51
|
Jo A, Deniz S, Cherian S, Xu J, Futagi D, DeVries SH, Zhu Y. Modular interneuron circuits control motion sensitivity in the mouse retina. Nat Commun 2023; 14:7746. [PMID: 38008788 PMCID: PMC10679153 DOI: 10.1038/s41467-023-43382-0] [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: 05/03/2023] [Accepted: 11/08/2023] [Indexed: 11/28/2023] Open
Abstract
Neural computations arise from highly precise connections between specific types of neurons. Retinal ganglion cells (RGCs) with similar stratification patterns are positioned to receive similar inputs but often display different response properties. In this study, we used intersectional mouse genetics to achieve single-cell type labeling and identified an object motion sensitive (OMS) AC type, COMS-AC(counter-OMS AC). Optogenetic stimulation revealed that COMS-AC makes glycinergic synapses with the OMS-insensitive HD2p-RGC, while chemogenetic inactivation showed that COMS-AC provides inhibitory control to HD2p-RGC during local motion. This local inhibition, combined with the inhibitory drive from TH2-AC during global motion, explains the OMS-insensitive feature of HD2p-RGC. In contrast, COMS-AC fails to make synapses with W3(UHD)-RGC, allowing it to exhibit OMS under the control of VGlut3-AC and TH2-AC. These findings reveal modular interneuron circuits that endow structurally similar RGC types with different responses and present a mechanism for redundancy-reduction in the retina to expand coding capacity.
Collapse
Affiliation(s)
- Andrew Jo
- Departments of Ophthalmology and Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Sercan Deniz
- Departments of Ophthalmology and Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Suraj Cherian
- Departments of Ophthalmology and Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Jian Xu
- Departments of Ophthalmology and Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Daiki Futagi
- Departments of Ophthalmology and Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Steven H DeVries
- Departments of Ophthalmology and Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Yongling Zhu
- Departments of Ophthalmology and Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
52
|
Chen APF, Chen L, Shi KW, Cheng E, Ge S, Xiong Q. Nigrostriatal dopamine modulates the striatal-amygdala pathway in auditory fear conditioning. Nat Commun 2023; 14:7231. [PMID: 37945595 PMCID: PMC10636191 DOI: 10.1038/s41467-023-43066-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 10/31/2023] [Indexed: 11/12/2023] Open
Abstract
The auditory striatum, a sensory portion of the dorsal striatum, plays an essential role in learning and memory. In contrast to its roles and underlying mechanisms in operant conditioning, however, little is known about its contribution to classical auditory fear conditioning. Here, we reveal the function of the auditory striatum in auditory-conditioned fear memory. We find that optogenetically inhibiting auditory striatal neurons impairs fear memory formation, which is mediated through the striatal-amygdala pathway. Using calcium imaging in behaving mice, we find that auditory striatal neuronal responses to conditioned tones potentiate across memory acquisition and expression. Furthermore, nigrostriatal dopaminergic projections plays an important role in modulating conditioning-induced striatal potentiation. Together, these findings demonstrate the existence of a nigro-striatal-amygdala circuit for conditioned fear memory formation and expression.
Collapse
Affiliation(s)
- Allen P F Chen
- Department of Neurobiology and Behavior, SUNY Stony Brook, Stony Brook, NY, 11794, USA
- Medical Scientist Training Program, Renaissance School of Medicine at SUNY Stony Brook, Stony Brook, NY, 11794, USA
| | - Lu Chen
- Department of Neurobiology and Behavior, SUNY Stony Brook, Stony Brook, NY, 11794, USA
| | - Kaiyo W Shi
- Department of Neurobiology and Behavior, SUNY Stony Brook, Stony Brook, NY, 11794, USA
| | - Eileen Cheng
- Department of Neurobiology and Behavior, SUNY Stony Brook, Stony Brook, NY, 11794, USA
- Department of Physiology and Biophysics, SUNY Stony Brook, Stony Brook, NY, 11794, USA
| | - Shaoyu Ge
- Department of Neurobiology and Behavior, SUNY Stony Brook, Stony Brook, NY, 11794, USA
| | - Qiaojie Xiong
- Department of Neurobiology and Behavior, SUNY Stony Brook, Stony Brook, NY, 11794, USA.
| |
Collapse
|
53
|
Jovanoski KD, Duquenoy L, Mitchell J, Kapoor I, Treiber CD, Croset V, Dempsey G, Parepalli S, Cognigni P, Otto N, Felsenberg J, Waddell S. Dopaminergic systems create reward seeking despite adverse consequences. Nature 2023; 623:356-365. [PMID: 37880370 PMCID: PMC10632144 DOI: 10.1038/s41586-023-06671-8] [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: 07/04/2022] [Accepted: 09/22/2023] [Indexed: 10/27/2023]
Abstract
Resource-seeking behaviours are ordinarily constrained by physiological needs and threats of danger, and the loss of these controls is associated with pathological reward seeking1. Although dysfunction of the dopaminergic valuation system of the brain is known to contribute towards unconstrained reward seeking2,3, the underlying reasons for this behaviour are unclear. Here we describe dopaminergic neural mechanisms that produce reward seeking despite adverse consequences in Drosophila melanogaster. Odours paired with optogenetic activation of a defined subset of reward-encoding dopaminergic neurons become cues that starved flies seek while neglecting food and enduring electric shock punishment. Unconstrained seeking of reward is not observed after learning with sugar or synthetic engagement of other dopaminergic neuron populations. Antagonism between reward-encoding and punishment-encoding dopaminergic neurons accounts for the perseverance of reward seeking despite punishment, whereas synthetic engagement of the reward-encoding dopaminergic neurons also impairs the ordinary need-dependent dopaminergic valuation of available food. Connectome analyses reveal that the population of reward-encoding dopaminergic neurons receives highly heterogeneous input, consistent with parallel representation of diverse rewards, and recordings demonstrate state-specific gating and satiety-related signals. We propose that a similar dopaminergic valuation system dysfunction is likely to contribute to maladaptive seeking of rewards by mammals.
Collapse
Affiliation(s)
| | - Lucille Duquenoy
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford, UK
| | - Jessica Mitchell
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford, UK
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Ishaan Kapoor
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford, UK
| | | | - Vincent Croset
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford, UK
- Department of Biosciences, Durham University, Durham, UK
| | - Georgia Dempsey
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford, UK
| | - Sai Parepalli
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford, UK
| | - Paola Cognigni
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford, UK
- Northern Medical Physics and Clinical Engineering, Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, UK
| | - Nils Otto
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford, UK
- Institute of Anatomy and Molecular Neurobiology, Westfälische Wilhelms-University, Münster, Germany
| | - Johannes Felsenberg
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford, UK
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Scott Waddell
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford, UK.
| |
Collapse
|
54
|
Caldwell M, Ayo-Jibunoh V, Mendoza JC, Brimblecombe KR, Reynolds LM, Zhu Jiang XY, Alarcon C, Fiore E, N Tomaio J, Phillips GR, Mingote S, Flores C, Casaccia P, Liu J, Cragg SJ, McCloskey DP, Yetnikoff L. Axo-glial interactions between midbrain dopamine neurons and oligodendrocyte lineage cells in the anterior corpus callosum. Brain Struct Funct 2023; 228:1993-2006. [PMID: 37668732 PMCID: PMC10516790 DOI: 10.1007/s00429-023-02695-y] [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: 06/14/2023] [Accepted: 08/09/2023] [Indexed: 09/06/2023]
Abstract
Oligodendrocyte progenitor cells (OPCs) receive synaptic innervation from glutamatergic and GABAergic axons and can be dynamically regulated by neural activity, resulting in activity-dependent changes in patterns of axon myelination. However, it remains unclear to what extent other types of neurons may innervate OPCs. Here, we provide evidence implicating midbrain dopamine neurons in the innervation of oligodendrocyte lineage cells in the anterior corpus callosum and nearby white matter tracts of male and female adult mice. Dopaminergic axon terminals were identified in the corpus callosum of DAT-Cre mice after injection of an eYFP reporter virus into the midbrain. Furthermore, fast-scan cyclic voltammetry revealed monoaminergic transients in the anterior corpus callosum, consistent with the anatomical findings. Using RNAscope, we further demonstrate that ~ 40% of Olig2 + /Pdfgra + cells and ~ 20% of Olig2 + /Pdgfra- cells in the anterior corpus callosum express Drd1 and Drd2 transcripts. These results suggest that oligodendrocyte lineage cells may respond to dopamine released from midbrain dopamine axons, which could affect myelination. Together, this work broadens our understanding of neuron-glia interactions with important implications for myelin plasticity by identifying midbrain dopamine axons as a potential regulator of corpus callosal oligodendrocyte lineage cells.
Collapse
Affiliation(s)
- Megan Caldwell
- CUNY Neuroscience Collaborative, The Graduate Center, City University of New York, 365 5Th Ave, New York, NY, 10016, USA
- Department of Psychology, College of Staten Island, City University of New York, 2800 Victory Boulevard, Staten Island, NY, 10314, USA
| | - Vanessa Ayo-Jibunoh
- Department of Psychology, College of Staten Island, City University of New York, 2800 Victory Boulevard, Staten Island, NY, 10314, USA
| | - Josue Criollo Mendoza
- Department of Biology, College of Staten Island, City University of New York, 2800 Victory Boulevard, Staten Island, NY, 10314, USA
| | - Katherine R Brimblecombe
- Centre for Integrative Neuroscience, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, OX1 3PT, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Lauren M Reynolds
- Plasticité du Cerveau, CNRS UMR8249, École Supérieure de Physique et de Chimie Industrielles de la Ville de Paris (ESPCI Paris), Paris, France
| | - Xin Yan Zhu Jiang
- Department of Biology, College of Staten Island, City University of New York, 2800 Victory Boulevard, Staten Island, NY, 10314, USA
| | - Colin Alarcon
- Department of Psychology, College of Staten Island, City University of New York, 2800 Victory Boulevard, Staten Island, NY, 10314, USA
| | - Elizabeth Fiore
- Department of Psychology, College of Staten Island, City University of New York, 2800 Victory Boulevard, Staten Island, NY, 10314, USA
| | - Jacquelyn N Tomaio
- CUNY Neuroscience Collaborative, The Graduate Center, City University of New York, 365 5Th Ave, New York, NY, 10016, USA
- Neuroscience Initiative, Advanced Science Research Center, Graduate Center of The City University of New York, New York, NY, USA
| | - Greg R Phillips
- CUNY Neuroscience Collaborative, The Graduate Center, City University of New York, 365 5Th Ave, New York, NY, 10016, USA
- Department of Biology, College of Staten Island, City University of New York, 2800 Victory Boulevard, Staten Island, NY, 10314, USA
- Center for Developmental Neuroscience, College of Staten Island, City University of New York, 2800 Victory Boulevard, Staten Island, NY, 10314, USA
| | - Susana Mingote
- CUNY Neuroscience Collaborative, The Graduate Center, City University of New York, 365 5Th Ave, New York, NY, 10016, USA
- Neuroscience Initiative, Advanced Science Research Center, Graduate Center of The City University of New York, New York, NY, USA
| | - Cecilia Flores
- Department of Psychiatry and of Neurology and Neuroscience, McGill University, and Douglas Mental Health University Institute, Montreal, QC, Canada
| | - Patrizia Casaccia
- Neuroscience Initiative, Advanced Science Research Center, Graduate Center of The City University of New York, New York, NY, USA
- Department of Neuroscience and Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jia Liu
- Neuroscience Initiative, Advanced Science Research Center, Graduate Center of The City University of New York, New York, NY, USA
| | - Stephanie J Cragg
- Centre for Integrative Neuroscience, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, OX1 3PT, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Dan P McCloskey
- CUNY Neuroscience Collaborative, The Graduate Center, City University of New York, 365 5Th Ave, New York, NY, 10016, USA
- Department of Psychology, College of Staten Island, City University of New York, 2800 Victory Boulevard, Staten Island, NY, 10314, USA
| | - Leora Yetnikoff
- CUNY Neuroscience Collaborative, The Graduate Center, City University of New York, 365 5Th Ave, New York, NY, 10016, USA.
- Department of Psychology, College of Staten Island, City University of New York, 2800 Victory Boulevard, Staten Island, NY, 10314, USA.
| |
Collapse
|
55
|
Lançon K, Séguéla P. Dysregulated neuromodulation in the anterior cingulate cortex in chronic pain. Front Pharmacol 2023; 14:1289218. [PMID: 37954846 PMCID: PMC10634228 DOI: 10.3389/fphar.2023.1289218] [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: 09/05/2023] [Accepted: 10/09/2023] [Indexed: 11/14/2023] Open
Abstract
Chronic pain is a significant global socioeconomic burden with limited long-term treatment options. The intractable nature of chronic pain stems from two primary factors: the multifaceted nature of pain itself and an insufficient understanding of the diverse physiological mechanisms that underlie its initiation and maintenance, in both the peripheral and central nervous systems. The development of novel non-opioidergic analgesic approaches is contingent on our ability to normalize the dysregulated nociceptive pathways involved in pathological pain processing. The anterior cingulate cortex (ACC) stands out due to its involvement in top-down modulation of pain perception, its abnormal activity in chronic pain conditions, and its contribution to cognitive functions frequently impaired in chronic pain states. Here, we review the roles of the monoamines dopamine (DA), norepinephrine (NE), serotonin (5-HT), and other neuromodulators in controlling the activity of the ACC and how chronic pain alters their signaling in ACC circuits to promote pathological hyperexcitability. Additionally, we discuss the potential of targeting these monoaminergic pathways as a therapeutic strategy for treating the cognitive and affective symptoms associated with chronic pain.
