1
|
Gaertner Z, Oram C, Schneeweis A, Schonfeld E, Bolduc C, Chen C, Dombeck D, Parisiadou L, Poulin JF, Awatramani R. Molecular and spatial transcriptomic classification of midbrain dopamine neurons and their alterations in a LRRK2 G2019S model of Parkinson's disease. eLife 2025; 13:RP101035. [PMID: 40353820 PMCID: PMC12068872 DOI: 10.7554/elife.101035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025] Open
Abstract
Several studies have revealed that midbrain dopamine (DA) neurons, even within a single neuroanatomical area, display heterogeneous properties. In parallel, studies using singlecell profiling techniques have begun to cluster DA neurons into subtypes based on their molecular signatures. Recent work has shown that molecularly defined DA subtypes within the substantia nigra (SNc) display distinctive anatomic and functional properties, and differential vulnerability in Parkinson's disease (PD). Based on these provocative results, a granular understanding of these putative subtypes and their alterations in PD models, is imperative. We developed an optimized pipeline for single-nuclear RNA sequencing (snRNA-seq) and generated a high-resolution hierarchically organized map revealing 20 molecularly distinct DA neuron subtypes belonging to three main families. We integrated this data with spatial MERFISH technology to map, with high definition, the location of these subtypes in the mouse midbrain, revealing heterogeneity even within neuroanatomical sub-structures. Finally, we demonstrate that in the preclinical LRRK2G2019S knock-in mouse model of PD, subtype organization and proportions are preserved. Transcriptional alterations occur in many subtypes including those localized to the ventral tier SNc, where differential expression is observed in synaptic pathways, which might account for previously described DA release deficits in this model. Our work provides an advancement of current taxonomic schemes of the mouse midbrain DA neuron subtypes, a high-resolution view of their spatial locations, and their alterations in a prodromal mouse model of PD.
Collapse
Affiliation(s)
- Zachary Gaertner
- Northwestern University Feinberg School of Medicine, Dept of NeurologyChicagoUnited States
- Northwestern University, Dept of NeurobiologyEvanstonUnited States
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research NetworkChevy ChaseUnited States
| | - Cameron Oram
- McGill University (Montreal Neurological Institute), Faculty of Medicine and Health Sciences, Dept of Neurology and NeurosurgeryMontrealCanada
| | - Amanda Schneeweis
- Northwestern University Feinberg School of Medicine, Dept of NeurologyChicagoUnited States
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research NetworkChevy ChaseUnited States
| | - Elan Schonfeld
- Northwestern University Feinberg School of Medicine, Dept of NeurologyChicagoUnited States
| | - Cyril Bolduc
- McGill University (Montreal Neurological Institute), Faculty of Medicine and Health Sciences, Dept of Neurology and NeurosurgeryMontrealCanada
| | - Chuyu Chen
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research NetworkChevy ChaseUnited States
- Northwestern University Feinberg School of Medicine, Dept of PharmacologyChicagoUnited States
| | - Daniel Dombeck
- Northwestern University, Dept of NeurobiologyEvanstonUnited States
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research NetworkChevy ChaseUnited States
| | - Loukia Parisiadou
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research NetworkChevy ChaseUnited States
| | - Jean-Francois Poulin
- McGill University (Montreal Neurological Institute), Faculty of Medicine and Health Sciences, Dept of Neurology and NeurosurgeryMontrealCanada
| | - Rajeshwar Awatramani
- Northwestern University Feinberg School of Medicine, Dept of NeurologyChicagoUnited States
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research NetworkChevy ChaseUnited States
| |
Collapse
|
2
|
Ceballos CC, Pena RFO. Dendritic synaptic integration modes under in vivo-like states. Biophys J 2025:S0006-3495(25)00274-7. [PMID: 40312913 DOI: 10.1016/j.bpj.2025.04.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 04/02/2025] [Accepted: 04/28/2025] [Indexed: 05/03/2025] Open
Abstract
The neural code remains undiscovered and understanding synaptic input integration under in vivo-like conditions is just the initial step toward unraveling it. Synaptic signals generate fast dendritic spikes through two main modes of temporal summation: coincidence detection and integration. In coincidence detection, dendrites fire only when multiple incoming signals arrive in rapid succession, whereas integration involves summation of postsynaptic potentials over longer periods with minimal membrane leakage. This process is influenced by ionic properties, especially as the membrane potential approaches the firing threshold, where inactivating currents play a critical role. However, the modulation of temporal summation by these currents under in vivo-like conditions has not been thoroughly studied. In our research, we used computer simulations of a single dendritic branch to investigate how three inactivating currents-A-type potassium, T-type calcium, and transient sodium-affect temporal summation. We found that calcium and sodium currents promote integrative behavior in dendrites, while potassium currents enhance their ability to act as coincidence detectors. By adjusting the levels of these currents in dendrites, neurons can flexibly switch between integration and coincidence detection modes, providing them with a versatile mechanism for complex tasks such as multiplexing. This flexibility could be key to understanding how neural circuits process information in real time.
