center, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China
| | - Chao-bao Liu
- School of Basic Medical Sciences, MOE Frontiers Center for Brain Science, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Pharmacology Research Center, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China
| | - Yi Wang
- School of Basic Medical Sciences, MOE Frontiers Center for Brain Science, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Pharmacology Research Center, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China
| | - Xi-xi Wang
- School of Basic Medical Sciences, MOE Frontiers Center for Brain Science, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Pharmacology Research Center, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China
| | - Yuan-yao Yang
- School of Basic Medical Sciences, MOE Frontiers Center for Brain Science, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Pharmacology Research Center, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China
| | - Chang-you Jiang
- School of Basic Medical Sciences, MOE Frontiers Center for Brain Science, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Pharmacology Research Center, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China
- Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China
| | - Qiu-min Le
- School of Basic Medical Sciences, MOE Frontiers Center for Brain Science, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Pharmacology Research Center, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China
- Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China
| | - Xing Liu
- School of Basic Medical Sciences, MOE Frontiers Center for Brain Science, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Pharmacology Research Center, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China
- Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China
| | - Lan Ma
- School of Basic Medical Sciences, MOE Frontiers Center for Brain Science, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Pharmacology Research Center, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China
- Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China
| | - Fei-fei Wang
- School of Basic Medical Sciences, MOE Frontiers Center for Brain Science, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Pharmacology Research Center, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China
- Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China
| |
Collapse
|
20
|
Blaess S, Krabbe S. Cell type specificity for circuit output in the midbrain dopaminergic system. Curr Opin Neurobiol 2023; 83:102811. [PMID: 37972537 DOI: 10.1016/j.conb.2023.102811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 09/14/2023] [Accepted: 10/19/2023] [Indexed: 11/19/2023]
Abstract
Midbrain dopaminergic neurons are a relatively small group of neurons in the mammalian brain controlling a wide range of behaviors. In recent years, increasingly sophisticated tracing, imaging, transcriptomic, and machine learning approaches have provided substantial insights into the anatomical, molecular, and functional heterogeneity of dopaminergic neurons. Despite this wealth of new knowledge, it remains unclear whether and how the diverse features defining dopaminergic subclasses converge to delineate functional ensembles within the dopaminergic system. Here, we review recent studies investigating various aspects of dopaminergic heterogeneity and discuss how development, behavior, and disease influence subtype characteristics. We then outline what further approaches could be pursued to gain a more inclusive picture of dopaminergic diversity, which could be crucial to understanding the functional architecture of this system.
Collapse
Affiliation(s)
- Sandra Blaess
- Neurodevelopmental Genetics, Institute of Reconstructive Neurobiology, Medical Faculty, University of Bonn, 53127 Bonn, Germany.
| | - Sabine Krabbe
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany.
| |
Collapse
|
21
|
Stetsenko A, Koos T. Neuronal implementation of the temporal difference learning algorithm in the midbrain dopaminergic system. Proc Natl Acad Sci U S A 2023; 120:e2309015120. [PMID: 37903252 PMCID: PMC10636325 DOI: 10.1073/pnas.2309015120] [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: 05/29/2023] [Accepted: 09/29/2023] [Indexed: 11/01/2023] Open
Abstract
The temporal difference learning (TDL) algorithm has been essential to conceptualizing the role of dopamine in reinforcement learning (RL). Despite its theoretical importance, it remains unknown whether a neuronal implementation of this algorithm exists in the brain. Here, we provide an interpretation of the recently described signaling properties of ventral tegmental area (VTA) GABAergic neurons and show that a circuitry of these neurons implements the TDL algorithm. Specifically, we identified the neuronal mechanism of three key components of the TDL model: a sustained state value signal encoded by an afferent input to the VTA, a temporal differentiation circuit formed by two types of VTA GABAergic neurons the combined output of which computes momentary reward prediction (RP) as the derivative of the state value, and the computation of reward prediction errors (RPEs) in dopamine neurons utilizing the output of the differentiation circuit. Using computational methods, we also show that this mechanism is optimally adapted to the biophysics of RPE signaling in dopamine neurons, mechanistically links the emergence of conditioned reinforcement to RP, and can naturally account for the temporal discounting of reinforcement. Elucidating the implementation of the TDL algorithm may further the investigation of RL in biological and artificial systems.
Collapse
Affiliation(s)
- Anya Stetsenko
- Center for Molecular and Behavioral Neuroscience, Rutgers University, Newark, NJ07102
| | - Tibor Koos
- Center for Molecular and Behavioral Neuroscience, Rutgers University, Newark, NJ07102
| |
Collapse
|
22
|
Tsou JH, Lee SR, Chiang CY, Yang YJ, Guo FY, Ni SY, Yau HJ. Negative Emotions Recruit the Parabrachial Nucleus Efferent to the VTA to Disengage Instrumental Food Seeking. J Neurosci 2023; 43:7276-7293. [PMID: 37684032 PMCID: PMC10621778 DOI: 10.1523/jneurosci.2114-22.2023] [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: 11/14/2022] [Revised: 08/14/2023] [Accepted: 08/29/2023] [Indexed: 09/10/2023] Open
Abstract
The parabrachial nucleus (PBN) interfaces between taste and feeding systems and is also an important hub for relaying distress information and threats. Despite that the PBN sends projections to the ventral tegmental area (VTA), a heterogeneous brain region that regulates motivational behaviors, the function of the PBN-to-VTA connection remains elusive. Here, by using male mice in several behavioral paradigms, we discover that VTA-projecting PBN neurons are significantly engaged in contextual fear, restraint or mild stress but not palatable feeding, visceral malaise, or thermal pain. These results suggest that the PBN-to-VTA input may relay negative emotions under threat. Consistent with this notion, optogenetic activation of PBN-to-VTA glutamatergic input results in aversion, which is sufficient to override palatable feeding. Moreover, in a palatable food-reinforced operant task, we demonstrate that transient optogenetic activation of PBN-to-VTA input during food reward retrieval disengages instrumental food-seeking behaviors but spares learned action-outcome association. By using an activity-dependent targeting approach, we show that VTA DA neurons are disengaged by the PBN afferent activation, implicating that VTA non-DA neurons may mediate PBN afferent regulation. We further show that optogenetic activation of VTA neurons functionally recruited by the PBN input results in aversion, dampens palatable feeding, and disengages palatable food self-administration behavior. Finally, we demonstrate that transient activation of VTA glutamatergic, but not GABAergic, neurons recapitulates the negative regulation of the PBN input on food self-administration behavior. Together, we reveal that the PBN-to-VTA input conveys negative affect, likely through VTA glutamatergic neurons, to disengage instrumental food-seeking behaviors.SIGNIFICANCE STATEMENT The PBN receives multiple inputs and thus is well positioned to route information of various modalities to engage different downstream circuits to attend or respond accordingly. We demonstrate that the PBN-to-VTA input conveys negative affect and then triggers adaptive prioritized responses to address pertinent needs by withholding ongoing behaviors, such as palatable food seeking or intake shown in the present study. It has evolutionary significance because preparing to cope with stressful situations or threats takes priority over food seeking to promote survival. Knowing how appropriate adaptive responses are generated will provide new insights into circuitry mechanisms of various coping behaviors to changing environmental stimuli.