Collapse
Affiliation(s)
| | - Philippe Séguéla
- Department of Neurology and Neurosurgery, Alan Edwards Centre for Research on Pain, Montréal Neurological Institute, McGill University, Montréal, QC, Canada
| |
Collapse
|
56
|
Jain S, Yee AG, Maas J, Gierok S, Xu H, Stansil J, Eriksen J, Nelson AB, Silm K, Ford CP, Edwards RH. Adaptor protein-3 produces synaptic vesicles that release phasic dopamine. Proc Natl Acad Sci U S A 2023; 120:e2309843120. [PMID: 37812725 PMCID: PMC10589613 DOI: 10.1073/pnas.2309843120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 09/06/2023] [Indexed: 10/11/2023] Open
Abstract
The burst firing of midbrain dopamine neurons releases a phasic dopamine signal that mediates reinforcement learning. At many synapses, however, high firing rates deplete synaptic vesicles (SVs), resulting in synaptic depression that limits release. What accounts for the increased release of dopamine by stimulation at high frequency? We find that adaptor protein-3 (AP-3) and its coat protein VPS41 promote axonal dopamine release by targeting vesicular monoamine transporter VMAT2 to the axon rather than dendrites. AP-3 and VPS41 also produce SVs that respond preferentially to high-frequency stimulation, independent of their role in axonal polarity. In addition, conditional inactivation of VPS41 in dopamine neurons impairs reinforcement learning, and this involves a defect in the frequency dependence of release rather than the amount of dopamine released. Thus, AP-3 and VPS41 promote the axonal polarity of dopamine release but enable learning by producing a distinct population of SVs tuned specifically to high firing frequency that confers the phasic release of dopamine.
Collapse
Affiliation(s)
- Shweta Jain
- Department of Physiology, University of California School of Medicine, San Francisco, CA94143
- Department of Neurology, University of California School of Medicine, San Francisco, CA94143
| | - Andrew G. Yee
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO80045
| | - James Maas
- Department of Physiology, University of California School of Medicine, San Francisco, CA94143
- Department of Neurology, University of California School of Medicine, San Francisco, CA94143
| | - Sarah Gierok
- Department of Physiology, University of California School of Medicine, San Francisco, CA94143
- Department of Neurology, University of California School of Medicine, San Francisco, CA94143
| | - Hongfei Xu
- Department of Physiology, University of California School of Medicine, San Francisco, CA94143
- Department of Neurology, University of California School of Medicine, San Francisco, CA94143
| | - Jasmine Stansil
- Department of Neurology, University of California School of Medicine, San Francisco, CA94143
| | - Jacob Eriksen
- Department of Physiology, University of California School of Medicine, San Francisco, CA94143
- Department of Neurology, University of California School of Medicine, San Francisco, CA94143
| | - Alexandra B. Nelson
- Department of Neurology, University of California School of Medicine, San Francisco, CA94143
- Aligning Science Across Parkinson’s Collaborative Research Network, Chevy Chase, MD20815
| | - Katlin Silm
- Department of Physiology, University of California School of Medicine, San Francisco, CA94143
- Department of Neurology, University of California School of Medicine, San Francisco, CA94143
| | - Christopher P. Ford
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO80045
- Aligning Science Across Parkinson’s Collaborative Research Network, Chevy Chase, MD20815
| | - Robert H. Edwards
- Department of Physiology, University of California School of Medicine, San Francisco, CA94143
- Department of Neurology, University of California School of Medicine, San Francisco, CA94143
- Aligning Science Across Parkinson’s Collaborative Research Network, Chevy Chase, MD20815
| |
Collapse
|
57
|
Cobb-Lewis DE, Sansalone L, Khaliq ZM. Contributions of the Sodium Leak Channel NALCN to Pacemaking of Medial Ventral Tegmental Area and Substantia Nigra Dopaminergic Neurons. J Neurosci 2023; 43:6841-6853. [PMID: 37640554 PMCID: PMC10573758 DOI: 10.1523/jneurosci.0930-22.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 08/04/2023] [Accepted: 08/10/2023] [Indexed: 08/31/2023] Open
Abstract
We tested the role of the sodium leak channel, NALCN, in pacemaking of dopaminergic neuron (DAN) subpopulations from adult male and female mice. In situ hybridization revealed NALCN RNA in all DANs, with lower abundance in medial ventral tegmental area (VTA) relative to substantia nigra pars compacta (SNc). Despite lower relative abundance of NALCN, we found that acute pharmacological blockade of NALCN in medial VTA DANs slowed pacemaking by 49.08%. We also examined the electrophysiological properties of projection-defined VTA DAN subpopulations identified by retrograde labeling. Inhibition of NALCN reduced pacemaking in DANs projecting to medial nucleus accumbens (NAc) and others projecting to lateral NAc by 70.74% and 31.98%, respectively, suggesting that NALCN is a primary driver of pacemaking in VTA DANs. In SNc DANs, potentiating NALCN by lowering extracellular calcium concentration speeded pacemaking in wildtype but not NALCN conditional knockout mice, demonstrating functional presence of NALCN. In contrast to VTA DANs, however, pacemaking in SNc DANs was unaffected by inhibition of NALCN. Instead, we found that inhibition of NALCN increased the gain of frequency-current plots at firing frequencies slower than spontaneous firing. Similarly, inhibition of the hyperpolarization-activated cyclic nucleotide-gated (HCN) conductance increased gain but had little effect on pacemaking. Interestingly, simultaneous inhibition of NALCN and HCN resulted in significant reduction in pacemaker rate. Thus, we found NALCN makes substantial contributions to driving pacemaking in VTA DAN subpopulations. In SNc DANs, NALCN is not critical for pacemaking but inhibition of NALCN makes cells more sensitive to hyperpolarizing stimuli.SIGNIFICANCE STATEMENT Pacemaking in midbrain dopaminergic neurons (DAN) relies on multiple subthreshold conductances, including a sodium leak. Whether the sodium leak channel, NALCN, contributes to pacemaking in DANs located in the VTA and the SNc has not yet been determined. Using electrophysiology and pharmacology, we show that NALCN plays a prominent role in driving pacemaking in projection-defined VTA DAN subpopulations. By contrast, pacemaking in SNc neurons does not rely on NALCN. Instead, the presence of NALCN regulates the excitability of SNc DANs by reducing the gain of the neuron's response to inhibitory stimuli. Together, these findings will inform future efforts to obtain DAN subpopulation-specific treatments for use in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Dana E Cobb-Lewis
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892
- Institute for Neuroscience, George Washington University School of Medicine and Health Sciences, Washington, DC 20037
| | - Lorenzo Sansalone
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892
| | - Zayd M Khaliq
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
58
|
Farries MA, Faust TW, Mohebi A, Berke JD. Selective encoding of reward predictions and prediction errors by globus pallidus subpopulations. Curr Biol 2023; 33:4124-4135.e5. [PMID: 37703876 PMCID: PMC10591972 DOI: 10.1016/j.cub.2023.08.042] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/04/2023] [Accepted: 08/15/2023] [Indexed: 09/15/2023]
Abstract
Basal ganglia (BG) circuits help guide and invigorate actions using predictions of future rewards (values). Within the BG, the globus pallidus pars externa (GPe) may play an essential role in aggregating and distributing value information. We recorded from the GPe in unrestrained rats performing both Pavlovian and instrumental tasks to obtain rewards and distinguished neuronal subtypes by their firing properties across the wake/sleep cycle and optogenetic tagging. In both tasks, the parvalbumin-positive (PV+), faster-firing "prototypical" neurons showed strong, sustained modulation by value, unlike other subtypes, including the "arkypallidal" cells that project back to striatum. Furthermore, we discovered that a distinct minority (7%) of GP cells display slower, pacemaker-like firing and encode reward prediction errors (RPEs) almost identically to midbrain dopamine neurons. These cell-specific forms of GPe value representation help define the circuit mechanisms by which the BG contribute to motivation and reinforcement learning.
Collapse
Affiliation(s)
- Michael A Farries
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO 80210, USA
| | - Thomas W Faust
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ali Mohebi
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Joshua D Berke
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Psychiatry and Behavioral Sciences, Neuroscience Graduate Program, Kavli Institute for Fundamental Neuroscience, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
59
|
Buck SA, Rubin SA, Kunkhyen T, Treiber CD, Xue X, Fenno LE, Mabry SJ, Sundar VR, Yang Z, Shah D, Ketchesin KD, Becker-Krail DD, Vasylieva I, Smith MC, Weisel FJ, Wang W, Erickson-Oberg MQ, O’Leary EI, Aravind E, Ramakrishnan C, Kim YS, Wu Y, Quick M, Coleman JA, MacDonald WA, Elbakri R, De Miranda BR, Palladino MJ, McCabe BD, Fish KN, Seney ML, Rayport S, Mingote S, Deisseroth K, Hnasko TS, Awatramani R, Watson AM, Waddell S, Cheetham CEJ, Logan RW, Freyberg Z. Sexually dimorphic mechanisms of VGLUT-mediated protection from dopaminergic neurodegeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.02.560584. [PMID: 37873436 PMCID: PMC10592912 DOI: 10.1101/2023.10.02.560584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Parkinson's disease (PD) targets some dopamine (DA) neurons more than others. Sex differences offer insights, with females more protected from DA neurodegeneration. The mammalian vesicular glutamate transporter VGLUT2 and Drosophila ortholog dVGLUT have been implicated as modulators of DA neuron resilience. However, the mechanisms by which VGLUT2/dVGLUT protects DA neurons remain unknown. We discovered DA neuron dVGLUT knockdown increased mitochondrial reactive oxygen species in a sexually dimorphic manner in response to depolarization or paraquat-induced stress, males being especially affected. DA neuron dVGLUT also reduced ATP biosynthetic burden during depolarization. RNA sequencing of VGLUT+ DA neurons in mice and flies identified candidate genes that we functionally screened to further dissect VGLUT-mediated DA neuron resilience across PD models. We discovered transcription factors modulating dVGLUT-dependent DA neuroprotection and identified dj-1β as a regulator of sex-specific DA neuron dVGLUT expression. Overall, VGLUT protects DA neurons from PD-associated degeneration by maintaining mitochondrial health.
Collapse
Affiliation(s)
- Silas A. Buck
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Sophie A. Rubin
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Tenzin Kunkhyen
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Christoph D. Treiber
- Centre for Neural Circuits & Behaviour, University of Oxford, Oxford OX1 3TA, UK
| | - Xiangning Xue
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15232, USA
| | - Lief E. Fenno
- Departments of Psychiatry and Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA
| | - Samuel J. Mabry
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Varun R. Sundar
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Zilu Yang
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Divia Shah
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Kyle D. Ketchesin
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Darius D. Becker-Krail
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Iaroslavna Vasylieva
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Megan C. Smith
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Florian J. Weisel
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Wenjia Wang
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15232, USA
| | - M. Quincy Erickson-Oberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Emma I. O’Leary
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Eshan Aravind
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Charu Ramakrishnan
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Yoon Seok Kim
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Yanying Wu
- Centre for Neural Circuits & Behaviour, University of Oxford, Oxford OX1 3TA, UK
| | - Matthias Quick
- Department of Psychiatry, Columbia University, New York, NY 10032, USA
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Jonathan A. Coleman
- Department of Structural Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | | | - Rania Elbakri
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Briana R. De Miranda
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Michael J. Palladino
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
- Pittsburgh Institute of Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Brian D. McCabe
- Brain Mind Institute, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Kenneth N. Fish
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Marianne L. Seney
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Stephen Rayport
- Department of Psychiatry, Columbia University, New York, NY 10032, USA
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Susana Mingote
- Department of Psychiatry, Columbia University, New York, NY 10032, USA
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA
- Neuroscience Initiative, Advanced Science Research Center, Graduate Center of The City University of New York, New York, NY 10031, USA
| | - Karl Deisseroth
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Thomas S. Hnasko
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA
- Research Service, VA San Diego Healthcare System, San Diego, CA 92161, USA
| | | | - Alan M. Watson
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Scott Waddell
- Centre for Neural Circuits & Behaviour, University of Oxford, Oxford OX1 3TA, UK
| | | | - Ryan W. Logan
- Department of Psychiatry, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
60
|
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
|
61
|
Azcorra M, Gaertner Z, Davidson C, He Q, Kim H, Nagappan S, Hayes CK, Ramakrishnan C, Fenno L, Kim YS, Deisseroth K, Longnecker R, Awatramani R, Dombeck DA. Unique functional responses differentially map onto genetic subtypes of dopamine neurons. Nat Neurosci 2023; 26:1762-1774. [PMID: 37537242 PMCID: PMC10545540 DOI: 10.1038/s41593-023-01401-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 07/05/2023] [Indexed: 08/05/2023]
Abstract
Dopamine neurons are characterized by their response to unexpected rewards, but they also fire during movement and aversive stimuli. Dopamine neuron diversity has been observed based on molecular expression profiles; however, whether different functions map onto such genetic subtypes remains unclear. In this study, we established that three genetic dopamine neuron subtypes within the substantia nigra pars compacta, characterized by the expression of Slc17a6 (Vglut2), Calb1 and Anxa1, each have a unique set of responses to rewards, aversive stimuli and accelerations and decelerations, and these signaling patterns are highly correlated between somas and axons within subtypes. Remarkably, reward responses were almost entirely absent in the Anxa1+ subtype, which instead displayed acceleration-correlated signaling. Our findings establish a connection between functional and genetic dopamine neuron subtypes and demonstrate that molecular expression patterns can serve as a common framework to dissect dopaminergic functions.