Collapse
Affiliation(s)
- Cesar C Ceballos
- Department of Biological Sciences, Florida Atlantic University, Jupiter, Florida
| | - Rodrigo F O Pena
- Department of Biological Sciences, Florida Atlantic University, Jupiter, Florida; Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, Florida.
| |
Collapse
|
3
|
Beaver ML, Evans RC. Muscarinic Receptor Activation Preferentially Inhibits Rebound in Vulnerable Dopaminergic Neurons. J Neurosci 2025; 45:e1443242025. [PMID: 40000233 PMCID: PMC12005241 DOI: 10.1523/jneurosci.1443-24.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 02/10/2025] [Accepted: 02/18/2025] [Indexed: 02/27/2025] Open
Abstract
Dopaminergic subpopulations of the substantia nigra pars compacta (SNc) differentially degenerate in Parkinson's disease and are characterized by unique electrophysiological properties. The vulnerable population expresses a T-type calcium channel-mediated afterdepolarization (ADP) and shows rebound activity upon release from inhibition, whereas the resilient population does not have an ADP and is slower to fire after hyperpolarization. This rebound activity can trigger dopamine release in the striatum, an important component of basal ganglia function. Using whole-cell patch-clamp electrophysiology on ex vivo slices from adult mice of both sexes, we find that muscarinic activation with the nonselective muscarinic agonist oxotremorine inhibits rebound activity more strongly in vulnerable versus resilient SNc neurons. Here, we show that this effect depends on the direct activation of muscarinic receptors on the SNc dopaminergic neurons. Through a series of pharmacological and transgenic knock-out experiments, we tested whether the muscarinic inhibition of rebound was mediated through the canonical rebound-related ion channels: T-type calcium channels, hyperpolarization-activated cation channels (HCN), and A-type potassium channels. We find that muscarinic receptor activation inhibits HCN-mediated current (I h) in vulnerable SNc neurons but that I h activity is not necessary for the muscarinic inhibition of rebound activity. Similarly, we find that oxotremorine inhibits rebound activity independently of T-type calcium channels and A-type potassium channels. Together these findings reveal new principles governing acetylcholine and dopamine interactions, showing that muscarinic receptors directly affect SNc rebound activity in the midbrain at the somatodendritic level and differentially modify information processing in distinct SNc subpopulations.
Collapse
Affiliation(s)
- Megan L Beaver
- Departments of Pharmacology & Physiology, Georgetown University Medical Center, Washington, DC 20007
- Neuroscience, Georgetown University Medical Center, Washington, DC 20007
| | - Rebekah C Evans
- Neuroscience, Georgetown University Medical Center, Washington, DC 20007
| |
Collapse
|
4
|
Gaertner Z, Oram C, Schneeweis A, Schonfeld E, Bolduc C, Chen C, Dombeck D, Parisiadou L, Poulin JF, Awatramani R. Molecular and spatial transcriptomic classification of midbrain dopamine neurons and their alterations in a LRRK2 G2019S model of Parkinson's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.06.597807. [PMID: 38895448 PMCID: PMC11185743 DOI: 10.1101/2024.06.06.597807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Several studies have revealed that midbrain dopamine (DA) neurons, even within a single neuroanatomical area, display heterogeneous properties. In parallel, studies using single cell profiling techniques have begun to cluster DA neurons into subtypes based on their molecular signatures. Recent work has shown that molecularly defined DA subtypes within the substantia nigra (SNc) display distinctive anatomic and functional properties, and differential vulnerability in Parkinson's disease (PD). Based on these provocative results, a granular understanding of these putative subtypes and their alterations in PD models, is imperative. We developed an optimized pipeline for single-nuclear RNA sequencing (snRNA-seq) and generated a high-resolution hierarchically organized map revealing 20 molecularly distinct DA neuron subtypes belonging to three main families. We integrated this data with spatial MERFISH technology to map, with high definition, the location of these subtypes in the mouse midbrain, revealing heterogeneity even within neuroanatomical sub-structures. Finally, we demonstrate that in the preclinical LRRK2G2019S knock-in mouse model of PD, subtype organization and proportions are preserved. Transcriptional alterations occur in many subtypes including those localized to the ventral tier SNc, where differential expression is observed in synaptic pathways, which might account for previously described DA release deficits in this model. Our work provides an advancement of current taxonomic schemes of the mouse midbrain DA neuron subtypes, a high-resolution view of their spatial locations, and their alterations in a prodromal mouse model of PD.