Collapse
Affiliation(s)
- Jen-Hui Tsou
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Synaptic Plasticity Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224
| | - Syun-Ruei Lee
- Laboratory for Neural Circuits and Behaviors, Graduate Institute of Brain and Mind Sciences, National Taiwan University, Taipei 10051, Taiwan
| | - Chia-Ying Chiang
- Laboratory for Neural Circuits and Behaviors, Graduate Institute of Brain and Mind Sciences, National Taiwan University, Taipei 10051, Taiwan
| | - Yi-Jie Yang
- Laboratory for Neural Circuits and Behaviors, Graduate Institute of Brain and Mind Sciences, National Taiwan University, Taipei 10051, Taiwan
| | - Fong-Yi Guo
- Laboratory for Neural Circuits and Behaviors, Graduate Institute of Brain and Mind Sciences, National Taiwan University, Taipei 10051, Taiwan
| | - Shih-Ying Ni
- School of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Hau-Jie Yau
- Laboratory for Neural Circuits and Behaviors, Graduate Institute of Brain and Mind Sciences, National Taiwan University, Taipei 10051, Taiwan
- Neurobiology and Cognitive Science Center, National Taiwan University, Taipei 10617, Taiwan
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Taiwan University and Academia Sinica, Taipei 115, Taiwan
- PhD Program in Translational Medicine, National Taiwan University and Academia Sinica, Taipei 115, Taiwan
| |
Collapse
|
23
|
Cui X, Tong Q, Xu H, Xie C, Xiao L. A putative loop connection between VTA dopamine neurons and nucleus accumbens encodes positive valence to compensate for hunger. Prog Neurobiol 2023; 229:102503. [PMID: 37451329 DOI: 10.1016/j.pneurobio.2023.102503] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 07/08/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
Dopamine (DA) signal play pivotal roles in regulating motivated behaviors, including feeding behavior, but the role of midbrain DA neurons in modulating food intake and neural circuitry mechanisms remain largely unknown. Here, we found that activating but not inhibiting ventral tegmental area (VTA) DA neurons reduces mouse food intake. Furthermore, DA neurons in ventral VTA, especially neurons projecting to the medial nucleus accumbens (NAc), are activated by refeeding in the 24 h fasted mice. Combing neural circuitry tracing, optogenetic, chemogenetic, and pharmacological manipulations, we established that the VTA→medial NAc→VTA loop circuit is critical for the VTA DA neurons activation-induced food intake reduction. Moreover, activating either VTA DA neurons or dopaminergic axons in medial NAc elevates positive valence, which will compensate for the hungry-induced food intake. Thus, our study identifies a subset of positive valence-encoded VTA DA neurons forming possible loop connections with medial NAc that are anorexigenic.
Collapse
Affiliation(s)
- Xiao Cui
- Department of Orthodontics, 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
| | - Qiuping Tong
- Department of Orthodontics, 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
- Department of Orthodontics, 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
| | - Chuantong Xie
- Department of Orthodontics, 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
- Department of Orthodontics, 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
|
24
|
Geisler CE, Hayes MR. Metabolic hormone action in the VTA: Reward-directed behavior and mechanistic insights. Physiol Behav 2023; 268:114236. [PMID: 37178855 PMCID: PMC10330780 DOI: 10.1016/j.physbeh.2023.114236] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/10/2023] [Accepted: 05/10/2023] [Indexed: 05/15/2023]
Abstract
Dysfunctional signaling in midbrain reward circuits perpetuates diseases characterized by compulsive overconsumption of rewarding substances such as substance abuse, binge eating disorder, and obesity. Ventral tegmental area (VTA) dopaminergic activity serves as an index for how rewarding stimuli are perceived and triggers behaviors necessary to obtain future rewards. The evolutionary linking of reward with seeking and consuming palatable foods ensured an organism's survival, and hormone systems that regulate appetite concomitantly developed to regulate motivated behaviors. Today, these same mechanisms serve to regulate reward-directed behavior around food, drugs, alcohol, and social interactions. Understanding how hormonal regulation of VTA dopaminergic output alters motivated behaviors is essential to leveraging therapeutics that target these hormone systems to treat addiction and disordered eating. This review will outline our current understanding of the mechanisms underlying VTA action of the metabolic hormones ghrelin, glucagon-like peptide-1, amylin, leptin, and insulin to regulate behavior around food and drugs of abuse, highlighting commonalities and differences in how these five hormones ultimately modulate VTA dopamine signaling.
Collapse
Affiliation(s)
- Caroline E Geisler
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Matthew R Hayes
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
25
|
Chuhma N, Oh SJ, Rayport S. The dopamine neuron synaptic map in the striatum. Cell Rep 2023; 42:112204. [PMID: 36867530 PMCID: PMC10657204 DOI: 10.1016/j.celrep.2023.112204] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 12/21/2022] [Accepted: 02/16/2023] [Indexed: 03/04/2023] Open
Abstract
Dopamine neurons project to the striatum to control movement, cognition, and motivation via slower volume transmission as well as faster dopamine, glutamate, and GABA synaptic actions capable of conveying the temporal information in dopamine neuron firing. To define the scope of these synaptic actions, recordings of dopamine-neuron-evoked synaptic currents were made in four major striatal neuron types, spanning the entire striatum. This revealed that inhibitory postsynaptic currents are widespread, while excitatory postsynaptic currents are localized to the medial nucleus accumbens and the anterolateral-dorsal striatum, and that all synaptic actions are weak in the posterior striatum. Synaptic actions in cholinergic interneurons are the strongest, variably mediating inhibition throughout the striatum and excitation in the medial accumbens, capable of controlling their activity. This mapping shows that dopamine neuron synaptic actions extend throughout the striatum, preferentially target cholinergic interneurons, and define distinct striatal subregions.
Collapse
Affiliation(s)
- Nao Chuhma
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA; Department of Psychiatry, Columbia University, New York, NY 10032, USA.
| | - Soo Jung Oh
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA; Department of Psychiatry, Columbia University, New York, NY 10032, USA
| | - Stephen Rayport
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA; Department of Psychiatry, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
26
|
Abstract
The midbrain dopamine (mDA) system is composed of molecularly and functionally distinct neuron subtypes that mediate specific behaviours and are linked to various brain diseases. Considerable progress has been made in identifying mDA neuron subtypes, and recent work has begun to unveil how these neuronal subtypes develop and organize into functional brain structures. This progress is important for further understanding the disparate physiological functions of mDA neurons and their selective vulnerability in disease, and will ultimately accelerate therapy development. This Review discusses recent advances in our understanding of molecularly defined mDA neuron subtypes and their circuits, ranging from early developmental events, such as neuron migration and axon guidance, to their wiring and function, and future implications for therapeutic strategies.
Collapse
|
27
|
Soares-Cunha C, Heinsbroek JA. Ventral pallidal regulation of motivated behaviors and reinforcement. Front Neural Circuits 2023; 17:1086053. [PMID: 36817646 PMCID: PMC9932340 DOI: 10.3389/fncir.2023.1086053] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/06/2023] [Indexed: 02/05/2023] Open
Abstract
The interconnected nuclei of the ventral basal ganglia have long been identified as key regulators of motivated behavior, and dysfunction of this circuit is strongly implicated in mood and substance use disorders. The ventral pallidum (VP) is a central node of the ventral basal ganglia, and recent studies have revealed complex VP cellular heterogeneity and cell- and circuit-specific regulation of reward, aversion, motivation, and drug-seeking behaviors. Although the VP is canonically considered a relay and output structure for this circuit, emerging data indicate that the VP is a central hub in an extensive network for reward processing and the regulation of motivation that extends beyond classically defined basal ganglia borders. VP neurons respond temporally faster and show more advanced reward coding and prediction error processing than neurons in the upstream nucleus accumbens, and regulate the activity of the ventral mesencephalon dopamine system. This review will summarize recent findings in the literature and provide an update on the complex cellular heterogeneity and cell- and circuit-specific regulation of motivated behaviors and reinforcement by the VP with a specific focus on mood and substance use disorders. In addition, we will discuss mechanisms by which stress and drug exposure alter the functioning of the VP and produce susceptibility to neuropsychiatric disorders. Lastly, we will outline unanswered questions and identify future directions for studies necessary to further clarify the central role of VP neurons in the regulation of motivated behaviors. Significance: Research in the last decade has revealed a complex cell- and circuit-specific role for the VP in reward processing and the regulation of motivated behaviors. Novel insights obtained using cell- and circuit-specific interrogation strategies have led to a major shift in our understanding of this region. Here, we provide a comprehensive review of the VP in which we integrate novel findings with the existing literature and highlight the emerging role of the VP as a linchpin of the neural systems that regulate motivation, reward, and aversion. In addition, we discuss the dysfunction of the VP in animal models of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Carina Soares-Cunha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Jasper A. Heinsbroek
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
28
|
Martinez Damonte V, Pomrenze MB, Manning CE, Casper C, Wolfden AL, Malenka RC, Kauer JA. Somatodendritic Release of Cholecystokinin Potentiates GABAergic Synapses Onto Ventral Tegmental Area Dopamine Cells. Biol Psychiatry 2023; 93:197-208. [PMID: 35961792 PMCID: PMC9976994 DOI: 10.1016/j.biopsych.2022.06.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 06/01/2022] [Accepted: 06/10/2022] [Indexed: 11/02/2022]
Abstract
BACKGROUND Neuropeptides are contained in nearly every neuron in the central nervous system and can be released not only from nerve terminals but also from somatodendritic sites. Cholecystokinin (CCK), among the most abundant neuropeptides in the brain, is expressed in the majority of midbrain dopamine neurons. Despite this high expression, CCK function within the ventral tegmental area (VTA) is not well understood. METHODS We confirmed CCK expression in VTA dopamine neurons through immunohistochemistry and in situ hybridization and detected optogenetically induced CCK release using an enzyme-linked immunosorbent assay. To investigate whether CCK modulates VTA circuit activity, we used whole-cell patch clamp recordings in mouse brain slices. We infused CCK locally in vivo and tested food intake and locomotion in fasted mice. We also used in vivo fiber photometry to measure Ca2+ transients in dopamine neurons during feeding. RESULTS Here we report that VTA dopamine neurons release CCK from somatodendritic regions, where it triggers long-term potentiation of GABAergic (gamma-aminobutyric acidergic) synapses. The somatodendritic release occurs during trains of optogenetic stimuli or prolonged but modest depolarization and is dependent on synaptotagmin-7 and T-type Ca2+ channels. Depolarization-induced long-term potentiation is blocked by a CCK2 receptor antagonist and mimicked by exogenous CCK. Local infusion of CCK in vivo inhibits food consumption and decreases distance traveled in an open field test. Furthermore, intra-VTA-infused CCK reduced dopamine cell Ca2+ signals during food consumption after an overnight fast and was correlated with reduced food intake. CONCLUSIONS Our experiments introduce somatodendritic neuropeptide release as a previously unknown feedback regulator of VTA dopamine cell excitability and dopamine-related behaviors.