Collapse
Affiliation(s)
- Maite Azcorra
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
- Department of Neurology, Northwestern University, Chicago, IL, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Zachary Gaertner
- Department of Neurology, Northwestern University, Chicago, IL, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Connor Davidson
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Qianzi He
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Hailey Kim
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Shivathmihai Nagappan
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Cooper K Hayes
- Department of Microbiology and Immunology, Northwestern University, Chicago, IL, USA
| | - Charu Ramakrishnan
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, USA
| | - Lief Fenno
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, USA
- Departments of Neuroscience & Psychiatry, The University of Texas at Austin, Austin, TX, USA
| | - Yoon Seok Kim
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, USA
| | - Karl Deisseroth
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, USA
| | - Richard Longnecker
- Department of Microbiology and Immunology, Northwestern University, Chicago, IL, USA
| | - Rajeshwar Awatramani
- Department of Neurology, Northwestern University, Chicago, IL, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| | - Daniel A Dombeck
- Department of Neurobiology, Northwestern University, Evanston, IL, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
62
|
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
|
63
|
Smith DM, Torregrossa MM. The ventral tegmental area dopamine to lateral amygdala projection supports cocaine cue associative learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.22.554187. [PMID: 37662292 PMCID: PMC10473658 DOI: 10.1101/2023.08.22.554187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Learning and memory mechanisms are critically involved in drug craving and relapse. Environmental cues paired with repeated drug use acquire incentive value such that exposure to the cues alone can trigger craving and relapse. The amygdala, particularly the lateral amygdala (LA), underlies cue-related learning processes that assign valence to environmental stimuli including drug-paired cues. Evidence suggests that the ventral tegmental area (VTA) dopamine (DA) projection to the LA participates in encoding reinforcing effects that act as a US in conditioned cue reward-seeking as DA released in the amygdala is important for emotional and behavioral functions. Here we used chemogenetics to manipulate these VTA DA inputs to the LA to determine the role of this projection for acquisition of drug-cue associations and reinstatement of drug-seeking. We found inhibiting DA input to the LA during cocaine self-administration slowed acquisition and weakened the ability of the previously cocaine-paired cue to elicit cocaine-seeking. Conversely, exciting the projection during self-administration boosted the salience of the cocaine-paired cue as indicated by enhanced responding during cue-induced reinstatement. Importantly, interfering with DA input to the LA had no impact on the ability of cocaine to elicit a place preference or induce reinstatement in response to a priming cocaine injection. Overall, we show that manipulation of projections underlying DA signaling in the LA may be useful for developing therapeutic interventions for substance use disorders.
Collapse
|
64
|
Jain S, Yee AG, Maas J, Gierok S, Xu H, Stansil J, Eriksen J, Nelson A, Silm K, Ford CP, Edwards RH. Adaptor Protein-3 Produces Synaptic Vesicles that Release Phasic Dopamine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.07.552338. [PMID: 37609166 PMCID: PMC10441354 DOI: 10.1101/2023.08.07.552338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
The burst firing of midbrain dopamine neurons releases a phasic dopamine signal that mediates reinforcement learning. At many synapses, however, high firing rates deplete synaptic vesicles (SVs), resulting in synaptic depression that limits release. What accounts for the increased release of dopamine by stimulation at high frequency? We find that adaptor protein-3 (AP-3) and its coat protein VPS41 promote axonal dopamine release by targeting vesicular monoamine transporter VMAT2 to the axon rather than dendrites. AP-3 and VPS41 also produce SVs that respond preferentially to high frequency stimulation, independent of their role in axonal polarity. In addition, conditional inactivation of VPS41 in dopamine neurons impairs reinforcement learning, and this involves a defect in the frequency dependence of release rather than the amount of dopamine released. Thus, AP-3 and VPS41 promote the axonal polarity of dopamine release but enable learning by producing a novel population of SVs tuned specifically to high firing frequency that confers the phasic release of dopamine.
Collapse
Affiliation(s)
- Shweta Jain
- Department of Physiology, UCSF School of Medicine, San Francisco USA
- Department of Neurology, UCSF School of Medicine, San Francisco USA
| | - Andrew G. Yee
- Department of Pharmacology, University of Colorado School of Medicine, Aurora USA
| | - James Maas
- Department of Physiology, UCSF School of Medicine, San Francisco USA
- Department of Neurology, UCSF School of Medicine, San Francisco USA
| | - Sarah Gierok
- Department of Physiology, UCSF School of Medicine, San Francisco USA
- Department of Neurology, UCSF School of Medicine, San Francisco USA
| | - Hongfei Xu
- Department of Physiology, UCSF School of Medicine, San Francisco USA
- Department of Neurology, UCSF School of Medicine, San Francisco USA
| | - Jasmine Stansil
- Department of Neurology, UCSF School of Medicine, San Francisco USA
| | - Jacob Eriksen
- Department of Physiology, UCSF School of Medicine, San Francisco USA
- Department of Neurology, UCSF School of Medicine, San Francisco USA
| | - Alexandra Nelson
- Department of Neurology, UCSF School of Medicine, San Francisco USA
| | - Katlin Silm
- Department of Physiology, UCSF School of Medicine, San Francisco USA
- Department of Neurology, UCSF School of Medicine, San Francisco USA
| | - Christopher P. Ford
- Department of Pharmacology, University of Colorado School of Medicine, Aurora USA
| | - Robert H. Edwards
- Department of Physiology, UCSF School of Medicine, San Francisco USA
- Department of Neurology, UCSF School of Medicine, San Francisco USA
| |
Collapse
|
65
|
Altbürger C, Holzhauser J, Driever W. CRISPR/Cas9-based QF2 knock-in at the tyrosine hydroxylase ( th) locus reveals novel th-expressing neuron populations in the zebrafish mid- and hindbrain. Front Neuroanat 2023; 17:1196868. [PMID: 37603776 PMCID: PMC10433395 DOI: 10.3389/fnana.2023.1196868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/30/2023] [Indexed: 08/23/2023] Open
Abstract
Catecholaminergic neuron clusters are among the most conserved neuromodulatory systems in vertebrates, yet some clusters show significant evolutionary dynamics. Because of their disease relevance, special attention has been paid to mammalian midbrain dopaminergic systems, which have important functions in motor control, reward, motivation, and cognitive function. In contrast, midbrain dopaminergic neurons in teleosts were thought to be lost secondarily. Here, we generated a CRISPR/Cas9-based knock-in transgene at the th locus, which allows the expression of the Q-system transcription factor QF2 linked to the Tyrosine hydroxylase open reading frame by an E2A peptide. The QF2 knock-in allele still expresses Tyrosine hydroxylase in catecholaminergic neurons. Coexpression analysis of QF2 driven expression of QUAS fluorescent reporter transgenes and of th mRNA and Th protein revealed that essentially all reporter expressing cells also express Th/th. We also observed a small group of previously unidentified cells expressing the reporter gene in the midbrain and a larger group close to the midbrain-hindbrain boundary. However, we detected no expression of the catecholaminergic markers ddc, slc6a3, or dbh in these neurons, suggesting that they are not actively transmitting catecholamines. The identified neurons in the midbrain are located in a GABAergic territory. A coexpression analysis with anatomical markers revealed that Th-expressing neurons in the midbrain are located in the tegmentum and those close to the midbrain-hindbrain boundary are located in the hindbrain. Our data suggest that zebrafish may still have some evolutionary remnants of midbrain dopaminergic neurons.
Collapse
Affiliation(s)
- Christian Altbürger
- Developmental Biology, Faculty of Biology, Institute of Biology I, Albert Ludwigs University Freiburg, Freiburg, Germany
- CIBSS and BIOSS - Centres for Biological Signalling Studies, Albert Ludwigs University Freiburg, Freiburg, Germany
| | - Jens Holzhauser
- Developmental Biology, Faculty of Biology, Institute of Biology I, Albert Ludwigs University Freiburg, Freiburg, Germany
| | - Wolfgang Driever
- Developmental Biology, Faculty of Biology, Institute of Biology I, Albert Ludwigs University Freiburg, Freiburg, Germany
- CIBSS and BIOSS - Centres for Biological Signalling Studies, Albert Ludwigs University Freiburg, Freiburg, Germany
| |
Collapse
|
66
|
Marie A, Kinet R, Helbling JC, Darricau M, Alfos S, Di Miceli M, Angelo MF, Foury A, Richard E, Trifilieff P, Mallet NP, Bosch-Bouju C. Impact of dietary vitamin A on striatal function in adult rats. FASEB J 2023; 37:e23037. [PMID: 37392372 DOI: 10.1096/fj.202300133r] [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: 01/25/2023] [Revised: 04/18/2023] [Accepted: 06/05/2023] [Indexed: 07/03/2023]
Abstract
The striatum is a brain structure involved in the control of voluntary movement. Striatum contains high amounts of retinoic acid, the active metabolite of vitamin A, as well as retinoid receptors, RARβ and RXRγ. Previous studies revealed that disruption of retinoid signaling initiated during development is deleterious for striatal physiology and related motor functions. However, the alteration of retinoid signaling, and the importance of vitamin A supply during adulthood on striatal physiology and function has never been established. In the present study, we investigated the impact of vitamin A supply on striatal function. Adult Sprague-Dawley rats were fed with three specific diets, either sub-deficient, sufficient, or enriched in vitamin A (0.4, 5, and 20 international units [IU] of retinol per g of diet, respectively) for 6 months. We first validated that vitamin A sub-deficient diet in adult rats constitutes a physiological model of retinoid signaling reduction in the striatum. We then revealed subtle alterations of fine motor skills in sub-deficient rats using a new behavioral apparatus specifically designed to test forepaw reach-and-grasp skills relying on striatal function. Finally, we showed using qPCR analysis and immunofluorescence that the striatal dopaminergic system per se was not affected by vitamin A sub-deficiency at adult age. Rather, cholinergic synthesis in the striatum and μ-opioid receptor expression in striosomes sub-territories were the most affected by vitamin A sub-deficiency starting at adulthood. Taken together these results revealed that retinoid signaling alteration at adulthood is associated with motor learning deficits together with discrete neurobiological alterations in the striatum.
Collapse
Affiliation(s)
- Anaïs Marie
- INRAE, Bordeaux INP, NutriNeuro, UMR 1286, University of Bordeaux, Bordeaux, France
| | - Rémi Kinet
- Institut des Maladies Neurodégénératives, UMR 5293, University of Bordeaux, Bordeaux, France
- CNRS UMR 5293, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| | | | - Morgane Darricau
- INRAE, Bordeaux INP, NutriNeuro, UMR 1286, University of Bordeaux, Bordeaux, France
- Institut des Maladies Neurodégénératives, UMR 5293, University of Bordeaux, Bordeaux, France
- CNRS UMR 5293, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| | - Serge Alfos
- INRAE, Bordeaux INP, NutriNeuro, UMR 1286, University of Bordeaux, Bordeaux, France
| | - Mathieu Di Miceli
- Worcester Biomedical Research Group, School of Science and the Environment, University of Worcester, Worcester, UK
| | | | - Aline Foury
- INRAE, Bordeaux INP, NutriNeuro, UMR 1286, University of Bordeaux, Bordeaux, France
| | - Emmanuel Richard
- INSERM, U1035, CHU Bordeaux, University of Bordeaux, Bordeaux, France
| | - Pierre Trifilieff
- INRAE, Bordeaux INP, NutriNeuro, UMR 1286, University of Bordeaux, Bordeaux, France
| | - Nicolas P Mallet
- Institut des Maladies Neurodégénératives, UMR 5293, University of Bordeaux, Bordeaux, France
- CNRS UMR 5293, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| | | |
Collapse
|
67
|
Neuman SS, Metzger JM, Bondarenko V, Wang Y, Felton J, Levine JE, Saha K, Gong S, Emborg ME. Striatonigral distribution of a fluorescent reporter following intracerebral delivery of genome editors. Front Bioeng Biotechnol 2023; 11:1237613. [PMID: 37564994 PMCID: PMC10410562 DOI: 10.3389/fbioe.2023.1237613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 07/18/2023] [Indexed: 08/12/2023] Open
Abstract
Introduction: Targeted gene editing is proposed as a therapeutic approach for numerous disorders, including neurological diseases. As the brain is organized into neural networks, it is critical to understand how anatomically connected structures are affected by genome editing. For example, neurons in the substantia nigra pars compacta (SNpc) project to the striatum, and the striatum contains neurons that project to the substantia nigra pars reticulata (SNpr). Methods: Here, we report the effect of injecting genome editors into the striatum of Ai14 reporter mice, which have a LoxP-flanked stop cassette that prevents expression of the red fluorescent protein tdTomato. Two weeks following intracerebral delivery of either synthetic nanocapsules (NCs) containing CRISPR ribonucleoprotein targeting the tdTomato stop cassette or adeno-associated virus (AAV) vectors expressing Cre recombinase, the brains were collected, and the presence of tdTomato was assessed in both the striatum and SN. Results: TdTomato expression was observed at the injection site in both the NC- and AAV-treated groups and typically colocalized with the neuronal marker NeuN. In the SN, tdTomato-positive fibers were present in the pars reticulata, and SNpr area expressing tdTomato correlated with the size of the striatal genome edited area. Conclusion: These results demonstrate in vivo anterograde axonal transport of reporter gene protein products to the SNpr following neuronal genome editing in the striatum.
Collapse
Affiliation(s)
- Samuel S. Neuman
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Jeanette M. Metzger
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Viktoriya Bondarenko
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Yuyuan Wang
- Department of Ophthalmology and Visual Sciences, University of Wisconsin–Madison, Madison, WI, United States
- Department of Biomedical Engineering, University of Wisconsin–Madison, Madison, WI, United States
- Wisconsin Institute for Discovery, University of Wisconsin–Madison, Madison, WI, United States
| | - Jesi Felton
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Jon E. Levine
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, United States
- Department of Neuroscience, University of Wisconsin–Madison, Madison, WI, United States
| | - Krishanu Saha
- Department of Biomedical Engineering, University of Wisconsin–Madison, Madison, WI, United States
- Wisconsin Institute for Discovery, University of Wisconsin–Madison, Madison, WI, United States
| | - Shaoqin Gong
- Department of Ophthalmology and Visual Sciences, University of Wisconsin–Madison, Madison, WI, United States
- Department of Biomedical Engineering, University of Wisconsin–Madison, Madison, WI, United States
- Wisconsin Institute for Discovery, University of Wisconsin–Madison, Madison, WI, United States
| | - Marina E. Emborg
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, United States
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
68
|
Faget L, Oriol L, Lee WC, Sargent C, Ramanathan D, Hnasko TS. Ventral pallidum GABA and glutamate neurons drive approach and avoidance through distinct modulation of VTA cell types. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.12.548750. [PMID: 37502884 PMCID: PMC10369949 DOI: 10.1101/2023.07.12.548750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
The ventral pallidum (VP) contains GABA and glutamate (Glut) neurons projecting to ventral tegmental area (VTA) whose stimulation drives approach and avoidance, respectively. Yet little is known about the cell-type-specific mechanisms by which VP projections to VTA drive behavior. Here, we found that both VP GABA and Glut neurons were activated during approach to reward or delivery of an aversive stimulus. Stimulation of VP GABA neurons inhibited VTA GABA, but activated dopamine (DA) and glutamate neurons. Remarkably, this cell-type-specific recruitment was behavior-contingent such that VTA recruitment was inhibited when evoked by the subject's own action. Conversely, VP Glut neurons activated VTA GABA, as well as DA and Glut neurons, despite driving aversion. However, VP Glut neurons evoked DA in reward-associated ventromedial nucleus accumbens (NAc), but reduced DA in aversion-associated dorsomedial NAc. These findings show how heterogeneous VP cell types can engage VTA cell types to shape approach and avoidance behaviors.