Collapse
Affiliation(s)
- Zachary Gaertner
- Northwestern University Feinberg School of Medicine, Dept of Neurology, Chicago, IL 60611
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Cameron Oram
- McGill University (Montreal Neurological Institute), Faculty of Medicine and Health Sciences, Dept of Neurology and Neurosurgery, Montreal (QC), Canada
| | - Amanda Schneeweis
- Northwestern University Feinberg School of Medicine, Dept of Neurology, Chicago, IL 60611
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Elan Schonfeld
- Northwestern University Feinberg School of Medicine, Dept of Neurology, Chicago, IL 60611
| | - Cyril Bolduc
- McGill University (Montreal Neurological Institute), Faculty of Medicine and Health Sciences, Dept of Neurology and Neurosurgery, Montreal (QC), Canada
| | - Chuyu Chen
- Northwestern University Feinberg School of Medicine, Dept of Pharmacology, Chicago, IL 60611
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Daniel Dombeck
- Northwestern University, Dept of Neurobiology, Evanston, IL 60201
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Loukia Parisiadou
- Northwestern University Feinberg School of Medicine, Dept of Pharmacology, Chicago, IL 60611
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Jean-Francois Poulin
- McGill University (Montreal Neurological Institute), Faculty of Medicine and Health Sciences, Dept of Neurology and Neurosurgery, Montreal (QC), Canada
| | - Rajeshwar Awatramani
- Northwestern University Feinberg School of Medicine, Dept of Neurology, Chicago, IL 60611
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| |
Collapse
|
5
|
Sansalone L, Evans RC, Twedell E, Zhang R, Khaliq ZM. Corticonigral projections recruit substantia nigra pars lateralis dopaminergic neurons for auditory threat memories. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.04.621665. [PMID: 39574768 PMCID: PMC11580856 DOI: 10.1101/2024.11.04.621665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/01/2024]
Abstract
Dopaminergic neurons (DANs) in the lateral substantia nigra project to the tail of striatum (TS), which is involved in threat conditioning. Auditory cortex also contributes to threatening behaviors, but whether it directly interacts with midbrain DANs and how these interactions might influence threat conditioning remain unclear. Here, functional mapping revealed robust excitatory input from auditory and temporal association cortexes to substantia nigra pars lateralis (SNL) DANs, but not to pars compacta (SNc) DANs. SNL DANs exhibited unique firing patterns, with irregular pacemaking and higher maximal firing, reflecting different channel complements than SNc DANs. Behaviorally, inhibiting cortex to SNL projections impaired memory retrieval during auditory threat conditioning. Thus, we demonstrate robust corticonigral projections to SNL DANs, contrasting with previous observations of sparse cortical input to substantia nigra DANs. These findings distinguish SNL DANs from other nigral populations, highlighting their role in threatening behaviors and expanding knowledge of cortex to midbrain interactions.
Collapse
Affiliation(s)
- Lorenzo Sansalone
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, Bethesda, MD, 20892
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Rebekah C. Evans
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, Bethesda, MD, 20892
| | - Emily Twedell
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, Bethesda, MD, 20892
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Renshu Zhang
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, Bethesda, MD, 20892
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Zayd M. Khaliq
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, Bethesda, MD, 20892
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| |
Collapse
|
6
|
Fallah M, Udobi KC, Swiatek AE, Scott CB, Evans RC. Inhibitory basal ganglia nuclei differentially innervate pedunculopontine nucleus subpopulations and evoke opposite motor and valence behaviors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.05.606694. [PMID: 39149277 PMCID: PMC11326182 DOI: 10.1101/2024.08.05.606694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
The canonical basal ganglia model predicts that the substantia nigra pars reticulata (SNr) and the globus pallidus externa (GPe) will have specific effects on locomotion: the SNr inhibiting locomotion and the GPe enhancing it. In this manuscript, we use in vivo optogenetics to show that a projection-defined neural subpopulation within each structure exerts non-canonical effects on locomotion. These non-canonical subpopulations are defined by their projection to the pedunculopontine nucleus (PPN) and mediate opposing effects on reward. To understand how these structures differentially modulate the PPN, we use ex vivo whole-cell recording with optogenetics to comprehensively dissect the SNr and GPe connections to regionally- and molecularly-defined populations of PPN neurons. The SNr inhibits all PPN subtypes, but most strongly inhibits caudal glutamatergic neurons. The GPe selectively inhibits caudal glutamatergic and GABAergic neurons, avoiding both cholinergic and rostral cells. This circuit characterization reveals non-canonical basal ganglia pathways for locomotion and valence.