Collapse
|
29
|
Koevoet D, Deschamps PKH, Kenemans JL. Catecholaminergic and cholinergic neuromodulation in autism spectrum disorder: A comparison to attention-deficit hyperactivity disorder. Front Neurosci 2023; 16:1078586. [PMID: 36685234 PMCID: PMC9853424 DOI: 10.3389/fnins.2022.1078586] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 12/15/2022] [Indexed: 01/09/2023] Open
Abstract
Autism spectrum disorder (ASD) is a heterogeneous neurodevelopmental disorder characterized by social impairments and restricted, repetitive behaviors. Treatment of ASD is notoriously difficult and might benefit from identification of underlying mechanisms that overlap with those disturbed in other developmental disorders, for which treatment options are more obvious. One example of the latter is attention-deficit hyperactivity disorder (ADHD), given the efficacy of especially stimulants in treatment of ADHD. Deficiencies in catecholaminergic systems [dopamine (DA), norepinephrine (NE)] in ADHD are obvious targets for stimulant treatment. Recent findings suggest that dysfunction in catecholaminergic systems may also be a factor in at least a subgroup of ASD. In this review we scrutinize the evidence for catecholaminergic mechanisms underlying ASD symptoms, and also include in this analysis a third classic ascending arousing system, the acetylcholinergic (ACh) network. We complement this with a comprehensive review of DA-, NE-, and ACh-targeted interventions in ASD, and an exploratory search for potential treatment-response predictors (biomarkers) in ASD, genetically or otherwise. Based on this review and analysis we propose that (1) stimulant treatment may be a viable option for an ASD subcategory, possibly defined by genetic subtyping; (2) cerebellar dysfunction is pronounced for a relatively small ADHD subgroup but much more common in ASD and in both cases may point toward NE- or ACh-directed intervention; (3) deficiency of the cortical salience network is sizable in subgroups of both disorders, and biomarkers such as eye blink rate and pupillometric data may predict the efficacy of targeting this underlying deficiency via DA, NE, or ACh in both ASD and ADHD.
Collapse
Affiliation(s)
- Damian Koevoet
- Experimental Psychology, Helmholtz Institute, Utrecht University, Utrecht, Netherlands,*Correspondence: Damian Koevoet,
| | - P. K. H. Deschamps
- Department of Psychiatry, University Medical Center Utrecht, Utrecht, Netherlands
| | - J. L. Kenemans
- Experimental Psychology, Helmholtz Institute, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
30
|
D1 receptor-expressing neurons in ventral tegmental area alleviate mouse anxiety-like behaviors via glutamatergic projection to lateral septum. Mol Psychiatry 2023; 28:625-638. [PMID: 36195641 PMCID: PMC9531220 DOI: 10.1038/s41380-022-01809-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 09/18/2022] [Accepted: 09/20/2022] [Indexed: 11/08/2022]
Abstract
Dopamine (DA) acts as a key regulator in controlling emotion, and dysfunction of DA signal has been implicated in the pathophysiology of some psychiatric disorders, including anxiety. Ventral tegmental area (VTA) is one of main regions with DA-producing neurons. VTA DAergic projections in mesolimbic brain regions play a crucial role in regulating anxiety-like behaviors, however, the function of DA signal within VTA in regulating emotion remains unclear. Here, we observe that pharmacological activation/inhibition of VTA D1 receptors will alleviate/aggravate mouse anxiety-like behaviors, and knockdown of VTA D1 receptor expression also exerts anxiogenic effect. With fluorescence in situ hybridization and electrophysiological recording, we find that D1 receptors are functionally expressed in VTA neurons. Silencing/activating VTA D1 neurons bidirectionally modulate mouse anxiety-like behaviors. Furthermore, knocking down D1 receptors in VTA DA and glutamate neurons elevates anxiety-like state, but in GABA neurons has the opposite effect. In addition, we identify the glutamatergic projection from VTA D1 neurons to lateral septum is mainly responsible for the anxiolytic effect induced by activating VTA D1 neurons. Thus, our study not only characterizes the functional expression of D1 receptors in VTA neurons, but also uncovers the pivotal role of DA signal within VTA in mediating anxiety-like behaviors.
Collapse
|
31
|
Yoshida J, Oñate M, Khatami L, Vera J, Nadim F, Khodakhah K. Cerebellar Contributions to the Basal Ganglia Influence Motor Coordination, Reward Processing, and Movement Vigor. J Neurosci 2022; 42:8406-8415. [PMID: 36351826 PMCID: PMC9665921 DOI: 10.1523/jneurosci.1535-22.2022] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 08/30/2022] [Accepted: 08/30/2022] [Indexed: 11/17/2022] Open
Abstract
Both the cerebellum and the basal ganglia are known for their roles in motor control and motivated behavior. These two systems have been classically considered as independent structures that coordinate their contributions to behavior via separate cortico-thalamic loops. However, recent evidence demonstrates the presence of a rich set of direct connections between these two regions. Although there is strong evidence for connections in both directions, for brevity we limit our discussion to the better-characterized connections from the cerebellum to the basal ganglia. We review two sets of such connections: disynaptic projections through the thalamus and direct monosynaptic projections to the midbrain dopaminergic nuclei, the VTA and the SNc. In each case, we review the evidence for these pathways from anatomic tracing and physiological recordings, and discuss their potential functional roles. We present evidence that the disynaptic pathway through the thalamus is involved in motor coordination, and that its dysfunction contributes to motor deficits, such as dystonia. We then discuss how cerebellar projections to the VTA and SNc influence dopamine release in the respective targets of these nuclei: the NAc and the dorsal striatum. We argue that the cerebellar projections to the VTA may play a role in reward-based learning and therefore contribute to addictive behavior, whereas the projection to the SNc may contribute to movement vigor. Finally, we speculate how these projections may explain many of the observations that indicate a role for the cerebellum in mental disorders, such as schizophrenia.