Collapse
Affiliation(s)
- Lauren Faget
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA
- Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA 92161, USA
| | - Lucie Oriol
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Wen-Chun Lee
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Cody Sargent
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Dhakshin Ramanathan
- Department of Psychiatry, University of California, San Diego, La Jolla, CA 92093, USA
- Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA 92161, USA
| | - Thomas S. Hnasko
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA
- Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA 92161, USA
| |
Collapse
|
69
|
Abstract
Striosomes form neurochemically specialized compartments of the striatum embedded in a large matrix made up of modules called matrisomes. Striosome-matrix architecture is multiplexed with the canonical direct-indirect organization of the striatum. Striosomal functions remain to be fully clarified, but key information is emerging. First, striosomes powerfully innervate nigral dopamine-containing neurons and can completely shut down their activity, with a following rebound excitation. Second, striosomes receive limbic and cognition-related corticostriatal afferents and are dynamically modulated in relation to value-based actions. Third, striosomes are spatially interspersed among matrisomes and interneurons and are influenced by local and global neuromodulatory and oscillatory activities. Fourth, striosomes tune engagement and the motivation to perform reinforcement learning, to manifest stereotypical behaviors, and to navigate valence conflicts and valence discriminations. We suggest that, at an algorithmic level, striosomes could serve as distributed scaffolds to provide formats of the striatal computations generated through development and refined through learning. We propose that striosomes affect subjective states. By transforming corticothalamic and other inputs to the functional formats of the striatum, they could implement state transitions in nigro-striato-nigral circuits to affect bodily and cognitive actions according to internal motives whose functions are compromised in neuropsychiatric conditions.
Collapse
Affiliation(s)
- Ann M Graybiel
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA;
| | - Ayano Matsushima
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA;
| |
Collapse
|
70
|
Soden ME, Yee JX, Zweifel LS. Circuit coordination of opposing neuropeptide and neurotransmitter signals. Nature 2023; 619:332-337. [PMID: 37380765 PMCID: PMC10947507 DOI: 10.1038/s41586-023-06246-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 05/22/2023] [Indexed: 06/30/2023]
Abstract
Fast-acting neurotransmitters and slow, modulatory neuropeptides are co-released from neurons in the central nervous system, albeit from distinct synaptic vesicles1. The mechanisms of how co-released neurotransmitters and neuropeptides that have opposing actions-for example, stimulatory versus inhibitory-work together to exert control of neural circuit output remain unclear. This has been difficult to resolve owing to the inability to selectively isolate these signalling pathways in a cell- and circuit-specific manner. Here we developed a genetic-based anatomical disconnect procedure that utilizes distinct DNA recombinases to independently facilitate CRISPR-Cas9 mutagenesis2 of neurotransmitter- and neuropeptide-related genes in distinct cell types in two different brain regions simultaneously. We demonstrate that neurons within the lateral hypothalamus that produce the stimulatory neuropeptide neurotensin and the inhibitory neurotransmitter GABA (γ-aminobutyric acid) utilize these signals to coordinately activate dopamine-producing neurons of the ventral tegmental area. We show that GABA release from lateral hypothalamus neurotensin neurons inhibits GABA neurons within the ventral tegmental area, disinhibiting dopamine neurons and causing a rapid rise in calcium, whereas neurotensin directly generates a slow inactivating calcium signal in dopamine neurons that is dependent on the expression of neurotensin receptor 1 (Ntsr1). We further show that these two signals work together to regulate dopamine neuron responses to maximize behavioural responding. Thus, a neurotransmitter and a neuropeptide with opposing signals can act on distinct timescales through different cell types to enhance circuit output and optimize behaviour.
Collapse
Affiliation(s)
- Marta E Soden
- Department of Pharmacology, University of Washington, Seattle, WA, USA.
| | - Joshua X Yee
- Department of Pharmacology, University of Washington, Seattle, WA, USA
| | - Larry S Zweifel
- Department of Pharmacology, University of Washington, Seattle, WA, USA.
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, USA.
| |
Collapse
|
71
|
Courtney CD, Pamukcu A, Chan CS. Cell and circuit complexity of the external globus pallidus. Nat Neurosci 2023; 26:1147-1159. [PMID: 37336974 PMCID: PMC11382492 DOI: 10.1038/s41593-023-01368-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 05/18/2023] [Indexed: 06/21/2023]
Abstract
The external globus pallidus (GPe) of the basal ganglia has been underappreciated owing to poor understanding of its cells and circuits. It was assumed that the GPe consisted of a homogeneous neuron population primarily serving as a 'relay station' for information flowing through the indirect basal ganglia pathway. However, the advent of advanced tools in rodent models has sparked a resurgence in interest in the GPe. Here, we review recent data that have unveiled the cell and circuit complexity of the GPe. These discoveries have revealed that the GPe does not conform to traditional views of the basal ganglia. In particular, recent evidence confirms that the afferent and efferent connections of the GPe span both the direct and the indirect pathways. Furthermore, the GPe displays broad interconnectivity beyond the basal ganglia, consistent with its emerging multifaceted roles in both motor and non-motor functions. In summary, recent data prompt new proposals for computational rules of the basal ganglia.
Collapse
Affiliation(s)
- Connor D Courtney
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Arin Pamukcu
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - C Savio Chan
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
72
|
Pagliaccio D, Wengler K, Durham K, Fontaine M, Rueppel M, Becker H, Bilek E, Pieper S, Risdon C, Horga G, Fitzgerald KD, Marsh R. Probing midbrain dopamine function in pediatric obsessive-compulsive disorder via neuromelanin-sensitive magnetic resonance imaging. Mol Psychiatry 2023; 28:3075-3082. [PMID: 37198261 PMCID: PMC10189717 DOI: 10.1038/s41380-023-02105-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 04/26/2023] [Accepted: 05/03/2023] [Indexed: 05/19/2023]
Abstract
Obsessive-compulsive disorder (OCD) is an impairing psychiatric condition, which often onsets in childhood. Growing research highlights dopaminergic alterations in adult OCD, yet pediatric studies are limited by methodological constraints. This is the first study to utilize neuromelanin-sensitive MRI as a proxy for dopaminergic function among children with OCD. N = 135 youth (6-14-year-olds) completed high-resolution neuromelanin-sensitive MRI across two sites; n = 64 had an OCD diagnosis. N = 47 children with OCD completed a second scan after cognitive-behavioral therapy. Voxel-wise analyses identified that neuromelanin-MRI signal was higher among children with OCD compared to those without (483 voxels, permutation-corrected p = 0.018). Effects were significant within both the substania nigra pars compacta (p = 0.004, Cohen's d = 0.51) and ventral tegmental area (p = 0.006, d = 0.50). Follow-up analyses indicated that more severe lifetime symptoms (t = -2.72, p = 0.009) and longer illness duration (t = -2.22, p = 0.03) related to lower neuromelanin-MRI signal. Despite significant symptom reduction with therapy (p < 0.001, d = 1.44), neither baseline nor change in neuromelanin-MRI signal associated with symptom improvement. Current results provide the first demonstration of the utility of neuromelanin-MRI in pediatric psychiatry, specifically highlighting in vivo evidence for midbrain dopamine alterations in treatment-seeking youth with OCD. Neuromelanin-MRI likely indexes accumulating alterations over time, herein, implicating dopamine hyperactivity in OCD. Given evidence of increased neuromelanin signal in pediatric OCD but negative association with symptom severity, additional work is needed to parse potential longitudinal or compensatory mechanisms. Future studies should explore the utility of neuromelanin-MRI biomarkers to identify early risk prior to onset, parse OCD subtypes or symptom heterogeneity, and explore prediction of pharmacotherapy response.
Collapse
Affiliation(s)
- David Pagliaccio
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA.
- New York State Psychiatric Institute, New York, NY, USA.
| | - Kenneth Wengler
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
| | - Katherine Durham
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
| | - Martine Fontaine
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
| | - Meryl Rueppel
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
| | - Hannah Becker
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Emily Bilek
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Sarah Pieper
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
| | - Caroline Risdon
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
| | - Guillermo Horga
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
| | - Kate D Fitzgerald
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
| | - Rachel Marsh
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
| |
Collapse
|
73
|
Caridade-Silva R, Araújo B, Martins-Macedo J, Teixeira FG. N-Acetylcysteine Treatment May Compensate Motor Impairments through Dopaminergic Transmission Modulation in a Striatal 6-Hydroxydopamine Parkinson's Disease Rat Model. Antioxidants (Basel) 2023; 12:1257. [PMID: 37371987 DOI: 10.3390/antiox12061257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/29/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Preventing degeneration and the loss of dopaminergic neurons (DAn) in the brain while mitigating motor symptoms remains a challenge in Parkinson's Disease (PD) treatment development. In light of this, developing or repositioning potential disease-modifying approaches is imperative to achieve meaningful translational gains in PD research. Under this concept, N-acetylcysteine (NAC) has revealed promising perspectives in preserving the dopaminergic system capability and modulating PD mechanisms. Although NAC has been shown to act as an antioxidant and (neuro)protector of the brain, it has yet to be acknowledged how this repurposed drug can improve motor symptomatology and provide disease-modifying properties in PD. Therefore, in the present work, we assessed the impact of NAC on motor and histological deficits in a striatal 6-hydroxydopamine (6-OHDA) rat model of PD. The results revealed that NAC enhanced DAn viability, as we found that it could restore dopamine transporter (DAT) levels compared to the untreated 6-OHDA group. Such findings were positively correlated with a significant amelioration in the motor outcomes of the 6-OHDA-treated animals, demonstrating that NAC may, somehow, be a modulator of PD degenerative mechanisms. Overall, we postulated a proof-of-concept milestone concerning the therapeutic application of NAC. Nevertheless, it is extremely important to understand the complexity of this drug and how its therapeutical properties interact with the cellular and molecular PD mechanisms.
Collapse
Affiliation(s)
- Rita Caridade-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, 4710-057/4805-017 Braga/Guimarães, Portugal
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- Center for Translational Health and Medical Biotechnology Research, School of Health, Polytechnic University of Porto, 4200-465 Porto, Portugal
| | - Bruna Araújo
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, 4710-057/4805-017 Braga/Guimarães, Portugal
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- Center for Translational Health and Medical Biotechnology Research, School of Health, Polytechnic University of Porto, 4200-465 Porto, Portugal
| | - Joana Martins-Macedo
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- Center for Translational Health and Medical Biotechnology Research, School of Health, Polytechnic University of Porto, 4200-465 Porto, Portugal
| | - Fábio G Teixeira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, 4710-057/4805-017 Braga/Guimarães, Portugal
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- Center for Translational Health and Medical Biotechnology Research, School of Health, Polytechnic University of Porto, 4200-465 Porto, Portugal
| |
Collapse
|
74
|
Borghammer P. The brain-first vs. body-first model of Parkinson's disease with comparison to alternative models. J Neural Transm (Vienna) 2023; 130:737-753. [PMID: 37062013 DOI: 10.1007/s00702-023-02633-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 04/03/2023] [Indexed: 04/17/2023]
Abstract
The ultimate origin of Lewy body disorders, including Parkinson's disease (PD) and Dementia with Lewy bodies (DLB), is still incompletely understood. Although a large number of pathogenic mechanisms have been implicated, accumulating evidence support that aggregation and neuron-to-neuron propagation of alpha-synuclein may be the core feature of these disorders. The synuclein, origin, and connectome (SOC) disease model of Lewy body disorders was recently introduced. This model is based on the hypothesis that in the majority of patients, the first alpha-synuclein pathology arises in single location and spreads from there. The most common origin sites are the enteric nervous system and the olfactory system. The SOC model predicts that gut-first pathology leads to a clinical body-first subtype characterized by prodromal autonomic symptoms and REM sleep behavior disorder. In contrast, olfactory-first pathology leads to a brain-first subtype with fewer non-motor symptoms before diagnosis. The SOC model further predicts that body-first patients are older, more commonly develop symmetric dopaminergic degeneration, and are at increased risk of dementia-compared to brain-first patients. In this review, the SOC model is explained and compared to alternative models of the pathogenesis of Lewy body disorders, including the Braak staging system, and the Unified Staging System for Lewy Body Disorders. Postmortem evidence from brain banks and clinical imaging data of dopaminergic and cardiac sympathetic loss is reviewed. It is concluded that these datasets seem to be more compatible with the SOC model than with those alternative disease models of Lewy body disorders.