Collapse
Affiliation(s)
- Michel Fallah
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC, USA 20007
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA 20007
- Senior author
| | - Kenea C Udobi
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA 20007
| | - Aleksandra E Swiatek
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA 20007
| | - Chelsea B Scott
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA 20007
| | - Rebekah C Evans
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA 20007
- Lead contact
| |
Collapse
|
7
|
Beaver ML, Evans RC. Muscarinic receptor activation preferentially inhibits rebound in vulnerable dopaminergic neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.30.605819. [PMID: 39131326 PMCID: PMC11312546 DOI: 10.1101/2024.07.30.605819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Dopaminergic subpopulations of the substantia nigra pars compacta (SNc) differentially degenerate in Parkinson's disease and are characterized by unique electrophysiological properties. The vulnerable population expresses a T-type calcium channel-mediated afterdepolarization (ADP) and shows rebound activity upon release from inhibition, whereas the resilient population does not have an ADP and is slower to fire after hyperpolarization. This rebound activity can trigger dopamine release in the striatum, an important component of basal ganglia function. Using whole-cell patch clamp electrophysiology on ex vivo slices from adult mice of both sexes, we find that muscarinic activation with the non-selective muscarinic agonist Oxotremorine inhibits rebound activity more strongly in vulnerable vs resilient SNc neurons. Here, we show that this effect depends on the direct activation of muscarinic receptors on the SNc dopaminergic neurons. Through a series of pharmacological and transgenic knock-out experiments, we tested whether the muscarinic inhibition of rebound was mediated through the canonical rebound-related ion channels: T-type calcium channels, hyperpolarization-activated cation channels (HCN), and A-type potassium channels. We find that muscarinic receptor activation inhibits HCN-mediated current (Ih) in vulnerable SNc neurons, but that Ih activity is not necessary for the muscarinic inhibition of rebound activity. Similarly, we find that Oxotremorine inhibits rebound activity independently of T-type calcium channels and A-type potassium channels. Together these findings reveal new principles governing acetylcholine and dopamine interactions, showing that muscarinic receptors directly affect SNc rebound activity in the midbrain at the somatodendritic level and differentially modify information processing in distinct SNc subpopulations.
Collapse
Affiliation(s)
- Megan L. Beaver
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington, DC, USA 20007
| | - Rebekah C. Evans
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA 20007
| |
Collapse
|
8
|
Sandoval A, Duran P, Corzo-López A, Fernández-Gallardo M, Muñoz-Herrera D, Leyva-Leyva M, González-Ramírez R, Felix R. The role of voltage-gated calcium channels in the pathogenesis of Parkinson's disease. Int J Neurosci 2024; 134:452-461. [PMID: 35993158 DOI: 10.1080/00207454.2022.2115905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 06/07/2022] [Accepted: 07/29/2022] [Indexed: 10/15/2022]
Abstract
Aim: Voltage-gated calcium (CaV) channels play an essential role in maintaining calcium homeostasis and regulating numerous physiological processes in neurons. Therefore, dysregulation of calcium signaling is relevant in many neurological disorders, including Parkinson's disease (PD). This review aims to introduce the role of CaV channels in PD and discuss some novel aspects of channel regulation and its impact on the molecular pathophysiology of the disease. Methods: an exhaustive search of the literature in the field was carried out using the PubMed database of The National Center for Biotechnology Information. Systematic searches were performed from the initial date of publication to May 2022. Results: Although α-synuclein aggregates are the main feature of PD, L-type calcium (CaV1) channels seem to play an essential role in the pathogenesis of PD. Changes in the functional expression of CaV1.3 channels alter Calcium homeostasis and contribute to the degeneration of dopaminergic neurons. Furthermore, recent studies suggest that CaV channel trafficking towards the cell membrane depends on the activity of the ubiquitin-proteasome system (UPS). In PD, there is an increase in the expression of L-type channels associated with a decrease in the expression of Parkin, an E3 enzyme of the UPS. Therefore, a link between Parkin and CaV channels could play a fundamental role in the pathogenesis of PD and, as such, could be a potentially attractive target for therapeutic intervention. Conclusion: The study of alterations in the functional expression of CaV channels will provide a framework to understand better the neurodegenerative processes that occur in PD and a possible path toward identifying new therapeutic targets to treat this condition.
Collapse
Affiliation(s)
- Alejandro Sandoval
- School of Medicine FES Iztacala, National Autonomous University of Mexico (UNAM), Tlalnepantla, Mexico
| | - Paz Duran
- Department of Cell Biology, Centre for Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - Alejandra Corzo-López
- Department of Cell Biology, Centre for Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | | | - David Muñoz-Herrera
- Department of Cell Biology, Centre for Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - Margarita Leyva-Leyva
- Department of Molecular Biology and Histocompatibility, "Dr. Manuel Gea González" General Hospital, Mexico City, Mexico
| | - Ricardo González-Ramírez
- Department of Molecular Biology and Histocompatibility, "Dr. Manuel Gea González" General Hospital, Mexico City, Mexico
| | - Ricardo Felix
- Department of Cell Biology, Centre for Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| |
Collapse
|
9
|
Seiler JL, Zhuang X, Nelson AB, Lerner TN. Dopamine across timescales and cell types: Relevance for phenotypes in Parkinson's disease progression. Exp Neurol 2024; 374:114693. [PMID: 38242300 DOI: 10.1016/j.expneurol.2024.114693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 01/09/2024] [Accepted: 01/15/2024] [Indexed: 01/21/2024]
Abstract
Dopamine neurons in the substantia nigra pars compacta (SNc) synthesize and release dopamine, a critical neurotransmitter for movement and learning. SNc dopamine neurons degenerate in Parkinson's Disease (PD), causing a host of motor and non-motor symptoms. Here, we review recent conceptual advances in our basic understanding of the dopamine system - including our rapidly advancing knowledge of dopamine neuron heterogeneity - with special attention to their importance for understanding PD. In PD patients, dopamine neuron degeneration progresses from lateral SNc to medial SNc, suggesting clinically relevant heterogeneity in dopamine neurons. With technical advances in dopamine system interrogation, we can understand the relevance of this heterogeneity for PD progression and harness it to develop new treatments.