Collapse
Affiliation(s)
- Junichi Yoshida
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Maritza Oñate
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Leila Khatami
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Jorge Vera
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Farzan Nadim
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461
- Department of Biological Sciences, New Jersey Institute of Technology, Newark, New Jersey, 07102
| | - Kamran Khodakhah
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461
| |
Collapse
|
32
|
Manning CE, Fritz M, Kauer JA. Function of Excitatory Periaqueductal Gray Synapses in the Ventral Tegmental Area following Inflammatory Injury. eNeuro 2022; 9:ENEURO.0324-22.2022. [PMID: 36635253 PMCID: PMC9797208 DOI: 10.1523/eneuro.0324-22.2022] [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: 08/13/2022] [Revised: 10/26/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022] Open
Abstract
Manipulating the activity of ventral tegmental area (VTA) dopamine (DA) neurons can drive nocifensive reflexes, and their firing rates are reduced following noxious stimuli. However, the pain-relevant inputs to the VTA remain incompletely understood. In this study, we used male and female mice in combination with identified dopamine and GABA neurons in the VTA that receive excitatory inputs from the periaqueductal gray (PAG), a nexus of ascending pain information. We tested whether PAG-VTA synapses undergo functional plasticity in response to a pain model using optical stimulation in conjunction with slice electrophysiology. We found that acute carrageenan inflammation does not significantly affect the strength of excitatory PAG synapses onto VTA DA neurons. However, at the PAG synapses on VTA GABA neurons, the subunit composition of NMDA receptors is altered; the complement of NR2D subunits at synaptic sites appears to be lost. Thus, our data support a model in which injury initially alters synapses on VTA GABA neurons. Over time, these alterations may increase tonic inhibition of VTA DA neurons leading to their reduced firing.
Collapse
Affiliation(s)
- Claire Elena Manning
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California 94305-5101
| | - Michael Fritz
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California 94305-5101
| | - Julie Ann Kauer
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California 94305-5101
| |
Collapse
|
33
|
van Hoogstraten WS, Lute MCC, Nusselder H, Kros L, van den Maagdenberg AMJM, De Zeeuw CI. cATR Tracing Approach to Identify Individual Intermediary Neurons Based on Their Input and Output: A Proof-of-Concept Study Connecting Cerebellum and Central Hubs Implicated in Developmental Disorders. Cells 2022; 11:cells11192978. [PMID: 36230940 PMCID: PMC9562212 DOI: 10.3390/cells11192978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/19/2022] [Accepted: 09/21/2022] [Indexed: 11/25/2022] Open
Abstract
Over the past decades, it has become increasingly clear that many neurodevelopmental disorders can be characterized by aberrations in the neuro-anatomical connectome of intermediary hubs. Yet, despite the advent in unidirectional transsynaptic tracing technologies, we are still lacking an efficient approach to identify individual neurons based on both their precise input and output relations, hampering our ability to elucidate the precise connectome in both the healthy and diseased condition. Here, we bridge this gap by combining anterograde transsynaptic- and retrograde (cATR) tracing in Ai14 reporter mice, using adeno-associated virus serotype 1 expressing Cre and cholera toxin subunit B as the anterograde and retrograde tracer, respectively. We have applied this innovative approach to selectively identify individual neurons in the brainstem that do not only receive input from one or more of the cerebellar nuclei (CN), but also project to the primary motor cortex (M1), the amygdala or the ventral tegmental area (VTA). Cells directly connecting CN to M1 were found mainly in the thalamus, while a large diversity of midbrain and brainstem areas connected the CN to the amygdala or VTA. Our data highlight that cATR allows for specific, yet brain-wide, identification of individual neurons that mediate information from a cerebellar nucleus to the cerebral cortex, amygdala or VTA via a disynaptic pathway. Given that the identified neurons in healthy subjects can be readily quantified, our data also form a solid foundation to make numerical comparisons with mouse mutants suffering from aberrations in their connectome due to a neurodevelopmental disorder.
Collapse
Affiliation(s)
| | - Marit C. C. Lute
- Department of Neuroscience, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands
| | - Hugo Nusselder
- Department of Neuroscience, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands
| | - Lieke Kros
- Department of Neuroscience, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands
| | - Arn M. J. M. van den Maagdenberg
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Department of Neurology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Chris I. De Zeeuw
- Department of Neuroscience, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands
- Netherlands Institute for Neuroscience, NIN-KNAW, 1105 BA Amsterdam, The Netherlands
- Correspondence:
| |
Collapse
|
34
|
Fifel K, El Farissi A, Cherasse Y, Yanagisawa M. Motivational and Valence-Related Modulation of Sleep/Wake Behavior are Mediated by Midbrain Dopamine and Uncoupled from the Homeostatic and Circadian Processes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200640. [PMID: 35794435 PMCID: PMC9403635 DOI: 10.1002/advs.202200640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 05/19/2022] [Indexed: 06/15/2023]
Abstract
Motivation and its hedonic valence are powerful modulators of sleep/wake behavior, yet its underlying mechanism is still poorly understood. Given the well-established role of midbrain dopamine (mDA) neurons in encoding motivation and emotional valence, here, neuronal mechanisms mediating sleep/wake regulation are systematically investigated by DA neurotransmission. It is discovered that mDA mediates the strong modulation of sleep/wake states by motivational valence. Surprisingly, this modulation can be uncoupled from the classically employed measures of circadian and homeostatic processes of sleep regulation. These results establish the experimental foundation for an additional new factor of sleep regulation. Furthermore, an electroencephalographic marker during wakefulness at the theta range is identified that can be used to reliably track valence-related modulation of sleep. Taken together, this study identifies mDA signaling as an important neural substrate mediating sleep modulation by motivational valence.
Collapse
Affiliation(s)
- Karim Fifel
- International Institute for Integrative Sleep Medicine (WPI‐IIIS)University of TsukubaTsukubaIbaraki305‐8577Japan
| | - Amina El Farissi
- International Institute for Integrative Sleep Medicine (WPI‐IIIS)University of TsukubaTsukubaIbaraki305‐8577Japan
| | - Yoan Cherasse
- International Institute for Integrative Sleep Medicine (WPI‐IIIS)University of TsukubaTsukubaIbaraki305‐8577Japan
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine (WPI‐IIIS)University of TsukubaTsukubaIbaraki305‐8577Japan
| |
Collapse
|
35
|
Souza R, Bueno D, Lima LB, Muchon MJ, Gonçalves L, Donato J, Shammah-Lagnado SJ, Metzger M. Top-down projections of the prefrontal cortex to the ventral tegmental area, laterodorsal tegmental nucleus, and median raphe nucleus. Brain Struct Funct 2022; 227:2465-2487. [DOI: 10.1007/s00429-022-02538-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 07/05/2022] [Indexed: 11/30/2022]
|
36
|
Hui M, Beier KT. Defining the interconnectivity of the medial prefrontal cortex and ventral midbrain. Front Mol Neurosci 2022; 15:971349. [PMID: 35935333 PMCID: PMC9354837 DOI: 10.3389/fnmol.2022.971349] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 07/05/2022] [Indexed: 11/21/2022] Open
Abstract
Dysfunction in dopamine (DA) signaling contributes to neurological disorders ranging from drug addiction and schizophrenia to depression and Parkinson’s Disease. How might impairment of one neurotransmitter come to effect these seemingly disparate diseases? One potential explanation is that unique populations of DA-releasing cells project to separate brain regions that contribute to different sets of behaviors. Though dopaminergic cells themselves are spatially restricted to the midbrain and constitute a relatively small proportion of all neurons, their projections influence many brain regions. DA is particularly critical for the activity and function of medial prefrontal cortical (mPFC) ensembles. The midbrain and mPFC exhibit reciprocal connectivity – the former innervates the mPFC, and in turn, the mPFC projects back to the midbrain. Viral mapping studies have helped elucidate the connectivity within and between these regions, which likely have broad implications for DA-dependent behaviors. In this review, we discuss advancements in our understanding of the connectivity between the mPFC and midbrain DA system, focusing primarily on rodent models.