Collapse
Affiliation(s)
- Per Borghammer
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
- Department of Nuclear Medicine and PET, Aarhus University Hospital, Palle Juul-Jensens Boulevard 165, J220, 8200, Aarhus, Denmark.
| |
Collapse
|
75
|
Jang G, Contreras Castillo S, Esteva E, Upadhaya S, Feng J, Adams NM, Richard E, Awatramani R, Sawai CM, Reizis B. Stem cell decoupling underlies impaired lymphoid development during aging. Proc Natl Acad Sci U S A 2023; 120:e2302019120. [PMID: 37216517 PMCID: PMC10236001 DOI: 10.1073/pnas.2302019120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 04/20/2023] [Indexed: 05/24/2023] Open
Abstract
Mammalian aging is associated with multiple defects of hematopoiesis, most prominently with the impaired development of T and B lymphocytes. This defect is thought to originate in hematopoietic stem cells (HSCs) of the bone marrow, specifically due to the age-dependent accumulation of HSCs with preferential megakaryocytic and/or myeloid potential ("myeloid bias"). Here, we tested this notion using inducible genetic labeling and tracing of HSCs in unmanipulated animals. We found that the endogenous HSC population in old mice shows reduced differentiation into all lineages including lymphoid, myeloid, and megakaryocytic. Single-cell RNA sequencing and immunophenotyping (CITE-Seq) showed that HSC progeny in old animals comprised balanced lineage spectrum including lymphoid progenitors. Lineage tracing using the aging-induced HSC marker Aldh1a1 confirmed the low contribution of old HSCs across all lineages. Competitive transplantations of total bone marrow cells with genetically marked HSCs revealed that the contribution of old HSCs was reduced, but compensated by other donor cells in myeloid cells but not in lymphocytes. Thus, the HSC population in old animals becomes globally decoupled from hematopoiesis, which cannot be compensated in lymphoid lineages. We propose that this partially compensated decoupling, rather than myeloid bias, is the primary cause of the selective impairment of lymphopoiesis in older mice.
Collapse
Affiliation(s)
- Geunhyo Jang
- Department of Pathology, New York University Grossman School of Medicine, New York, NY10016
| | | | - Eduardo Esteva
- Department of Pathology, New York University Grossman School of Medicine, New York, NY10016
- Applied Bioinformatics Laboratories, New York University Grossman School of Medicine, New York, NY10016
| | - Samik Upadhaya
- Department of Pathology, New York University Grossman School of Medicine, New York, NY10016
| | - Jue Feng
- Department of Pathology, New York University Grossman School of Medicine, New York, NY10016
| | - Nicholas M. Adams
- Department of Pathology, New York University Grossman School of Medicine, New York, NY10016
| | - Elodie Richard
- INSERM Unit 1312 Bordeaux Institute of Oncology, University of Bordeaux33076Bordeaux, France
| | - Rajeshwar Awatramani
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL60611
| | - Catherine M. Sawai
- INSERM Unit 1312 Bordeaux Institute of Oncology, University of Bordeaux33076Bordeaux, France
| | - Boris Reizis
- Department of Pathology, New York University Grossman School of Medicine, New York, NY10016
| |
Collapse
|
76
|
Jin R, Sun S, Hu Y, Zhang H, Sun X. Neuropeptides Modulate Feeding via the Dopamine Reward Pathway. Neurochem Res 2023:10.1007/s11064-023-03954-4. [PMID: 37233918 DOI: 10.1007/s11064-023-03954-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 05/13/2023] [Accepted: 05/16/2023] [Indexed: 05/27/2023]
Abstract
Dopamine (DA) is a catecholamine neurotransmitter widely distributed in the central nervous system. It participates in various physiological functions, such as feeding, anxiety, fear, sleeping and arousal. The regulation of feeding is exceptionally complex, involving energy homeostasis and reward motivation. The reward system comprises the ventral tegmental area (VTA), nucleus accumbens (NAc), hypothalamus, and limbic system. This paper illustrates the detailed mechanisms of eight typical orexigenic and anorexic neuropeptides that regulate food intake through the reward system. According to recent literature, neuropeptides released from the hypothalamus and other brain regions regulate reward feeding predominantly through dopaminergic neurons projecting from the VTA to the NAc. In addition, their effect on the dopaminergic system is mediated by the prefrontal cortex, paraventricular thalamus, laterodorsal tegmental area, amygdala, and complex neural circuits. Research on neuropeptides involved in reward feeding can help identify more targets to treat diseases with metabolic disorders, such as obesity.
Collapse
Affiliation(s)
- Ruijie Jin
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, China
| | - Shanbin Sun
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, China
| | - Yang Hu
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, China
| | - Hongfei Zhang
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, China
| | - Xiangrong Sun
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
77
|
Peters C, He S, Fermani F, Lim H, Ding W, Mayer C, Klein R. Transcriptomics reveals amygdala neuron regulation by fasting and ghrelin thereby promoting feeding. SCIENCE ADVANCES 2023; 9:eadf6521. [PMID: 37224253 DOI: 10.1126/sciadv.adf6521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 04/19/2023] [Indexed: 05/26/2023]
Abstract
The central amygdala (CeA) consists of numerous genetically defined inhibitory neurons that control defensive and appetitive behaviors including feeding. Transcriptomic signatures of cell types and their links to function remain poorly understood. Using single-nucleus RNA sequencing, we describe nine CeA cell clusters, of which four are mostly associated with appetitive and two with aversive behaviors. To analyze the activation mechanism of appetitive CeA neurons, we characterized serotonin receptor 2a (Htr2a)-expressing neurons (CeAHtr2a) that comprise three appetitive clusters and were previously shown to promote feeding. In vivo calcium imaging revealed that CeAHtr2a neurons are activated by fasting, the hormone ghrelin, and the presence of food. Moreover, these neurons are required for the orexigenic effects of ghrelin. Appetitive CeA neurons responsive to fasting and ghrelin project to the parabrachial nucleus (PBN) causing inhibition of target PBN neurons. These results illustrate how the transcriptomic diversification of CeA neurons relates to fasting and hormone-regulated feeding behavior.
Collapse
Affiliation(s)
- Christian Peters
- Department of Molecules-Signaling-Development, Max-Planck Institute for Biological Intelligence, 82152 Martinsried, Germany
| | - Songwei He
- Department of Molecules-Signaling-Development, Max-Planck Institute for Biological Intelligence, 82152 Martinsried, Germany
| | - Federica Fermani
- Department of Molecules-Signaling-Development, Max-Planck Institute for Biological Intelligence, 82152 Martinsried, Germany
| | - Hansol Lim
- Department of Molecules-Signaling-Development, Max-Planck Institute for Biological Intelligence, 82152 Martinsried, Germany
| | - Wenyu Ding
- Department of Molecules-Signaling-Development, Max-Planck Institute for Biological Intelligence, 82152 Martinsried, Germany
| | - Christian Mayer
- Laboratory of Neurogenomics, Max-Planck Institute for Biological Intelligence, 82152 Martinsried, Germany
| | - Rüdiger Klein
- Department of Molecules-Signaling-Development, Max-Planck Institute for Biological Intelligence, 82152 Martinsried, Germany
| |
Collapse
|
78
|
He CB, Jin Y, Li Y, Zhang Q, Yang B, Xu M, Yang J, Yi XN, Dong YL, Wang J, Li YQ. Collateral projections from the ventral tegmental area/substantia nigra pars compacta to the nucleus accumbens and insular cortex in the rat. Anat Sci Int 2023:10.1007/s12565-023-00728-4. [PMID: 37160827 DOI: 10.1007/s12565-023-00728-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 04/28/2023] [Indexed: 05/11/2023]
Abstract
Midbrain dopaminergic (DAergic) regions including ventral tegmental area (VTA) and substantia nigra pars compacta (SNc) are involved in diverse brain functions. Previous studies demonstrated that the VTA/SNc to nucleus accumbens (NAc) pathway is critical in reward and motivation. Moreover, DAergic innervations within the insular cortex (IC) are reported to play important roles in pain regulation. To investigate whether VTA/SNc sends collateral projections to NAc and IC, we injected retrograde tracer Fluoro-Gold (FG) into the NAc and Fluorescent retrograde tracer beads (RetroBeads) into the ipsilateral IC in rats. Then, to detect whether collateral projection neurons participate in neuropathic pain, parts of the rats received the spare nerve injury (SNI) surgery. The immunofluorescence staining results showed that FG, RetroBeads, and FG/RetroBeads double-labeled neurons were distributed in the VTA/SNc bilaterally with an ipsilateral predominance. The proportion of FG/RetroBeads double-labeled neurons to the total number of FG and RetroBeads-labeled neurons was 16.7% and 30.3%, respectively. About 90.3% of FG/RetroBeads double-labeled neurons showed DAergic neuron marker tyrosine hydroxylase (TH)-immunoreactive (IR), whereas, only 7.5% exhibited a subset of GABAergic inhibitory projection neuron marker parvalbumin (PV)-IR. One week after SNI, about 53.1% and 33.6% of FG- and RetroBeads-labeled neurons were FG/Fos- and RetroBeads/Fos-IR neurons, respectively. Finally, about 35.9% of the FG/RetroBeads double-labeled neurons showed Fos-IR. The present study indicates that parts of DAergic and PV-IR GABAergic neurons in the VTA/SNc send collateral projections to both NAc and IC, which are activated under SNI-induced neuropathic pain, and probably contribute to the regulation of nociception.
Collapse
Affiliation(s)
- Cheng-Bo He
- Department of Human Anatomy, Basic Medical College, Zunyi Medical University, Zunyi, 563006, China
- Department of Anatomy & K. K. Leung Brain Research Centre, Air Force Military Medical University, Xi'an, 710032, China
| | - Yuan Jin
- Department of Human Anatomy, Basic Medical College, Zunyi Medical University, Zunyi, 563006, China
| | - Yan Li
- Department of Human Anatomy, Basic Medical College, Zunyi Medical University, Zunyi, 563006, China
| | - Qian Zhang
- Department of Human Anatomy, Basic Medical College, Zunyi Medical University, Zunyi, 563006, China
| | - Bai Yang
- Department of Anatomy & K. K. Leung Brain Research Centre, Air Force Military Medical University, Xi'an, 710032, China
| | - Mang Xu
- Department of Anatomy, Basic Medical College, Dali University, Dali, 671000, China
| | - Juan Yang
- Key Laboratory of Brain Science Research and Transformation in Tropical Environment of Hainan Province, Haikou, 571199, China
| | - Xi-Nan Yi
- Key Laboratory of Brain Science Research and Transformation in Tropical Environment of Hainan Province, Haikou, 571199, China
| | - Yu-Lin Dong
- Department of Anatomy & K. K. Leung Brain Research Centre, Air Force Military Medical University, Xi'an, 710032, China
| | - Jian Wang
- Department of Cardiothoracic Surgery, General Hospital of Western Theater Command, Chengdu, 610083, China.
| | - Yun-Qing Li
- Department of Anatomy & K. K. Leung Brain Research Centre, Air Force Military Medical University, Xi'an, 710032, China.
- Department of Anatomy, Basic Medical College, Dali University, Dali, 671000, China.
- Key Laboratory of Brain Science Research and Transformation in Tropical Environment of Hainan Province, Haikou, 571199, China.
| |
Collapse
|
79
|
Perisse E, Miranda M, Trouche S. Modulation of aversive value coding in the vertebrate and invertebrate brain. Curr Opin Neurobiol 2023; 79:102696. [PMID: 36871400 DOI: 10.1016/j.conb.2023.102696] [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: 12/02/2022] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 03/06/2023]
Abstract
Avoiding potentially dangerous situations is key for the survival of any organism. Throughout life, animals learn to avoid environments, stimuli or actions that can lead to bodily harm. While the neural bases for appetitive learning, evaluation and value-based decision-making have received much attention, recent studies have revealed more complex computations for aversive signals during learning and decision-making than previously thought. Furthermore, previous experience, internal state and systems level appetitive-aversive interactions seem crucial for learning specific aversive value signals and making appropriate choices. The emergence of novel methodologies (computation analysis coupled with large-scale neuronal recordings, neuronal manipulations at unprecedented resolution offered by genetics, viral strategies and connectomics) has helped to provide novel circuit-based models for aversive (and appetitive) valuation. In this review, we focus on recent vertebrate and invertebrate studies yielding strong evidence that aversive value information can be computed by a multitude of interacting brain regions, and that past experience can modulate future aversive learning and therefore influence value-based decisions.
Collapse
Affiliation(s)
- Emmanuel Perisse
- Institute of Functional Genomics, University of Montpellier, CNRS, Inserm, 141 rue de la Cardonille, 34094 Montpellier Cedex 5, France.
| | - Magdalena Miranda
- Institute of Functional Genomics, University of Montpellier, CNRS, Inserm, 141 rue de la Cardonille, 34094 Montpellier Cedex 5, France
| | - Stéphanie Trouche
- Institute of Functional Genomics, University of Montpellier, CNRS, Inserm, 141 rue de la Cardonille, 34094 Montpellier Cedex 5, France.
| |
Collapse
|
80
|
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
|
81
|
Margolis EB, Moulton MG, Lambeth PS, O'Meara MJ. The life and times of endogenous opioid peptides: Updated understanding of synthesis, spatiotemporal dynamics, and the clinical impact in alcohol use disorder. Neuropharmacology 2023; 225:109376. [PMID: 36516892 PMCID: PMC10548835 DOI: 10.1016/j.neuropharm.2022.109376] [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: 08/22/2022] [Revised: 12/03/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022]
Abstract
The opioid G-protein coupled receptors (GPCRs) strongly modulate many of the central nervous system structures that contribute to neurological and psychiatric disorders including pain, major depressive disorder, and substance use disorders. To better treat these and related diseases, it is essential to understand the signaling of their endogenous ligands. In this review, we focus on what is known and unknown about the regulation of the over two dozen endogenous peptides with high affinity for one or more of the opioid receptors. We briefly describe which peptides are produced, with a particular focus on the recently proposed possible synthesis pathways for the endomorphins. Next, we describe examples of endogenous opioid peptide expression organization in several neural circuits and how they appear to be released from specific neural compartments that vary across brain regions. We discuss current knowledge regarding the strength of neural activity required to drive endogenous opioid peptide release, clues about how far peptides diffuse from release sites, and their extracellular lifetime after release. Finally, as a translational example, we discuss the mechanisms of action of naltrexone (NTX), which is used clinically to treat alcohol use disorder. NTX is a synthetic morphine analog that non-specifically antagonizes the action of most endogenous opioid peptides developed in the 1960s and FDA approved in the 1980s. We review recent studies clarifying the precise endogenous activity that NTX prevents. Together, the works described here highlight the challenges and opportunities the complex opioid system presents as a therapeutic target.