Collapse
Affiliation(s)
- Jillian L Seiler
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Xiaowen Zhuang
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA; Weill Institute for Neuroscience, University of California San Francisco, San Francisco, CA, USA; Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, CA, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Alexandra B Nelson
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA; Neuroscience Graduate Program, University of California San Francisco, San Francisco, CA, USA; Weill Institute for Neuroscience, University of California San Francisco, San Francisco, CA, USA; Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, CA, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| | - Talia N Lerner
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Northwestern University Interdepartmental Neuroscience Program (NUIN), Evanston, IL, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
10
|
Chen RYT, Evans RC. Comparing tonic and phasic dendritic calcium in cholinergic pedunculopontine neurons and dopaminergic substantia nigra neurons. Eur J Neurosci 2024; 59:1638-1656. [PMID: 38383047 PMCID: PMC10987283 DOI: 10.1111/ejn.16281] [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: 08/28/2023] [Revised: 01/25/2024] [Accepted: 01/30/2024] [Indexed: 02/23/2024]
Abstract
Several brainstem nuclei degenerate in Parkinson's disease (PD). In addition to the well-characterized dopaminergic neurons of the substantia nigra pars compacta (SNc), the cholinergic neurons of the pedunculopontine nucleus (PPN) also degenerate in PD. One leading hypothesis of selective vulnerability is that pacemaking activity and the activation of low-threshold L-type calcium current are major contributors to tonic calcium load and cellular stress in SNc dopaminergic neurons. However, it is not yet clear whether the vulnerable PPN cholinergic neurons share this property. Therefore, we used two-photon dendritic calcium imaging and whole-cell electrophysiology to evaluate the role of L-type calcium channels in tonic and phasic dendritic calcium signals in PPN and SNc neurons. In addition, we investigated N- and P/Q-type calcium channel regulation of firing properties and dendritic calcium in PPN neurons. We found that blocking L-type channels reduces tonic firing rate and dendritic calcium levels in SNc neurons. By contrast, the tonic calcium load in PPN neurons did not depend on L-, N- or P/Q-type channels. However, we found that blocking either L-type (with nifedipine) or N- and P/Q-type (with omega-conotoxin MVIIC) channels reduces phasic calcium influx in PPN dendrites. Together, these findings show that L-type calcium channels play different roles in the activity of SNc and PPN neurons, and suggest that low-threshold L-type channels are not responsible for tonic calcium levels in PPN cholinergic neurons and are therefore not likely to be a source of selective vulnerability in these cells.
Collapse
Affiliation(s)
- Rita Yu-Tzu Chen
- Department of Neuroscience, Georgetown University Medical Center, Washington DC
| | - Rebekah C. Evans
- Department of Neuroscience, Georgetown University Medical Center, Washington DC
| |
Collapse
|
11
|
Hammer N, Vogel P, Lee S, Roeper J. Optogenetic action potentials and intrinsic pacemaker interplay in retrogradely identified midbrain dopamine neurons. Eur J Neurosci 2024; 59:1311-1331. [PMID: 38056070 DOI: 10.1111/ejn.16208] [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: 07/29/2023] [Revised: 11/09/2023] [Accepted: 11/15/2023] [Indexed: 12/08/2023]
Abstract
Dissecting the diversity of midbrain dopamine (DA) neurons by optotagging is a promising addition to better identify their functional properties and contribution to motivated behavior. Retrograde molecular targeting of DA neurons with specific axonal projection allows further refinement of this approach. Here, we focus on adult mouse DA neurons in the substantia nigra pars compacta (SNc) projecting to dorsal striatum (DS) by demonstrating the selectivity of a floxed AAV9-based retrograde channelrhodopsin-eYFP (ChR-eYFP) labeling approach in DAT-cre mice. Furthermore, we show the utility of a sparse labeling version for anatomical single-cell reconstruction and demonstrate that ChR-eYFR expressing DA neurons retain intrinsic functional properties indistinguishable from conventionally retrogradely red-beads-labeled neurons. We systematically explore the properties of optogenetically evoked action potentials (oAPs) and their interaction with intrinsic pacemaking in this defined subpopulation of DA neurons. We found that the shape of the oAP and its first derivative, as a proxy for extracellularly recorded APs, is highly distinct from spontaneous APs (sAPs) of the same neurons and systematically varies across the pacemaker duty cycle. The timing of the oAP also affects the backbone oscillator of the intrinsic pacemaker by introducing transient "compensatory pauses". Characterizing this systematic interplay between oAPs and sAPs in defined DA neurons will also facilitate a refinement of DA neuron optotagging in vivo.