Collapse
Affiliation(s)
- May Hui
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, United States
| | - Kevin T. Beier
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, United States
- Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, United States
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, United States
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA, United States
- UCI Mind, University of California, Irvine, Irvine, CA, United States
- *Correspondence: Kevin T. Beier,
| |
Collapse
|
37
|
Zhang J, Sun Z, Duan F, Shi L, Zhang Y, Solé‐Casals J, Caiafa CF. Cerebral cortex layer segmentation using diffusion magnetic resonance imaging in vivo with applications to laminar connections and working memory analysis. Hum Brain Mapp 2022; 43:5220-5234. [PMID: 35778791 PMCID: PMC9812233 DOI: 10.1002/hbm.25998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 06/10/2022] [Accepted: 06/15/2022] [Indexed: 01/15/2023] Open
Abstract
Understanding the laminar brain structure is of great help in further developing our knowledge of the functions of the brain. However, since most layer segmentation methods are invasive, it is difficult to apply them to the human brain in vivo. To systematically explore the human brain's laminar structure noninvasively, the K-means clustering algorithm was used to automatically segment the left hemisphere into two layers, the superficial and deep layers, using a 7 Tesla (T) diffusion magnetic resonance imaging (dMRI)open dataset. The obtained layer thickness was then compared with the layer thickness of the BigBrain reference dataset, which segmented the neocortex into six layers based on the von Economo atlas. The results show a significant correlation not only between our automatically segmented superficial layer thickness and the thickness of layers 1-3 from the reference histological data, but also between our automatically segmented deep layer thickness and the thickness of layers 4-6 from the reference histological data. Second, we constructed the laminar connections between two pairs of unidirectional connected regions, which is consistent with prior research. Finally, we conducted the laminar analysis of the working memory, which was challenging to do in the past, and explained the conclusions of the functional analysis. Our work successfully demonstrates that it is possible to segment the human cortex noninvasively into layers using dMRI data and further explores the mechanisms of the human brain.
Collapse
Affiliation(s)
- Jie Zhang
- College of Artificial IntelligenceNankai UniversityTianjinChina
| | - Zhe Sun
- Computational Engineering Applications UnitHead Office for Information Systems and Cybersecurity, RIKENSaitamaJapan
| | - Feng Duan
- College of Artificial IntelligenceNankai UniversityTianjinChina
| | - Liang Shi
- College of Artificial IntelligenceNankai UniversityTianjinChina
| | - Yu Zhang
- Department of Bioengineering and Department of Electrical and Computer EngineeringLehigh UniversityBethlehemPennsylvaniaUSA
| | - Jordi Solé‐Casals
- College of Artificial IntelligenceNankai UniversityTianjinChina,Department of PsychiatryUniversity of CambridgeCambridgeUK,Data and Signal Processing Research GroupUniversity of Vic‐Central University of CataloniaVicCataloniaSpain
| | - Cesar F. Caiafa
- College of Artificial IntelligenceNankai UniversityTianjinChina,Instituto Argentino de Radioastronomía‐ CCT La Plata, CONICET/CIC‐PBA/UNLP, 1894 V.ElisaArgentina
| |
Collapse
|
38
|
Beier K. Modified viral-genetic mapping reveals local and global connectivity relationships of ventral tegmental area dopamine cells. eLife 2022; 11:e76886. [PMID: 35604019 PMCID: PMC9173742 DOI: 10.7554/elife.76886] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 05/20/2022] [Indexed: 11/13/2022] Open
Abstract
Dopamine cells in the ventral tegmental area (VTADA) are critical for a variety of motivated behaviors. These cells receive synaptic inputs from over 100 anatomically defined brain regions, which enables control from a distributed set of inputs across the brain. Extensive efforts have been made to map inputs to VTA cells based on neurochemical phenotype and output site. However, all of these studies have the same fundamental limitation that inputs local to the VTA cannot be properly assessed due to non-Cre-dependent uptake of EnvA-pseudotyped virus. Therefore, the quantitative contribution of local inputs to the VTA, including GABAergic, DAergic, and serotonergic, is not known. Here, I used a modified viral-genetic strategy that enables examination of both local and long-range inputs to VTADA cells in mice. I found that nearly half of the total inputs to VTADA cells are located locally, revealing a substantial portion of inputs that have been missed by previous analyses. The majority of inhibition to VTADA cells arises from the substantia nigra pars reticulata, with large contributions from the VTA and the substantia nigra pars compacta. In addition to receiving inputs from VTAGABA neurons, DA neurons are connected with other DA neurons within the VTA as well as the nearby retrorubal field. Lastly, I show that VTADA neurons receive inputs from distributed serotonergic neurons throughout the midbrain and hindbrain, with the majority arising from the dorsal raphe. My study highlights the importance of using the appropriate combination of viral-genetic reagents to unmask the complexity of connectivity relationships to defined cells in the brain.
Collapse
Affiliation(s)
- Kevin Beier
- Department of Physiology and Biophysics, Neurobiology and Behavior, Biomedical Engineering, Pharmaceutical Sciences, Center for the Neurobiology of Learning and Memory, University of California, IrvineIrvineUnited States
| |
Collapse
|
39
|
Cai J, Tong Q. Anatomy and Function of Ventral Tegmental Area Glutamate Neurons. Front Neural Circuits 2022; 16:867053. [PMID: 35669454 PMCID: PMC9164627 DOI: 10.3389/fncir.2022.867053] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/30/2022] [Indexed: 11/29/2022] Open
Abstract
The ventral tegmental area (VTA) is well known for regulating reward consumption, learning, memory, and addiction behaviors through mediating dopamine (DA) release in downstream regions. Other than DA neurons, the VTA is known to be heterogeneous and contains other types of neurons, including glutamate neurons. In contrast to the well-studied and established functions of DA neurons, the role of VTA glutamate neurons is understudied, presumably due to their relatively small quantity and a lack of effective means to study them. Yet, emerging studies have begun to reveal the importance of glutamate release from VTA neurons in regulating diverse behavioral repertoire through a complex intra-VTA and long-range neuronal network. In this review, we summarize the features of VTA glutamate neurons from three perspectives, namely, cellular properties, neural connections, and behavioral functions. Delineation of VTA glutamatergic pathways and their interactions with VTA DA neurons in regulating behaviors may reveal previously unappreciated functions of the VTA in other physiological processes.
Collapse
Affiliation(s)
- Jing Cai
- Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, UTHealth McGovern Medical School, Houston, TX, United States
- Neuroscience Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
| | - Qingchun Tong
- Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, UTHealth McGovern Medical School, Houston, TX, United States
- Neuroscience Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
| |
Collapse
|
40
|
Tian G, Hui M, Macchia D, Derdeyn P, Rogers A, Hubbard E, Liu C, Vasquez JJ, Taniguchi L, Bartas K, Carroll S, Beier KT. An extended amygdala-midbrain circuit controlling cocaine withdrawal-induced anxiety and reinstatement. Cell Rep 2022; 39:110775. [PMID: 35508124 PMCID: PMC9225486 DOI: 10.1016/j.celrep.2022.110775] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/29/2021] [Accepted: 04/12/2022] [Indexed: 12/20/2022] Open
Abstract
Although midbrain dopamine (DA) circuits are central to motivated behaviors, our knowledge of how experience modifies these circuits to facilitate subsequent behavioral adaptations is limited. Here we demonstrate the selective role of a ventral tegmental area DA projection to the amygdala (VTADA→amygdala) for cocaine-induced anxiety but not cocaine reward or sensitization. Our rabies virus-mediated circuit mapping approach reveals a persistent elevation in spontaneous and task-related activity of inhibitory GABAergic cells from the bed nucleus of the stria terminalis (BNST) and downstream VTADA→amygdala cells that can be detected even after a single cocaine exposure. Activity in BNSTGABA→midbrain cells is related to cocaine-induced anxiety but not reward or sensitization, and silencing this projection prevents development of anxiety during protracted withdrawal after cocaine administration. Finally, we observe that VTADA→amygdala cells are strongly activated after a challenge exposure to cocaine and that activity in these cells is necessary and sufficient for reinstatement of cocaine place preference.