Collapse
Affiliation(s)
- Elyssa B Margolis
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA; Neuroscience Graduate Program, University of California, San Francisco, CA, USA.
| | - Madelyn G Moulton
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Philip S Lambeth
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Matthew J O'Meara
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
82
|
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
|
83
|
Kielbinski M, Bernacka J, Zajda K, Wawrzczak-Bargieła A, Maćkowiak M, Przewlocki R, Solecki W. Acute stress modulates noradrenergic signaling in the ventral tegmental area-amygdalar circuit. J Neurochem 2023; 164:598-612. [PMID: 36161462 DOI: 10.1111/jnc.15698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 09/09/2022] [Accepted: 09/22/2022] [Indexed: 11/29/2022]
Abstract
Noradrenergic neurotransmission is a critical mediator of stress responses. In turn, exposure to stress induces noradrenergic system adaptations, some of which are implicated in the etiology of stress-related disorders. Adrenergic receptors (ARs) in the ventral tegmental area (VTA) have been demonstrated to regulate phasic dopamine (DA) release in the forebrain, necessary for behavioral responses to conditional cues. However, the impact of stress on noradrenergic modulation of the VTA has not been previously explored. We demonstrate that ARs in the VTA regulate dopaminergic activity in the VTA-BLA (basolateral amygdala) circuit, a key system for processing stress-related stimuli; and that such control is altered by acute stress. We utilized fast-scan cyclic voltammetry to assess the effects of intra-VTA microinfusion of α1 -AR and α2 -AR antagonists (terazosin and RX-821002, respectively), on electrically evoked phasic DA release in the BLA in stress-naïve and stressed (unavoidable electric shocks - UES) anesthetized male Sprague-Dawley rats. In addition, we used western blotting to explore UES-induced alterations in AR protein level in the VTA. Intra-VTA terazosin or RX-821002 dose-dependently attenuated DA release in the BLA. Interestingly, UES decreased the effects of intra-VTA α2 -AR blockade on DA release (24 h but not 7 days after stress), while the effects of terazosin were unchanged. Despite changes in α2 -AR physiological function in the VTA, UES did not alter α2 -AR protein levels in either intracellular or membrane fractions. These findings demonstrate that NA-ergic modulation of the VTA-BLA circuit undergoes significant alterations in response to acute stress, with α2 -AR signaling indicated as a key target.
Collapse
Affiliation(s)
- Michal Kielbinski
- Department of Neurobiology and Neuropsychology, Jagiellonian University, Institute of Applied Psychology, Krakow, Poland
| | - Joanna Bernacka
- Department of Neurobiology and Neuropsychology, Jagiellonian University, Institute of Applied Psychology, Krakow, Poland.,Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Katarzyna Zajda
- Department of Neurobiology and Neuropsychology, Jagiellonian University, Institute of Applied Psychology, Krakow, Poland
| | - Agnieszka Wawrzczak-Bargieła
- Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Marzena Maćkowiak
- Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Ryszard Przewlocki
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Wojciech Solecki
- Department of Neurobiology and Neuropsychology, Jagiellonian University, Institute of Applied Psychology, Krakow, Poland
| |
Collapse
|
84
|
Moya NA, Yun S, Fleps SW, Martin MM, Nadel JA, Beutler LR, Zweifel LS, Parker JG. The effect of selective nigrostriatal dopamine excess on behaviors linked to the cognitive and negative symptoms of schizophrenia. Neuropsychopharmacology 2023; 48:690-699. [PMID: 36380221 PMCID: PMC9938164 DOI: 10.1038/s41386-022-01492-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 10/16/2022] [Accepted: 10/25/2022] [Indexed: 11/16/2022]
Abstract
Excess dopamine release in the dorsal striatum (DS) is linked to psychosis. Antipsychotics are thought to work by blocking striatal D2 dopamine receptors, but they lack efficacy for the negative and cognitive symptoms of schizophrenia. These observations and the fact that increasing brain-wide dopamine improves cognition have fueled the dogma that excess dopamine is not involved in negative and cognitive symptoms. However, this idea has never been explicitly tested with DS-pathway specificity. To determine if excess DS dopamine is involved in cognitive and negative symptoms, we selectively re-expressed excitatory TRPV1 receptors in DS-projecting dopamine neurons of Trpv1 knockout mice. We treated these mice with capsaicin (TRPV1 agonist) to selectively activate these neurons, validated this approach with fiber photometry, and assessed its effects on social interaction and working memory, behavioral constructs related to negative and cognitive symptoms. We combined this manipulation with antipsychotic treatment (haloperidol) and compared it to brain-wide dopamine release via amphetamine treatment. We found that selectively activating DS-projecting dopamine neurons increased DS (but not cortical) dopamine release and increased locomotor activity. Surprisingly, this manipulation also impaired social interaction and working memory. Haloperidol normalized locomotion, but only partially rescued working memory and had no effect on social interaction. By contrast, amphetamine increased locomotion but did not impair social interaction or working memory. These results suggest that excess dopamine release, when restricted to the DS, causes behavioral deficits linked to negative and cognitive symptoms. Future therapies should address this disregarded role for excess striatal dopamine in the treatment-resistant symptoms of psychosis.
Collapse
Affiliation(s)
- Nicolette A Moya
- Departments of Neuroscience and Pharmacology, Northwestern University, Chicago, IL, USA
| | - Seongsik Yun
- Departments of Neuroscience and Pharmacology, Northwestern University, Chicago, IL, USA
| | - Stefan W Fleps
- Departments of Neuroscience and Pharmacology, Northwestern University, Chicago, IL, USA
| | - Madison M Martin
- Departments of Neuroscience and Pharmacology, Northwestern University, Chicago, IL, USA
| | - Jacob A Nadel
- Departments of Neuroscience and Pharmacology, Northwestern University, Chicago, IL, USA
| | - Lisa R Beutler
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University, Chicago, IL, USA
| | - Larry S Zweifel
- Departments of Psychiatry and Behavioral Sciences and Pharmacology, University of Washington, Seattle, WA, USA
| | - Jones G Parker
- Departments of Neuroscience and Pharmacology, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
85
|
Bienkowski MS. Further refining the boundaries of the hippocampus CA2 with gene expression and connectivity: Potential subregions and heterogeneous cell types. Hippocampus 2023; 33:150-160. [PMID: 36786207 PMCID: PMC9987718 DOI: 10.1002/hipo.23508] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 01/20/2023] [Accepted: 01/23/2023] [Indexed: 02/15/2023]
Abstract
Over the last two decades, the definition of hippocampal area CA2 has evolved from Lorente de Nó's original Golgi-based morphological description with the discovery of specific CA2 gene expression markers. Exploiting the specificity of these molecules has allowed for the genetic dissection of CA2 structure and function in transgenic mice. With this change in criteria, the anatomical boundaries of the CA2 have expanded across the hippocampal axis but the CA2's full rostrocaudal extent is not consistently delineated across atlases. The Hippocampus Gene Expression Atlas (HGEA) provides a comprehensive map of 20 gene expression domains across the entire mouse hippocampus including the CA2. In this commentary, I will review the consensus gene expression patterns that demarcate the expanded CA2 boundaries in the HGEA. Using DropViz single-cell transcriptomics and Mouse Connectome Project connectomics data, I will then suggest potential differences in CA2 cell type heterogeneity and connectivity that may identify and characterize further CA2 subregions.
Collapse
Affiliation(s)
- Michael S Bienkowski
- USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, California, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine of University of Southern California, Los Angeles, California, USA
- USC Center for Integrative Connectomics, Keck School of Medicine of University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
86
|
Bandala C, Cárdenas-Rodríguez N, Mendoza-Torreblanca JG, Contreras-García IJ, Martínez-López V, Cruz-Hernández TR, Carro-Rodríguez J, Vargas-Hernández MA, Ignacio-Mejía I, Alfaro-Rodriguez A, Lara-Padilla E. Therapeutic Potential of Dopamine and Related Drugs as Anti-Inflammatories and Antioxidants in Neuronal and Non-Neuronal Pathologies. Pharmaceutics 2023; 15:pharmaceutics15020693. [PMID: 36840015 PMCID: PMC9966027 DOI: 10.3390/pharmaceutics15020693] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/11/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
Dopamine (DA), its derivatives, and dopaminergic drugs are compounds widely used in the management of diseases related to the nervous system. However, DA receptors have been identified in nonneuronal tissues, which has been related to their therapeutic potential in pathologies such as sepsis or septic shock, blood pressure, renal failure, diabetes, and obesity, among others. In addition, DA and dopaminergic drugs have shown anti-inflammatory and antioxidant properties in different kinds of cells. AIM To compile the mechanism of action of DA and the main dopaminergic drugs and show the findings that support the therapeutic potential of these molecules for the treatment of neurological and non-neurological diseases considering their antioxidant and anti-inflammatory actions. METHOD We performed a review article. An exhaustive search for information was carried out in specialized databases such as PubMed, PubChem, ProQuest, EBSCO, Scopus, Science Direct, Web of Science, Bookshelf, DrugBank, Livertox, and Clinical Trials. RESULTS We showed that DA and dopaminergic drugs have emerged for the management of neuronal and nonneuronal diseases with important therapeutic potential as anti-inflammatories and antioxidants. CONCLUSIONS DA and DA derivatives can be an attractive treatment strategy and a promising approach to slowing the progression of disorders through repositioning.
Collapse
Affiliation(s)
- Cindy Bandala
- Neurociencia Básica, Instituto Nacional de Rehabilitación LGII, Secretaría de Salud, Mexico City 14389, Mexico
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico
- Correspondence: (C.B.); (E.L.-P.); Tel.: +52-(55)-5999-1000 (ext. 19307) (C.B.); +52-(55)-57296000 (ext. 62712) (E.L.-P.)
| | - Noemi Cárdenas-Rodríguez
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico
- Laboratorio de Neurociencias, Subdirección de Medicina Experimental, Instituto Nacional de Pediatría, Mexico City 04530, Mexico
| | | | | | - Valentín Martínez-López
- Unidad de Ingeniería de Tejidos, Terapia Celular y Medicina Regenerativa, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City 14389, Mexico
| | | | - Jazmín Carro-Rodríguez
- Escuela de Biología Experimental, Unidad Iztapalapa, Universidad Autónoma Metropolitana, Mexico City 09340, Mexico
| | | | - Iván Ignacio-Mejía
- Laboratorio de Medicina Traslacional, Escuela Militar de Graduados de Sanidad, Mexico City 11200, Mexico
| | - Alfonso Alfaro-Rodriguez
- Neurociencia Básica, Instituto Nacional de Rehabilitación LGII, Secretaría de Salud, Mexico City 14389, Mexico
| | - Eleazar Lara-Padilla
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico
- Correspondence: (C.B.); (E.L.-P.); Tel.: +52-(55)-5999-1000 (ext. 19307) (C.B.); +52-(55)-57296000 (ext. 62712) (E.L.-P.)
| |
Collapse
|
87
|
Bimpisidis Z, Serra GP, König N, Wallén-Mackenzie Å. Increased sucrose consumption in mice gene-targeted for Vmat2 selectively in NeuroD6-positive neurons of the ventral tegmental area. Front Mol Neurosci 2023; 16:1069834. [PMID: 36825278 PMCID: PMC9941196 DOI: 10.3389/fnmol.2023.1069834] [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: 10/14/2022] [Accepted: 01/11/2023] [Indexed: 02/10/2023] Open
Abstract
Ventral tegmental area (VTA) dopamine (DA) neurons are implicated in reward processing, motivation, reward prediction error, and in substance use disorder. Recent studies have identified distinct neuronal subpopulations within the VTA that can be clustered based on their molecular identity, neurotransmitter profile, physiology, projections and behavioral role. One such subpopulation is characterized by expression of the NeuroD6 gene, and projects primarily to the nucleus accumbens medial shell. We recently showed that optogenetic stimulation of these neurons induces real-time place preference while their targeted deletion of the Vmat2 gene caused altered response to rewarding substances, including ethanol and psychostimulants. Based on these recent findings, we wanted to further investigate the involvement of the NeuroD6-positive VTA subpopulation in reward processing. Using the same NeuroD6Cre+/wt ;Vmat2flox/flox mice as in our prior study, we now addressed the ability of the mice to process sucrose reward. In order to assess appetitive behavior and motivation to obtain sucrose reward, we tested conditional knockout (cKO) and control littermate mice in an operant sucrose self-administration paradigm. We observed that cKO mice demonstrate higher response rates to the operant task and consume more sucrose rewards than control mice. However, their motivation to obtain sucrose is identical to that of control mice. Our results highlight previous observations that appetitive behavior and motivation to obtain rewards can be served by distinct neuronal circuits, and demonstrate that the NeuroD6 VTA subpopulation is involved in mediating the former, but not the latter. Together with previous studies on the NeuroD6 subpopulation, our findings pinpoint the importance of unraveling the molecular and functional role of VTA subpopulations in order to better understand normal behavior and psychiatric disease.
Collapse
Affiliation(s)
| | - Gian Pietro Serra
- Unit of Comparative Physiology, Department of Organismal Biology, Uppsala University, Uppsala, Sweden
| | - Niclas König
- Unit of Comparative Physiology, Department of Organismal Biology, Uppsala University, Uppsala, Sweden
| | - Åsa Wallén-Mackenzie
- Unit of Comparative Physiology, Department of Organismal Biology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
88
|
Tsetsenis T, Broussard JI, Dani JA. Dopaminergic regulation of hippocampal plasticity, learning, and memory. Front Behav Neurosci 2023; 16:1092420. [PMID: 36778837 PMCID: PMC9911454 DOI: 10.3389/fnbeh.2022.1092420] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 12/30/2022] [Indexed: 01/28/2023] Open
Abstract
The hippocampus is responsible for encoding behavioral episodes into short-term and long-term memory. The circuits that mediate these processes are subject to neuromodulation, which involves regulation of synaptic plasticity and local neuronal excitability. In this review, we present evidence to demonstrate the influence of dopaminergic neuromodulation on hippocampus-dependent memory, and we address the controversy surrounding the source of dopamine innervation. First, we summarize historical and recent retrograde and anterograde anatomical tracing studies of direct dopaminergic projections from the ventral tegmental area and discuss dopamine release from the adrenergic locus coeruleus. Then, we present evidence of dopaminergic modulation of synaptic plasticity in the hippocampus. Plasticity mechanisms are examined in brain slices and in recordings from in vivo neuronal populations in freely moving rodents. Finally, we review pharmacological, genetic, and circuitry research that demonstrates the importance of dopamine release for learning and memory tasks while dissociating anatomically distinct populations of direct dopaminergic inputs.