Collapse
Affiliation(s)
- Niklas Hammer
- Institute of Neurophysiology, Neuroscience Center, Goethe University Frankfurt, Germany
| | - Pascal Vogel
- Institute of Neurophysiology, Neuroscience Center, Goethe University Frankfurt, Germany
| | - Sanghun Lee
- Institute of Neurophysiology, Neuroscience Center, Goethe University Frankfurt, Germany
| | - Jochen Roeper
- Institute of Neurophysiology, Neuroscience Center, Goethe University Frankfurt, Germany
| |
Collapse
|
12
|
Bu M, Follett J, Deng I, Tatarnikov I, Wall S, Guenther D, Maczis M, Wimsatt G, Milnerwood A, Moehle MS, Khoshbouei H, Farrer MJ. Inhibition of LRRK2 kinase activity rescues deficits in striatal dopamine physiology in VPS35 p.D620N knock-in mice. NPJ Parkinsons Dis 2023; 9:167. [PMID: 38110354 PMCID: PMC10728137 DOI: 10.1038/s41531-023-00609-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 11/24/2023] [Indexed: 12/20/2023] Open
Abstract
Dysregulation of dopamine neurotransmission profoundly affects motor, motivation and learning behaviors, and can be observed during the prodromal phase of Parkinson's disease (PD). However, the mechanism underlying these pathophysiological changes remains to be elucidated. Mutations in vacuolar protein sorting 35 (VPS35) and leucine-rich repeat kinase 2 (LRRK2) both lead to autosomal dominant PD, and VPS35 and LRRK2 may physically interact to govern the trafficking of synaptic cargos within the endo-lysosomal network in a kinase-dependent manner. To better understand the functional role of VPS35 and LRRK2 on dopamine physiology, we examined Vps35 haploinsufficient (Haplo) and Vps35 p.D620N knock-in (VKI) mice and how their behavior, dopamine kinetics and biochemistry are influenced by LRRK2 kinase inhibitors. We found Vps35 p.D620N significantly elevates LRRK2-mediated phosphorylation of Rab10, Rab12 and Rab29. In contrast, Vps35 haploinsufficiency reduces phosphorylation of Rab12. While striatal dopamine transporter (DAT) expression and function is similarly impaired in both VKI and Haplo mice, that physiology is normalized in VKI by treatment with the LRRK2 kinase inhibitor, MLi-2. As a corollary, VKI animals show a significant increase in amphetamine induced hyperlocomotion, compared to Haplo mice, that is also abolished by MLi-2. Taken together, these data show Vps35 p.D620N confers a gain-of-function with respect to LRRK2 kinase activity, and that VPS35 and LRRK2 functionally interact to regulate DAT function and striatal dopamine transmission.
Collapse
Affiliation(s)
- Mengfei Bu
- Department of Neurology, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Jordan Follett
- Department of Neurology, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Isaac Deng
- Department of Neurology, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Igor Tatarnikov
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Shannon Wall
- Department of Neurology, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Dylan Guenther
- Department of Neurology, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Melissa Maczis
- Department of Neurology, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Genevieve Wimsatt
- Department of Neurology, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Austen Milnerwood
- Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Mark S Moehle
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| | - Habibeh Khoshbouei
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Matthew J Farrer
- Department of Neurology, University of Florida, Gainesville, FL, USA.
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
13
|
Stetsenko A, Koos T. Neuronal implementation of the temporal difference learning algorithm in the midbrain dopaminergic system. Proc Natl Acad Sci U S A 2023; 120:e2309015120. [PMID: 37903252 PMCID: PMC10636325 DOI: 10.1073/pnas.2309015120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 09/29/2023] [Indexed: 11/01/2023] Open
Abstract
The temporal difference learning (TDL) algorithm has been essential to conceptualizing the role of dopamine in reinforcement learning (RL). Despite its theoretical importance, it remains unknown whether a neuronal implementation of this algorithm exists in the brain. Here, we provide an interpretation of the recently described signaling properties of ventral tegmental area (VTA) GABAergic neurons and show that a circuitry of these neurons implements the TDL algorithm. Specifically, we identified the neuronal mechanism of three key components of the TDL model: a sustained state value signal encoded by an afferent input to the VTA, a temporal differentiation circuit formed by two types of VTA GABAergic neurons the combined output of which computes momentary reward prediction (RP) as the derivative of the state value, and the computation of reward prediction errors (RPEs) in dopamine neurons utilizing the output of the differentiation circuit. Using computational methods, we also show that this mechanism is optimally adapted to the biophysics of RPE signaling in dopamine neurons, mechanistically links the emergence of conditioned reinforcement to RP, and can naturally account for the temporal discounting of reinforcement. Elucidating the implementation of the TDL algorithm may further the investigation of RL in biological and artificial systems.