Collapse
Affiliation(s)
- Guilian Tian
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92617, USA
| | - May Hui
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92617, USA
| | - Desiree Macchia
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92617, USA
| | - Pieter Derdeyn
- Program in Mathematical, Computational, and Systems Biology, University of California, Irvine, Irvine, CA 92617, USA
| | - Alexandra Rogers
- Interdepartmental Neuroscience Program, University of California, Irvine, Irvine, CA 92617, USA
| | - Elizabeth Hubbard
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92617, USA
| | - Chengfeng Liu
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92617, USA
| | - Jose J Vasquez
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92617, USA
| | - Lara Taniguchi
- Interdepartmental Neuroscience Program, University of California, Irvine, Irvine, CA 92617, USA
| | - Katrina Bartas
- Program in Mathematical, Computational, and Systems Biology, University of California, Irvine, Irvine, CA 92617, USA
| | - Sean Carroll
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92617, USA
| | - Kevin T Beier
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92617, USA; Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92617, USA; Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92617, USA; Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92617, USA; Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA 92617, USA; UCI Mind, University of California, Irvine, Irvine, CA 92617, USA.
| |
Collapse
|
41
|
Urrutia-Piñones J, Morales-Moraga C, Sanguinetti-González N, Escobar AP, Chiu CQ. Long-Range GABAergic Projections of Cortical Origin in Brain Function. Front Syst Neurosci 2022; 16:841869. [PMID: 35392440 PMCID: PMC8981584 DOI: 10.3389/fnsys.2022.841869] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/10/2022] [Indexed: 12/12/2022] Open
Abstract
The study of long-range GABAergic projections has traditionally been focused on those with subcortical origin. In the last few years, cortical GABAergic neurons have been shown to not only mediate local inhibition, but also extend long-range axons to remote cortical and subcortical areas. In this review, we delineate the different types of long-range GABAergic neurons (LRGNs) that have been reported to arise from the hippocampus and neocortex, paying attention to the anatomical and functional circuits they form to understand their role in behavior. Although cortical LRGNs are similar to their interneuron and subcortical counterparts, they comprise distinct populations that show specific patterns of cortico-cortical and cortico-fugal connectivity. Functionally, cortical LRGNs likely induce timed disinhibition in target regions to synchronize network activity. Thus, LRGNs are emerging as a new element of cortical output, acting in concert with long-range excitatory projections to shape brain function in health and disease.
Collapse
Affiliation(s)
- Jocelyn Urrutia-Piñones
- Ph.D. Program in Neuroscience, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Facultad de Ciencias, Instituto de Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
| | - Camila Morales-Moraga
- Facultad de Ciencias, Instituto de Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| | - Nicole Sanguinetti-González
- Ph.D. Program in Neuroscience, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Facultad de Ciencias, Instituto de Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
| | - Angelica P. Escobar
- Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Centro de Neurobiología y Fisiopatología Integrativa, Universidad de Valparaíso, Valparaíso, Chile
| | - Chiayu Q. Chiu
- Facultad de Ciencias, Instituto de Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
42
|
Hasegawa E, Miyasaka A, Sakurai K, Cherasse Y, Li Y, Sakurai T. Rapid eye movement sleep is initiated by basolateral amygdala dopamine signaling in mice. Science 2022; 375:994-1000. [PMID: 35239361 DOI: 10.1126/science.abl6618] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The sleep cycle is characterized by alternating non-rapid eye movement (NREM) and rapid eye movement (REM) sleeps. The mechanisms by which this cycle is generated are incompletely understood. We found that a transient increase of dopamine (DA) in the basolateral amygdala (BLA) during NREM sleep terminates NREM sleep and initiates REM sleep. DA acts on dopamine receptor D2 (Drd2)-expressing neurons in the BLA to induce the NREM-to-REM transition. This mechanism also plays a role in cataplectic attacks-a pathological intrusion of REM sleep into wakefulness-in narcoleptics. These results show a critical role of DA signaling in the BLA in initiating REM sleep and provide a neuronal basis for sleep cycle generation.
Collapse
Affiliation(s)
- Emi Hasegawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
- Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Ai Miyasaka
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Katsuyasu Sakurai
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yoan Cherasse
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China
| | - Takeshi Sakurai
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
- Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
- Life Science Center for Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
43
|
Derdeyn P, Hui M, Macchia D, Beier KT. Uncovering the Connectivity Logic of the Ventral Tegmental Area. Front Neural Circuits 2022; 15:799688. [PMID: 35153681 PMCID: PMC8832514 DOI: 10.3389/fncir.2021.799688] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/14/2021] [Indexed: 11/13/2022] Open
Abstract
Decades of research have revealed the remarkable complexity of the midbrain dopamine (DA) system, which comprises cells principally located in the ventral tegmental area (VTA) and substantia nigra pars compacta (SNc). Neither homogenous nor serving a singular function, the midbrain DA system is instead composed of distinct cell populations that (1) receive different sets of inputs, (2) project to separate forebrain sites, and (3) are characterized by unique transcriptional and physiological signatures. To appreciate how these differences relate to circuit function, we first need to understand the anatomical connectivity of unique DA pathways and how this connectivity relates to DA-dependent motivated behavior. We and others have provided detailed maps of the input-output relationships of several subpopulations of midbrain DA cells and explored the roles of these different cell populations in directing behavioral output. In this study, we analyze VTA inputs and outputs as a high dimensional dataset (10 outputs, 22 inputs), deploying computational techniques well-suited to finding interpretable patterns in such data. In addition to reinforcing our previous conclusion that the connectivity in the VTA is dependent on spatial organization, our analysis also uncovered a set of inputs elevated onto each projection-defined VTADA cell type. For example, VTADA→NAcLat cells receive preferential innervation from inputs in the basal ganglia, while VTADA→Amygdala cells preferentially receive inputs from populations sending a distributed input across the VTA, which happen to be regions associated with the brain's stress circuitry. In addition, VTADA→NAcMed cells receive ventromedially biased inputs including from the preoptic area, ventral pallidum, and laterodorsal tegmentum, while VTADA→mPFC cells are defined by dominant inputs from the habenula and dorsal raphe. We also go on to show that the biased input logic to the VTADA cells can be recapitulated using projection architecture in the ventral midbrain, reinforcing our finding that most input differences identified using rabies-based (RABV) circuit mapping reflect projection archetypes within the VTA.
Collapse
Affiliation(s)
- Pieter Derdeyn
- Program in Mathematical, Computational, and Systems Biology, University of California, Irvine, Irvine, CA, United States
| | - May Hui
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, United States
| | - Desiree Macchia
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, United States
| | - Kevin T. Beier
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, United States
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, United States
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
- Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, United States
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
44
|
Mercer Lindsay N, Chen C, Gilam G, Mackey S, Scherrer G. Brain circuits for pain and its treatment. Sci Transl Med 2021; 13:eabj7360. [PMID: 34757810 DOI: 10.1126/scitranslmed.abj7360] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Nicole Mercer Lindsay
- Department of Cell Biology and Physiology, UNC Neuroscience Center, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Department of Biology, CNC Program, Department of Applied Physics, Stanford University, Stanford, CA 94305, USA
| | - Chong Chen
- Department of Cell Biology and Physiology, UNC Neuroscience Center, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Gadi Gilam
- Division of Pain Medicine, Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Sean Mackey
- Division of Pain Medicine, Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Grégory Scherrer
- Department of Cell Biology and Physiology, UNC Neuroscience Center, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,New York Stem Cell Foundation-Robertson Investigator, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
45
|
Vincent C, Gilabert-Juan J, Gibel-Russo R, Alvarez-Fischer D, Krebs MO, Le Pen G, Prochiantz A, Di Nardo AA. Non-cell-autonomous OTX2 transcription factor regulates anxiety-related behavior in the mouse. Mol Psychiatry 2021; 26:6469-6480. [PMID: 33963285 PMCID: PMC8760049 DOI: 10.1038/s41380-021-01132-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/06/2021] [Accepted: 04/14/2021] [Indexed: 02/03/2023]
Abstract
The OTX2 homeoprotein transcription factor is expressed in the dopaminergic neurons of the ventral tegmental area, which projects to limbic structures controlling complex behaviors. OTX2 is also produced in choroid plexus epithelium, from which it is secreted into cerebrospinal fluid and transferred to limbic structure parvalbumin interneurons. Previously, adult male mice subjected to early-life stress were found susceptible to anxiety-like behaviors, with accompanying OTX2 expression changes in ventral tegmental area or choroid plexus. Here, we investigated the consequences of reduced OTX2 levels in Otx2 heterozygote mice, as well as in Otx2+/AA and scFvOtx2tg/0 mouse models for decreasing OTX2 transfer from choroid plexus to parvalbumin interneurons. Both male and female adult mice show anxiolysis-like phenotypes in all three models. In Otx2 heterozygote mice, we observed no changes in dopaminergic neuron numbers and morphology in ventral tegmental area, nor in their metabolic output and projections to target structures. However, we found reduced expression of parvalbumin in medial prefrontal cortex, which could be rescued in part by adult overexpression of Otx2 specifically in choroid plexus, resulting in increased anxiety-like behavior. Taken together, OTX2 synthesis by the choroid plexus followed by its secretion into the cerebrospinal fluid is an important regulator of anxiety-related phenotypes in the mouse.