Collapse
Affiliation(s)
- Theodoros Tsetsenis
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States,*Correspondence: Theodoros Tsetsenis John I. Broussard John A. Dani
| | - John I. Broussard
- Department of Neurobiology and Anatomy, UT Health Houston McGovern Medical School, Houston, TX, United States,*Correspondence: Theodoros Tsetsenis John I. Broussard John A. Dani
| | - John A. Dani
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States,*Correspondence: Theodoros Tsetsenis John I. Broussard John A. Dani
| |
Collapse
|
89
|
Tomagra G, Franchino C, Cesano F, Chiarion G, de lure A, Carbone E, Calabresi P, Mesin L, Picconi B, Marcantoni A, Carabelli V. Alpha-synuclein oligomers alter the spontaneous firing discharge of cultured midbrain neurons. Front Cell Neurosci 2023; 17:1078550. [PMID: 36744002 PMCID: PMC9896582 DOI: 10.3389/fncel.2023.1078550] [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: 10/24/2022] [Accepted: 01/06/2023] [Indexed: 01/21/2023] Open
Abstract
The aim of this work was to monitor the effects of extracellular α-synuclein on the firing activity of midbrain neurons dissociated from substantia nigra TH-GFP mice embryos and cultured on microelectrode arrays (MEA). We monitored the spontaneous firing discharge of the network for 21 days after plating and the role of glutamatergic and GABAergic inputs in regulating burst generation and network synchronism. Addition of GABA A , AMPA and NMDA antagonists did not suppress the spontaneous activity but allowed to identify three types of neurons that exhibited different modalities of firing and response to applied L-DOPA: high-rate (HR) neurons, low-rate pacemaking (LR-p), and low-rate non-pacemaking (LR-np) neurons. Most HR neurons were insensitive to L-DOPA, while the majority of LR-p neurons responded with a decrease of the firing discharge; less defined was the response of LR-np neurons. The effect of exogenous α-synuclein (α-syn) on the firing discharge of midbrain neurons was then studied by varying the exposure time (0-48 h) and the α-syn concentration (0.3-70 μM), while the formation of α-syn oligomers was monitored by means of AFM. Independently of the applied concentration, acute exposure to α-syn monomers did not exert any effect on the spontaneous firing rate of HR, LR-p, and LR-np neurons. On the contrary, after 48 h exposure, the firing activity was drastically altered at late developmental stages (14 days in vitro, DIV, neurons): α-syn oligomers progressively reduced the spontaneous firing discharge (IC50 = 1.03 μM), impaired burst generation and network synchronism, proportionally to the increased oligomer/monomer ratio. Different effects were found on early-stage developed neurons (9 DIV), whose firing discharge remained unaltered, regardless of the applied α-syn concentration and the exposure time. Our findings unravel, for the first time, the variable effects of exogenous α-syn at different stages of midbrain network development and provide new evidence for the early detection of neuronal function impairment associated to aggregated forms of α-syn.
Collapse
Affiliation(s)
- Giulia Tomagra
- Drug Science Department, University of Torino, Turin, Italy
- Nanostructured Interfaces and Surfaces Inter-Departmental Research Centre, Turin, Italy
| | | | - Federico Cesano
- Nanostructured Interfaces and Surfaces Inter-Departmental Research Centre, Turin, Italy
- Department of Chemistry and INSTM-UdR Torino, Turin, Italy
| | - Giovanni Chiarion
- Mathematical Biology and Physiology, Department of Electronics and Telecommunications, Turin, Italy
| | - Antonio de lure
- Laboratory Experimental Neurophysiology, IRCCS San Raffaele Rome, Rome, Italy
| | - Emilio Carbone
- Drug Science Department, University of Torino, Turin, Italy
- Nanostructured Interfaces and Surfaces Inter-Departmental Research Centre, Turin, Italy
| | - Paolo Calabresi
- Neurological Clinic, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Neurology, Department of Neuroscience, Faculty of Medicine, Università Cattolica del “Sacro Cuore,”Rome, Italy
| | - Luca Mesin
- Mathematical Biology and Physiology, Department of Electronics and Telecommunications, Turin, Italy
| | - Barbara Picconi
- Laboratory Experimental Neurophysiology, IRCCS San Raffaele Rome, Rome, Italy
- Dipartimento di Scienze Umane e Promozione della Qualitá della Vita, Telematic University San Raffaele Roma, Rome, Italy
| | - Andrea Marcantoni
- Drug Science Department, University of Torino, Turin, Italy
- Nanostructured Interfaces and Surfaces Inter-Departmental Research Centre, Turin, Italy
| | - Valentina Carabelli
- Drug Science Department, University of Torino, Turin, Italy
- Nanostructured Interfaces and Surfaces Inter-Departmental Research Centre, Turin, Italy
| |
Collapse
|
90
|
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
|
91
|
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
|
92
|
Shen Z, Li W, Chang W, Yue N, Yu J. Sex differences in chronic pain-induced mental disorders: Mechanisms of cerebral circuitry. Front Mol Neurosci 2023; 16:1102808. [PMID: 36891517 PMCID: PMC9986270 DOI: 10.3389/fnmol.2023.1102808] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 01/16/2023] [Indexed: 02/22/2023] Open
Abstract
Mental disorders such as anxiety and depression induced by chronic pain are common in clinical practice, and there are significant sex differences in their epidemiology. However, the circuit mechanism of this difference has not been fully studied, as preclinical studies have traditionally excluded female rodents. Recently, this oversight has begun to be resolved and studies including male and female rodents are revealing sex differences in the neurobiological processes behind mental disorder features. This paper reviews the structural functions involved in the injury perception circuit and advanced emotional cortex circuit. In addition, we also summarize the latest breakthroughs and insights into sex differences in neuromodulation through endogenous dopamine, 5-hydroxytryptamine, GABAergic inhibition, norepinephrine, and peptide pathways like oxytocin, as well as their receptors. By comparing sex differences, we hope to identify new therapeutic targets to offer safer and more effective treatments.
Collapse
Affiliation(s)
- Zuqi Shen
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Li
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Weiqi Chang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Na Yue
- Weifang Maternal and Child Health Hospital, Weifang, China
| | - Jin Yu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai, China
| |
Collapse
|
93
|
Molas S, Zhao-Shea R, Freels TG, Tapper AR. Viral Tracing Confirms Paranigral Ventral Tegmental Area Dopaminergic Inputs to the Interpeduncular Nucleus Where Dopamine Release Encodes Motivated Exploration. eNeuro 2023; 10:ENEURO.0282-22.2022. [PMID: 36599671 PMCID: PMC9840383 DOI: 10.1523/eneuro.0282-22.2022] [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: 07/12/2022] [Revised: 12/07/2022] [Accepted: 12/20/2022] [Indexed: 01/05/2023] Open
Abstract
Midbrain dopaminergic (DAergic) neurons of the ventral tegmental area (VTA) are engaged by rewarding stimuli and encode reward prediction error to update goal-directed learning. However, recent data indicate that VTA DAergic neurons are functionally heterogeneous with emerging roles in aversive signaling, salience, and novelty, based in part on anatomic location and projection, highlighting a need to functionally characterize the repertoire of VTA DAergic efferents in motivated behavior. Previous work identifying a mesointerpeduncular circuit consisting of VTA DAergic neurons projecting to the interpeduncular nucleus (IPN), a midbrain area implicated in aversion, anxiety-like behavior, and familiarity, has recently come into question. To verify the existence of this circuit, we combined presynaptic targeted and retrograde viral tracing in the dopamine transporter-Cre mouse line. Consistent with previous reports, synaptic tracing revealed that axon terminals from the VTA innervate the caudal IPN; whereas, retrograde tracing revealed DAergic VTA neurons, predominantly in the paranigral region, project to the nucleus accumbens shell, as well as the IPN. To test whether functional DAergic neurotransmission exists in the IPN, we expressed the genetically encoded DA sensor, dLight 1.2, in the IPN of C57BL/6J mice and measured IPN DA signals in vivo during social and anxiety-like behavior using fiber photometry. We observed an increase in IPN DA signal during social investigation of a novel but not familiar conspecific and during exploration of the anxiogenic open arms of the elevated plus maze. Together, these data confirm VTA DAergic neuron projections to the IPN and implicate this circuit in encoding motivated exploration.
Collapse
Affiliation(s)
- Susanna Molas
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605
| | - Rubing Zhao-Shea
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605
| | - Timothy G Freels
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605
| | - Andrew R Tapper
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605
| |
Collapse
|
94
|
Manion MTC, Glasper ER, Wang KH. A sex difference in mouse dopaminergic projections from the midbrain to basolateral amygdala. Biol Sex Differ 2022; 13:75. [PMID: 36585727 PMCID: PMC9801632 DOI: 10.1186/s13293-022-00486-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 12/20/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Dopaminergic circuits play important roles in the motivational control of behavior and dysfunction in dopaminergic circuits have been implicated in several psychiatric disorders, such as schizophrenia and depression. While these disorders exhibit different incidence rates in men and women, the potential sex differences in the underlying neural circuits are not well-understood. Previous anatomical tracing studies in mammalian species have revealed a prominent circuit projection connecting the dopaminergic midbrain ventral tegmental area (VTA) to the basolateral amygdala (BLA), which is involved in emotional processing and associative learning. However, whether there is any sex difference in this anatomical circuit remains unknown. METHODS To study the potential sex differences in the VTA-to-BLA dopaminergic circuit, we injected two viral vectors encoding fluorescent reporters of axons and synaptic boutons (AAV-FLEX-tdTomato and AAV-FLEX-SynaptophysinGFP, respectively) into the VTA of a mouse transgenic driver line (tyrosine hydroxylase promoter-driven Cre, or TH-Cre), which restricts the reporter expression to dopaminergic neurons. We then used confocal fluorescent microscopy to image the distribution and density of dopaminergic axons and synaptic boutons in serial sections of both male and female mouse brain. RESULTS We found that the overall labeling intensity of VTA-to-BLA dopaminergic projections is intermediate among forebrain dopaminergic pathways, significantly higher than the projections to the prefrontal cortex, but lower than the projections to the nucleus accumbens. Within the amygdala areas, dopaminergic axons are concentrated in BLA. Although the size of BLA and the density of dopaminergic axons within BLA are similar between male and female mice, the density of dopaminergic synaptic boutons in BLA is significantly higher in male brain than female brain. CONCLUSIONS Our results demonstrate an anatomical sex difference in mouse dopaminergic innervations from the VTA to BLA. This finding may provide a structural foundation to study neural circuit mechanisms underlying sex differences in motivational and emotional behaviors and related psychiatric dysfunctions.
Collapse
Affiliation(s)
- Matthew T. C. Manion
- grid.416868.50000 0004 0464 0574Unit on Neural Circuits and Adaptive Behaviors, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892 USA ,grid.164295.d0000 0001 0941 7177Department of Psychology, University of Maryland, College Park, MD 20742 USA ,grid.164295.d0000 0001 0941 7177Program in Neuroscience and Cognitive Science, University of Maryland, College Park, MD 20742 USA
| | - Erica R. Glasper
- grid.164295.d0000 0001 0941 7177Department of Psychology, University of Maryland, College Park, MD 20742 USA ,grid.164295.d0000 0001 0941 7177Program in Neuroscience and Cognitive Science, University of Maryland, College Park, MD 20742 USA ,grid.261331.40000 0001 2285 7943Department of Neuroscience and Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH 43235 USA
| | - Kuan Hong Wang
- grid.416868.50000 0004 0464 0574Unit on Neural Circuits and Adaptive Behaviors, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892 USA ,grid.412750.50000 0004 1936 9166Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY 14642 USA
| |
Collapse
|
95
|
Requie LM, Gómez-Gonzalo M, Speggiorin M, Managò F, Melone M, Congiu M, Chiavegato A, Lia A, Zonta M, Losi G, Henriques VJ, Pugliese A, Pacinelli G, Marsicano G, Papaleo F, Muntoni AL, Conti F, Carmignoto G. Astrocytes mediate long-lasting synaptic regulation of ventral tegmental area dopamine neurons. Nat Neurosci 2022; 25:1639-1650. [PMID: 36396976 DOI: 10.1038/s41593-022-01193-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 10/03/2022] [Indexed: 11/18/2022]
Abstract
The plasticity of glutamatergic transmission in the ventral tegmental area (VTA) represents a fundamental mechanism in the modulation of dopamine neuron burst firing and phasic dopamine release at target regions. These processes encode basic behavioral responses, including locomotor activity, learning and motivated behaviors. Here we describe a hitherto unidentified mechanism of long-term synaptic plasticity in mouse VTA. We found that the burst firing in individual dopamine neurons induces a long-lasting potentiation of excitatory synapses on adjacent dopamine neurons that crucially depends on Ca2+ elevations in astrocytes, mediated by endocannabinoid CB1 and dopamine D2 receptors co-localized at the same astrocytic process, and activation of pre-synaptic metabotropic glutamate receptors. Consistent with these findings, selective in vivo activation of astrocytes increases the burst firing of dopamine neurons in the VTA and induces locomotor hyperactivity. Astrocytes play, therefore, a key role in the modulation of VTA dopamine neuron functional activity.