Collapse
Affiliation(s)
- Anya Stetsenko
- Center for Molecular and Behavioral Neuroscience, Rutgers University, Newark, NJ07102
| | - Tibor Koos
- Center for Molecular and Behavioral Neuroscience, Rutgers University, Newark, NJ07102
| |
Collapse
|
14
|
Chen RYT, Evans RC. Comparing tonic and phasic calcium in the dendrites of vulnerable midbrain neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.28.555184. [PMID: 37693427 PMCID: PMC10491175 DOI: 10.1101/2023.08.28.555184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Several midbrain nuclei degenerate in Parkinson's Disease (PD). Many of these nuclei share the common characteristics that are thought to contribute to their selective vulnerability, including pacemaking activity and high levels of calcium influx. In addition to the well-characterized dopaminergic neurons of the substantia nigra pars compacta (SNc), the cholinergic neurons of the pedunculopontine nucleus (PPN) also degenerate in PD. It is well established that the low-threshold L-type calcium current is a main contributor to tonic calcium in SNc dopaminergic neurons and is hypothesized to contribute to their selective vulnerability. However, it is not yet clear whether the vulnerable PPN cholinergic neurons share this property. Therefore, we used two-photon dendritic calcium imaging and whole-cell electrophysiology to evaluate the role of L-type calcium channels in the tonic and phasic activity of PPN neurons and the corresponding dendritic calcium signal and directly compare these characteristics to SNc neurons. We found that blocking L-type channels reduces tonic firing rate and dendritic calcium levels in SNc neurons. By contrast, the calcium load in PPN neurons during pacemaking did not depend on L-type channels. However, we find that blocking L-type channels reduces phasic calcium influx in PPN dendrites. Together, these findings show that L-type calcium channels play different roles in the activity of SNc and PPN neurons, and suggest that low-threshold L-type channels are not responsible for tonic calcium levels in PPN cholinergic neurons and are therefore not likely to be a source of selective vulnerability in these cells.
Collapse
Affiliation(s)
- Rita Yu-Tzu Chen
- Department of Neuroscience, Georgetown University Medical Center, Washington DC
| | - Rebekah C. Evans
- Department of Neuroscience, Georgetown University Medical Center, Washington DC
| |
Collapse
|
15
|
Topczewska A, Giacalone E, Pratt WS, Migliore M, Dolphin AC, Shah MM. T-type Ca 2+ and persistent Na + currents synergistically elevate ventral, not dorsal, entorhinal cortical stellate cell excitability. Cell Rep 2023; 42:112699. [PMID: 37368752 PMCID: PMC10687207 DOI: 10.1016/j.celrep.2023.112699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 03/08/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Dorsal and ventral medial entorhinal cortex (mEC) regions have distinct neural network firing patterns to differentially support functions such as spatial memory. Accordingly, mEC layer II dorsal stellate neurons are less excitable than ventral neurons. This is partly because the densities of inhibitory conductances are higher in dorsal than ventral neurons. Here, we report that T-type Ca2+ currents increase 3-fold along the dorsal-ventral axis in mEC layer II stellate neurons, with twice as much CaV3.2 mRNA in ventral mEC compared with dorsal mEC. Long depolarizing stimuli trigger T-type Ca2+ currents, which interact with persistent Na+ currents to elevate the membrane voltage and spike firing in ventral, not dorsal, neurons. T-type Ca2+ currents themselves prolong excitatory postsynaptic potentials (EPSPs) to enhance their summation and spike coupling in ventral neurons only. These findings indicate that T-type Ca2+ currents critically influence the dorsal-ventral mEC stellate neuron excitability gradient and, thereby, mEC dorsal-ventral circuit activity.