Collapse
Affiliation(s)
- Clémentine Vincent
- Centre for Interdisciplinary Research in Biology (CIRB), CNRS UMR 7241, INSERM U1050, Labex MemoLife, PSL Research University, Collège de France, Paris, France
- Institut NeuroMyoGène, CNRS UMR 5310, INSERM U1217, Université Claude Bernard Lyon 1, Lyon, France
| | - Javier Gilabert-Juan
- Centre for Interdisciplinary Research in Biology (CIRB), CNRS UMR 7241, INSERM U1050, Labex MemoLife, PSL Research University, Collège de France, Paris, France
- Department of Anatomy, Histology and Neuroscience, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Rachel Gibel-Russo
- Centre for Interdisciplinary Research in Biology (CIRB), CNRS UMR 7241, INSERM U1050, Labex MemoLife, PSL Research University, Collège de France, Paris, France
| | | | - Marie-Odile Krebs
- Laboratoire de Physiopathologie des Maladies Psychiatriques, INSERM U1266, Institut de Psychiatrie et Neurosciences de Paris, Université de Paris, Paris, France
- Institut de Psychiatrie, CNRS GDR 3557, Paris, France
- Faculté de Médecine, Université de Paris, Pôle Hospitalo-Universitaire Evaluation Prévention et Innovation Thérapeutique, GHU Paris Psychiatrie et Neurosciences site Sainte-Anne, Paris, France
| | - Gwenaëlle Le Pen
- Laboratoire de Physiopathologie des Maladies Psychiatriques, INSERM U1266, Institut de Psychiatrie et Neurosciences de Paris, Université de Paris, Paris, France
| | - Alain Prochiantz
- Centre for Interdisciplinary Research in Biology (CIRB), CNRS UMR 7241, INSERM U1050, Labex MemoLife, PSL Research University, Collège de France, Paris, France.
| | - Ariel A Di Nardo
- Centre for Interdisciplinary Research in Biology (CIRB), CNRS UMR 7241, INSERM U1050, Labex MemoLife, PSL Research University, Collège de France, Paris, France.
| |
Collapse
|
46
|
Islam KUS, Meli N, Blaess S. The Development of the Mesoprefrontal Dopaminergic System in Health and Disease. Front Neural Circuits 2021; 15:746582. [PMID: 34712123 PMCID: PMC8546303 DOI: 10.3389/fncir.2021.746582] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 09/10/2021] [Indexed: 12/18/2022] Open
Abstract
Midbrain dopaminergic neurons located in the substantia nigra and the ventral tegmental area are the main source of dopamine in the brain. They send out projections to a variety of forebrain structures, including dorsal striatum, nucleus accumbens, and prefrontal cortex (PFC), establishing the nigrostriatal, mesolimbic, and mesoprefrontal pathways, respectively. The dopaminergic input to the PFC is essential for the performance of higher cognitive functions such as working memory, attention, planning, and decision making. The gradual maturation of these cognitive skills during postnatal development correlates with the maturation of PFC local circuits, which undergo a lengthy functional remodeling process during the neonatal and adolescence stage. During this period, the mesoprefrontal dopaminergic innervation also matures: the fibers are rather sparse at prenatal stages and slowly increase in density during postnatal development to finally reach a stable pattern in early adulthood. Despite the prominent role of dopamine in the regulation of PFC function, relatively little is known about how the dopaminergic innervation is established in the PFC, whether and how it influences the maturation of local circuits and how exactly it facilitates cognitive functions in the PFC. In this review, we provide an overview of the development of the mesoprefrontal dopaminergic system in rodents and primates and discuss the role of altered dopaminergic signaling in neuropsychiatric and neurodevelopmental disorders.
Collapse
Affiliation(s)
- K Ushna S Islam
- Neurodevelopmental Genetics, Institute of Reconstructive Neurobiology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Norisa Meli
- Neurodevelopmental Genetics, Institute of Reconstructive Neurobiology, Medical Faculty, University of Bonn, Bonn, Germany.,Institute of Neuropathology, Section for Translational Epilepsy Research, Medical Faculty, University of Bonn, Bonn, Germany
| | - Sandra Blaess
- Neurodevelopmental Genetics, Institute of Reconstructive Neurobiology, Medical Faculty, University of Bonn, Bonn, Germany
| |
Collapse
|
47
|
Beier KT. The Serendipity of Viral Trans-Neuronal Specificity: More Than Meets the Eye. Front Cell Neurosci 2021; 15:720807. [PMID: 34671244 PMCID: PMC8521040 DOI: 10.3389/fncel.2021.720807] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 09/07/2021] [Indexed: 12/25/2022] Open
Abstract
Trans-neuronal viruses are frequently used as neuroanatomical tools for mapping neuronal circuits. Specifically, recombinant one-step rabies viruses (RABV) have been instrumental in the widespread application of viral circuit mapping, as these viruses have enabled labs to map the direct inputs onto defined cell populations. Within the neuroscience community, it is widely believed that RABV spreads directly between neurons via synaptic connections, a hypothesis based principally on two observations. First, the virus labels neurons in a pattern consistent with known anatomical connectivity. Second, few glial cells appear to be infected following RABV injections, despite the fact that glial cells are abundant in the brain. However, there is no direct evidence that RABV can actually be transmitted through synaptic connections. Here we review the immunosubversive mechanisms that are critical to RABV’s success for infiltration of the central nervous system (CNS). These include interfering with and ultimately killing migratory T cells while maintaining levels of interferon (IFN) signaling in the brain parenchyma. Finally, we critically evaluate studies that support or are against synaptically-restricted RABV transmission and the implications of viral-host immune responses for RABV transmission in the brain.
Collapse
Affiliation(s)
- Kevin Thomas Beier
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
48
|
Ventral tegmental area GABAergic inhibition of cholinergic interneurons in the ventral nucleus accumbens shell promotes reward reinforcement. Nat Neurosci 2021; 24:1414-1428. [PMID: 34385700 PMCID: PMC8823543 DOI: 10.1038/s41593-021-00898-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 06/24/2021] [Indexed: 02/07/2023]
Abstract
The long-range GABAergic input from the ventral tegmental area (VTA) to the nucleus accumbens (NAc) is relatively understudied, and therefore its role in reward processing has remained unknown. In the present study, we show, in both male and female mice, that long-range GABAergic projections from the VTA to the ventral NAc shell, but not to the dorsal NAc shell or NAc core, are engaged in reward and reinforcement behavior. We show that this GABAergic projection exclusively synapses on to cholinergic interneurons (CINs) in the ventral NAc shell, thereby serving a specialized function in modulating reinforced reward behavior through the inhibition of ventral NAc shell CINs. These findings highlight the diversity in the structural and functional topography of VTA GABAergic projections, and their neuromodulatory interactions across the dorsoventral gradient of the NAc shell. They also further our understanding of neuronal circuits that are directly implicated in neuropsychiatric conditions such as depression and addiction.
Collapse
|
49
|
Morigaki R, Miyamoto R, Matsuda T, Miyake K, Yamamoto N, Takagi Y. Dystonia and Cerebellum: From Bench to Bedside. Life (Basel) 2021; 11:776. [PMID: 34440520 PMCID: PMC8401781 DOI: 10.3390/life11080776] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/20/2021] [Accepted: 07/29/2021] [Indexed: 12/31/2022] Open
Abstract
Dystonia pathogenesis remains unclear; however, findings from basic and clinical research suggest the importance of the interaction between the basal ganglia and cerebellum. After the discovery of disynaptic pathways between the two, much attention has been paid to the cerebellum. Basic research using various dystonia rodent models and clinical studies in dystonia patients continues to provide new pieces of knowledge regarding the role of the cerebellum in dystonia genesis. Herein, we review basic and clinical articles related to dystonia focusing on the cerebellum, and clarify the current understanding of the role of the cerebellum in dystonia pathogenesis. Given the recent evidence providing new hypotheses regarding dystonia pathogenesis, we discuss how the current evidence answers the unsolved clinical questions.
Collapse
Affiliation(s)
- Ryoma Morigaki
- Department of Advanced Brain Research, Institute of Biomedical Sciences, Graduate School of Medicine, Tokushima University, Tokushima 770-8501, Japan; (N.Y.); (Y.T.)