Collapse
Affiliation(s)
- Linda Maria Requie
- Neuroscience Institute, Section of Padova, National Research Council (CNR) and Department of Biomedical Sciences, Università degli Studi di Padova, Padova, Italy
| | - Marta Gómez-Gonzalo
- Neuroscience Institute, Section of Padova, National Research Council (CNR) and Department of Biomedical Sciences, Università degli Studi di Padova, Padova, Italy.
| | - Michele Speggiorin
- Neuroscience Institute, Section of Padova, National Research Council (CNR) and Department of Biomedical Sciences, Università degli Studi di Padova, Padova, Italy
| | - Francesca Managò
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano di Tecnologia (IIT), Genova, Italy
| | - Marcello Melone
- Department of Experimental and Clinical Medicine, Section of Neuroscience & Cell Biology, Università Politecnica delle Marche, and Center for Neurobiology of Aging, Ancona, Italy
| | - Mauro Congiu
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, Università degli Studi di Cagliari, Cagliari, Italy.,Neuroscience Institute, Section of Cagliari, National Research Council (CNR), Cagliari, Italy
| | - Angela Chiavegato
- Neuroscience Institute, Section of Padova, National Research Council (CNR) and Department of Biomedical Sciences, Università degli Studi di Padova, Padova, Italy
| | - Annamaria Lia
- Neuroscience Institute, Section of Padova, National Research Council (CNR) and Department of Biomedical Sciences, Università degli Studi di Padova, Padova, Italy
| | - Micaela Zonta
- Neuroscience Institute, Section of Padova, National Research Council (CNR) and Department of Biomedical Sciences, Università degli Studi di Padova, Padova, Italy
| | - Gabriele Losi
- Neuroscience Institute, Section of Padova, National Research Council (CNR) and Department of Biomedical Sciences, Università degli Studi di Padova, Padova, Italy.,Nanoscienze Institute, National Research Council (CNR), Modena, Italy
| | - Vanessa Jorge Henriques
- Neuroscience Institute, Section of Padova, National Research Council (CNR) and Department of Biomedical Sciences, Università degli Studi di Padova, Padova, Italy
| | - Arianna Pugliese
- Department of Experimental and Clinical Medicine, Section of Neuroscience & Cell Biology, Università Politecnica delle Marche, and Center for Neurobiology of Aging, Ancona, Italy
| | - Giada Pacinelli
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano di Tecnologia (IIT), Genova, Italy.,Padova Neuroscience Center (PNC), University of Padova, Padova, Italy
| | - Giovanni Marsicano
- University of Bordeaux and Interdisciplinary Institute for Neuroscience (CNRS), Bordeaux, France
| | - Francesco Papaleo
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano di Tecnologia (IIT), Genova, Italy
| | - Anna Lisa Muntoni
- Neuroscience Institute, Section of Cagliari, National Research Council (CNR), Cagliari, Italy
| | - Fiorenzo Conti
- Department of Experimental and Clinical Medicine, Section of Neuroscience & Cell Biology, Università Politecnica delle Marche, and Center for Neurobiology of Aging, Ancona, Italy
| | - Giorgio Carmignoto
- Neuroscience Institute, Section of Padova, National Research Council (CNR) and Department of Biomedical Sciences, Università degli Studi di Padova, Padova, Italy.
| |
Collapse
|
96
|
Stewart A, Mayer FP, Gowrishankar R, Davis GL, Areal LB, Gresch PJ, Katamish RM, Peart R, Stilley SE, Spiess K, Rabil MJ, Diljohn FA, Wiggins AE, Vaughan RA, Hahn MK, Blakely RD. Behaviorally penetrant, anomalous dopamine efflux exposes sex and circuit dependent regulation of dopamine transporters. Mol Psychiatry 2022; 27:4869-4880. [PMID: 36117213 DOI: 10.1038/s41380-022-01773-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 08/18/2022] [Accepted: 08/31/2022] [Indexed: 01/19/2023]
Abstract
Virtually all neuropsychiatric disorders display sex differences in prevalence, age of onset, and/or clinical symptomology. Although altered dopamine (DA) signaling is a feature of many of these disorders, sex-dependent mechanisms uniquely responsive to DA that drive sex-dependent behaviors remain unelucidated. Previously, we established that anomalous DA efflux (ADE) is a prominent feature of the DA transporter (DAT) variant Val559, a coding substitution identified in two male-biased disorders: attention-deficit/hyperactivity disorder and autism spectrum disorder. In vivo, Val559 ADE induces activation of nigrostriatal D2-type DA autoreceptors (D2ARs) that magnifies inappropriate, nonvesicular DA release by elevating phosphorylation and surface trafficking of ADE-prone DAT proteins. Here we demonstrate that DAT Val559 mice exhibit sex-dependent alterations in psychostimulant responses, social behavior, and cognitive performance. In a search for underlying mechanisms, we discovered that the ability of ADE to elicit D2AR regulation of DAT is both sex and circuit-dependent, with dorsal striatum D2AR/DAT coupling evident only in males, whereas D2AR/DAT coupling in the ventral striatum is exclusive to females. Moreover, systemic administration of the D2R antagonist sulpiride, which precludes ADE-driven DAT trafficking, can normalize DAT Val559 behavioral changes unique to each sex and without effects on the opposite sex or wildtype mice. Our studies support the sex- and circuit dependent capacity of D2ARs to regulate DAT as a critical determinant of the sex-biased effects of perturbed DA signaling in neurobehavioral disorders. Moreover, our work provides a cogent example of how a shared biological insult drives alternative physiological and behavioral trajectories as opposed to resilience.
Collapse
Affiliation(s)
- Adele Stewart
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA.,Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Felix P Mayer
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA
| | | | - Gwynne L Davis
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA
| | - Lorena B Areal
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA
| | - Paul J Gresch
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA.,Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Rania M Katamish
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA
| | - Rodeania Peart
- Wilkes Honors College, Florida Atlantic University, Jupiter, FL, USA
| | - Samantha E Stilley
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA
| | - Keeley Spiess
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA
| | - Maximilian J Rabil
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA
| | | | - Angelica E Wiggins
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA
| | - Roxanne A Vaughan
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Maureen K Hahn
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA.,Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Randy D Blakely
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA. .,Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA.
| |
Collapse
|
97
|
Adamson A, Buck SA, Freyberg Z, De Miranda BR. Sex Differences in Dopaminergic Vulnerability to Environmental Toxicants - Implications for Parkinson's Disease. Curr Environ Health Rep 2022; 9:563-573. [PMID: 36201109 PMCID: PMC10201647 DOI: 10.1007/s40572-022-00380-6] [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] [Accepted: 08/24/2022] [Indexed: 01/31/2023]
Abstract
PURPOSE OF REVIEW Sex dimorphism in Parkinson's disease (PD) is an ostensible feature of the neurological disorder, particularly as men are 1.5-2 times more likely to develop PD than women. Clinical features of the disease, such as presentation at onset, most prevalent symptoms, and response to treatment, are also affected by sex. Despite these well-known sex differences in PD risk and phenotype, the mechanisms that impart sex dimorphisms in PD remain poorly understood. RECENT FINDINGS As PD incidence is influenced by environmental factors, an intriguing pattern has recently emerged in research studies suggesting a male-specific vulnerability to dopaminergic neurodegeneration caused by neurotoxicant exposure, with relative protection in females. These new experimental data have uncovered potential mechanisms that provide clues to the source of sex differences in dopaminergic neurodegeneration and other PD pathology such as alpha-synuclein toxicity. In this review, we discuss the emerging evidence of increased male sensitivity to neurodegeneration from environmental exposures. We examine mechanisms underlying dopaminergic neurodegeneration and PD-related pathologies with evidence supporting the roles of estrogen, SRY expression, the vesicular glutamate transporter VGLUT2, and the microbiome as prospective catalysts for male vulnerability. We also highlight the importance of including sex as a biological variable, particularly when evaluating dopaminergic neurotoxicity in the context of PD.
Collapse
Affiliation(s)
- Ashley Adamson
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, 1719 6th Ave South, CIRC 560, Birmingham, AL, 35294, USA
| | - Silas A Buck
- Center for Neuroscience, Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Zachary Freyberg
- Center for Neuroscience, Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Briana R De Miranda
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, 1719 6th Ave South, CIRC 560, Birmingham, AL, 35294, USA.
| |
Collapse
|
98
|
He XB, Guo F, Li K, Yan J, Lee SH. Timing of MeCP2 Expression Determines Midbrain Dopamine Neuron Phenotype Specification. Stem Cells 2022; 40:1043-1055. [PMID: 36041430 DOI: 10.1093/stmcls/sxac061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 08/22/2022] [Indexed: 11/14/2022]
Abstract
Midbrain dopamine (DA) neurons are associated with locomotor and psychiatric disorders. DA phenotype is specified in ancestral neural precursor cells (NPCs) and maintained throughout neuronal differentiation. Here we show that endogenous expression of MeCP2 coincides with DA phenotype specification in mouse mesencephalon, and premature expression of MeCP2 prevents in vitro cultured NPCs from acquiring DA phenotype through interfering NURR1 transactivation of DA phenotype genes. By contrast, ectopic MeCP2 expression does not disturb DA phenotype in the DA neurons. By analyzing the dynamic change of DNA methylation along DA neuronal differentiation at the promoter of DA phenotype gene tyrosine hydroxylase (Th), we show that Th expression is determined by TET1-mediated de-methylation of NURR1 binding sites within Th promoter. Chromatin immunoprecipitation assays demonstrate that premature MeCP2 dominates the DNA binding of the corresponding sites thereby blocking TET1 function in DA NPCs, whereas TET1-mediated de-methylation prevents excessive MeCP2 binding in DA neurons. The significance of temporal DNA methylation status is further confirmed by targeted methylation/demethylation experiments showing that targeted de-methylation in DA NPCs protects DA phenotype specification from ectopic MeCP2 expression, whereas targeted methylation disturbs phenotype maintenance in MeCP2-overexpressed DA neurons. These findings suggest the appropriate timing of MeCP2 expression as a novel determining factor for guiding NPCs into DA lineage.
Collapse
Affiliation(s)
- Xi-Biao He
- Laboratory of Stem Cell Biology and Epigenetics, College of Basic Medical Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, People's Republic of China
| | - Fang Guo
- Laboratory of Stem Cell Biology and Epigenetics, College of Basic Medical Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, People's Republic of China
| | - Kexuan Li
- Laboratory of Stem Cell Biology and Epigenetics, College of Basic Medical Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, People's Republic of China
| | - Jiaqing Yan
- Laboratory of Stem Cell Biology and Epigenetics, College of Basic Medical Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, People's Republic of China
| | - Sang-Hun Lee
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| |
Collapse
|
99
|
Chang S, Fermani F, Lao CL, Huang L, Jakovcevski M, Di Giaimo R, Gagliardi M, Menegaz D, Hennrich AA, Ziller M, Eder M, Klein R, Cai N, Deussing JM. Tripartite extended amygdala-basal ganglia CRH circuit drives locomotor activation and avoidance behavior. SCIENCE ADVANCES 2022; 8:eabo1023. [PMID: 36383658 PMCID: PMC9668302 DOI: 10.1126/sciadv.abo1023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 09/30/2022] [Indexed: 06/16/2023]
Abstract
An adaptive stress response involves various mediators and circuits orchestrating a complex interplay of physiological, emotional, and behavioral adjustments. We identified a population of corticotropin-releasing hormone (CRH) neurons in the lateral part of the interstitial nucleus of the anterior commissure (IPACL), a subdivision of the extended amygdala, which exclusively innervate the substantia nigra (SN). Specific stimulation of this circuit elicits hyperactivation of the hypothalamic-pituitary-adrenal axis, locomotor activation, and avoidance behavior contingent on CRH receptor type 1 (CRHR1) located at axon terminals in the SN, which originate from external globus pallidus (GPe) neurons. The neuronal activity prompting the observed behavior is shaped by IPACLCRH and GPeCRHR1 neurons coalescing in the SN. These results delineate a previously unidentified tripartite CRH circuit functionally connecting extended amygdala and basal ganglia nuclei to drive locomotor activation and avoidance behavior.
Collapse
Affiliation(s)
- Simon Chang
- Molecular Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Federica Fermani
- Molecules-Signaling-Development, Max Planck Institute for Biological Intelligence (in foundation), Martinsried, Germany
| | - Chu-Lan Lao
- Collaborative Research Centre/Sonderforschungsbereich (SFB) 870, Viral Vector Facility, Munich, Germany
| | - Lianyun Huang
- Translational Genetics, Helmholtz Pioneer Campus, Helmholtz Zentrum München, Munich, Germany
| | - Mira Jakovcevski
- Molecular Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Rossella Di Giaimo
- Developmental Neurobiology, Max Planck Institute of Psychiatry, Munich, Germany
- Department of Biology, University of Naples Federico II, Naples Italy
| | - Miriam Gagliardi
- Genomics of Complex Diseases, Max Planck Institute of Psychiatry, Munich, Germany
| | - Danusa Menegaz
- Scientific Core Unit Electrophysiology, Max Planck Institute of Psychiatry, Munich, Germany
| | - Alexandru Adrian Hennrich
- Max von Pettenkofer-Institute Virology, Medical Faculty, and Gene Center, Ludwig Maximilians University Munich, Munich, Germany
| | - Michael Ziller
- Scientific Core Unit Electrophysiology, Max Planck Institute of Psychiatry, Munich, Germany
| | - Matthias Eder
- Scientific Core Unit Electrophysiology, Max Planck Institute of Psychiatry, Munich, Germany
| | - Rüdiger Klein
- Molecules-Signaling-Development, Max Planck Institute for Biological Intelligence (in foundation), Martinsried, Germany
| | - Na Cai
- Translational Genetics, Helmholtz Pioneer Campus, Helmholtz Zentrum München, Munich, Germany
| | - Jan M. Deussing
- Molecular Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| |
Collapse
|
100
|
Gaertner Z, Azcorra M, Dombeck DA, Awatramani R. Molecular heterogeneity in the substantia nigra: A roadmap for understanding PD motor pathophysiology. Neurobiol Dis 2022; 175:105925. [DOI: 10.1016/j.nbd.2022.105925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 11/03/2022] [Accepted: 11/09/2022] [Indexed: 11/13/2022] Open
|