- Department of Neurosurgery, Institute of Biomedical Sciences, Graduate School of Medicine, Tokushima University, Tokushima 770-8501, Japan; (T.M.); (K.M.)
| | - Ryosuke Miyamoto
- Department of Neurology, Institute of Biomedical Sciences, Graduate School of Medicine, Tokushima University, Tokushima 770-8501, Japan;
| | - Taku Matsuda
- Department of Neurosurgery, Institute of Biomedical Sciences, Graduate School of Medicine, Tokushima University, Tokushima 770-8501, Japan; (T.M.); (K.M.)
| | - Kazuhisa Miyake
- Department of Neurosurgery, Institute of Biomedical Sciences, Graduate School of Medicine, Tokushima University, Tokushima 770-8501, Japan; (T.M.); (K.M.)
| | - Nobuaki Yamamoto
- Department of Advanced Brain Research, Institute of Biomedical Sciences, Graduate School of Medicine, Tokushima University, Tokushima 770-8501, Japan; (N.Y.); (Y.T.)
- Department of Neurology, Institute of Biomedical Sciences, Graduate School of Medicine, Tokushima University, Tokushima 770-8501, Japan;
| | - Yasushi Takagi
- Department of Advanced Brain Research, Institute of Biomedical Sciences, Graduate School of Medicine, Tokushima University, Tokushima 770-8501, Japan; (N.Y.); (Y.T.)
- Department of Neurosurgery, Institute of Biomedical Sciences, Graduate School of Medicine, Tokushima University, Tokushima 770-8501, Japan; (T.M.); (K.M.)
| |
Collapse
|
50
|
Nguyen C, Mondoloni S, Le Borgne T, Centeno I, Come M, Jehl J, Solié C, Reynolds LM, Durand-de Cuttoli R, Tolu S, Valverde S, Didienne S, Hannesse B, Fiancette JF, Pons S, Maskos U, Deroche-Gamonet V, Dalkara D, Hardelin JP, Mourot A, Marti F, Faure P. Nicotine inhibits the VTA-to-amygdala dopamine pathway to promote anxiety. Neuron 2021; 109:2604-2615.e9. [PMID: 34242565 DOI: 10.1016/j.neuron.2021.06.013] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 03/27/2021] [Accepted: 06/09/2021] [Indexed: 11/28/2022]
Abstract
Nicotine stimulates dopamine (DA) neurons of the ventral tegmental area (VTA) to establish and maintain reinforcement. Nicotine also induces anxiety through an as yet unknown circuitry. We found that nicotine injection drives opposite functional responses of two distinct populations of VTA DA neurons with anatomically segregated projections: it activates neurons that project to the nucleus accumbens (NAc), whereas it inhibits neurons that project to the amygdala nuclei (Amg). We further show that nicotine mediates anxiety-like behavior by acting on β2-subunit-containing nicotinic acetylcholine receptors of the VTA. Finally, using optogenetics, we bidirectionally manipulate the VTA-NAc and VTA-Amg pathways to dissociate their contributions to anxiety-like behavior. We show that inhibition of VTA-Amg DA neurons mediates anxiety-like behavior, while their activation prevents the anxiogenic effects of nicotine. These distinct subpopulations of VTA DA neurons with opposite responses to nicotine may differentially drive the anxiogenic and the reinforcing effects of nicotine.
Collapse
Affiliation(s)
- Claire Nguyen
- ESPCI, Laboratoire de plasticité du cerveau UMR8249, 10 rue Vauquelin, 75005 Paris, France; Sorbonne Université, Inserm, UMR8246 CNRS, Neuroscience Paris Seine - IBPS, 75005 Paris, France
| | - Sarah Mondoloni
- Sorbonne Université, Inserm, UMR8246 CNRS, Neuroscience Paris Seine - IBPS, 75005 Paris, France
| | - Tinaïg Le Borgne
- ESPCI, Laboratoire de plasticité du cerveau UMR8249, 10 rue Vauquelin, 75005 Paris, France; Sorbonne Université, Inserm, UMR8246 CNRS, Neuroscience Paris Seine - IBPS, 75005 Paris, France
| | - Ines Centeno
- Sorbonne Université, Inserm, UMR8246 CNRS, Neuroscience Paris Seine - IBPS, 75005 Paris, France
| | - Maxime Come
- ESPCI, Laboratoire de plasticité du cerveau UMR8249, 10 rue Vauquelin, 75005 Paris, France; Sorbonne Université, Inserm, UMR8246 CNRS, Neuroscience Paris Seine - IBPS, 75005 Paris, France
| | - Joachim Jehl
- ESPCI, Laboratoire de plasticité du cerveau UMR8249, 10 rue Vauquelin, 75005 Paris, France; Sorbonne Université, Inserm, UMR8246 CNRS, Neuroscience Paris Seine - IBPS, 75005 Paris, France
| | - Clément Solié
- ESPCI, Laboratoire de plasticité du cerveau UMR8249, 10 rue Vauquelin, 75005 Paris, France; Sorbonne Université, Inserm, UMR8246 CNRS, Neuroscience Paris Seine - IBPS, 75005 Paris, France
| | - Lauren M Reynolds
- ESPCI, Laboratoire de plasticité du cerveau UMR8249, 10 rue Vauquelin, 75005 Paris, France; Sorbonne Université, Inserm, UMR8246 CNRS, Neuroscience Paris Seine - IBPS, 75005 Paris, France
| | | | - Stefania Tolu
- Sorbonne Université, Inserm, UMR8246 CNRS, Neuroscience Paris Seine - IBPS, 75005 Paris, France
| | - Sébastien Valverde
- Sorbonne Université, Inserm, UMR8246 CNRS, Neuroscience Paris Seine - IBPS, 75005 Paris, France
| | - Steve Didienne
- ESPCI, Laboratoire de plasticité du cerveau UMR8249, 10 rue Vauquelin, 75005 Paris, France; Sorbonne Université, Inserm, UMR8246 CNRS, Neuroscience Paris Seine - IBPS, 75005 Paris, France
| | - Bernadette Hannesse
- Sorbonne Université, Inserm, UMR8246 CNRS, Neuroscience Paris Seine - IBPS, 75005 Paris, France
| | - Jean-François Fiancette
- Neurocentre Magendie, Inserm U1215, Université de Bordeaux, 146 rue Léo Saignat, 33077 Bordeaux, France
| | - Stéphanie Pons
- Institut Pasteur, Unité Neurobiologie intégrative des systèmes cholinergiques, Département de neuroscience, 75724 Paris Cedex, France
| | - Uwe Maskos
- Institut Pasteur, Unité Neurobiologie intégrative des systèmes cholinergiques, Département de neuroscience, 75724 Paris Cedex, France
| | - Véronique Deroche-Gamonet
- Neurocentre Magendie, Inserm U1215, Université de Bordeaux, 146 rue Léo Saignat, 33077 Bordeaux, France
| | - Deniz Dalkara
- Sorbonne Université, Inserm, CNRS, Institut de la Vision, Paris, France
| | - Jean-Pierre Hardelin
- ESPCI, Laboratoire de plasticité du cerveau UMR8249, 10 rue Vauquelin, 75005 Paris, France; Sorbonne Université, Inserm, UMR8246 CNRS, Neuroscience Paris Seine - IBPS, 75005 Paris, France
| | - Alexandre Mourot
- ESPCI, Laboratoire de plasticité du cerveau UMR8249, 10 rue Vauquelin, 75005 Paris, France; Sorbonne Université, Inserm, UMR8246 CNRS, Neuroscience Paris Seine - IBPS, 75005 Paris, France
| | - Fabio Marti
- ESPCI, Laboratoire de plasticité du cerveau UMR8249, 10 rue Vauquelin, 75005 Paris, France; Sorbonne Université, Inserm, UMR8246 CNRS, Neuroscience Paris Seine - IBPS, 75005 Paris, France.
| | - Philippe Faure
- ESPCI, Laboratoire de plasticité du cerveau UMR8249, 10 rue Vauquelin, 75005 Paris, France; Sorbonne Université, Inserm, UMR8246 CNRS, Neuroscience Paris Seine - IBPS, 75005 Paris, France.
| |
Collapse
|