Collapse
Affiliation(s)
| | | | - Wendy S Pratt
- Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Michele Migliore
- Institute of Biophysics, National Research Council, 90146 Palermo, Italy
| | - Annette C Dolphin
- Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Mala M Shah
- Pharmacology, School of Pharmacy, University College London, London WC1N 4AX, UK.
| |
Collapse
|
16
|
Liu Y, Yuan Y, Yan Y, Wang R, Wang Z, Liu X, Zhang Y, Hua J, Wang Y, Zhao L. Mitochondrial pyruvate carrier 1 alleviates hypoxic-ischemic brain injury in rats. Life Sci 2023; 325:121686. [PMID: 37030616 DOI: 10.1016/j.lfs.2023.121686] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 03/28/2023] [Accepted: 04/05/2023] [Indexed: 04/10/2023]
Abstract
AIMS Mitochondrial dysfunction is a critical pathological change in cerebral ischemia. Mitochondrial pyruvate carrier 1 (MPC1) is a mitochondrial inner membrane protein carrier participating in pyruvate transport. The work is aiming to figure out the effect of MPC1 on cerebral ischemia. MAIN METHODS Bilateral internal carotid artery embolization (BICAO) rats model and cells model from oxygen glucose deprivation/reoxygenation (OGD/R) were used to simulate cerebral ischemia in vivo and in vitro. The effect of MPC1 on cerebral ischemia was detected by imaging, behavioral test, immunofluorescence, flow cytometry, transmission electron microscopy, Western blot and RT-Q-PCR. RNA-sequence (RNA-seq) was applied to explore the potential molecular mechanisms underlying the role of MPC1 in cerebral ischemia. KEY FINDING After BICAO or OGD/R treatment, MPC1 expression in ischemic cortical neurons was significantly decreased, and MPC1 deficiency significantly reduced cerebral blood flow, decreased locomotion activities, and exacerbated neuronal injury. Moreover, MPC1 deficiency obviously aggravated oxidative stress, structural disruption and dysfunction of mitochondria, autophagy and calcium overload of ischemic cortical neurons. Interestingly, MPC1 overexpression remarkably reversed neuronal loss and persisting neuronal deficits induced by OGD. Using RNA-seq, 38 MPC1-associated differentially expressed genes were involved in oxidative stress, autophagy and calcium overload. Our results further confirmed that MPC1 could alleviate autophagy via the PI3K/Akt/mTOR pathway in the ischemic cortical neurons. SIGNIFICANCE MPC1 may exert neuroprotective effects by attenuating oxidative stress, mitochondrial dysfunction, calcium overload and autophagy during cerebral ischemia. MPC1-related genes identified by RNA-seq may be a novel therapeutic target for cerebral ischemia.
Collapse
Affiliation(s)
- Yue Liu
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yuan Yuan
- Department of Pathology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yi Yan
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Ruyue Wang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Zhaohui Wang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiao Liu
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Ying Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jianyu Hua
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yue Wang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Li Zhao
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.
| |
Collapse
|
17
|
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
|
18
|
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: 12] [Impact Index Per Article: 6.0] [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
|
19
|
Kuo CS, Darmanis S, Diaz de Arce A, Liu Y, Almanzar N, Wu TTH, Quake SR, Krasnow MA. Neuroendocrinology of the lung revealed by single-cell RNA sequencing. eLife 2022; 11:e78216. [PMID: 36469459 PMCID: PMC9721618 DOI: 10.7554/elife.78216] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 11/15/2022] [Indexed: 12/12/2022] Open
Abstract
Pulmonary neuroendocrine cells (PNECs) are sensory epithelial cells that transmit airway status to the brain via sensory neurons and locally via calcitonin gene-related peptide (CGRP) and γ- aminobutyric acid (GABA). Several other neuropeptides and neurotransmitters have been detected in various species, but the number, targets, functions, and conservation of PNEC signals are largely unknown. We used scRNAseq to profile hundreds of the rare mouse and human PNECs. This revealed over 40 PNEC neuropeptide and peptide hormone genes, most cells expressing unique combinations of 5-18 genes. Peptides are packaged in separate vesicles, their release presumably regulated by the distinct, multimodal combinations of sensors we show are expressed by each PNEC. Expression of the peptide receptors predicts an array of local cell targets, and we show the new PNEC signal angiotensin directly activates one subtype of innervating sensory neuron. Many signals lack lung targets so may have endocrine activity like those of PNEC-derived carcinoid tumors. PNECs are an extraordinarily rich and diverse signaling hub rivaling the enteroendocrine system.
Collapse
Affiliation(s)
- Christin S Kuo
- Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
- Department of Biochemistry and Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| | - Spyros Darmanis
- Department of Bioengineering, Stanford UniversityStanfordUnited States
| | - Alex Diaz de Arce
- Department of Biochemistry and Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| | - Yin Liu
- Department of Biochemistry and Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| | - Nicole Almanzar
- Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
| | - Timothy Ting-Hsuan Wu
- Department of Biochemistry and Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| | - Stephen R Quake
- Department of Bioengineering, Stanford UniversityStanfordUnited States
- Chan-Zuckerburg BiohubSan FranciscoUnited States
| | - Mark A Krasnow
- Department of Biochemistry and Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| |
Collapse
|
20
|
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
|
21
|
Condon AF, Asad N, Dore TM, Williams JT. Co-activation of GPCRs facilitate GIRK-dependent current. J Physiol 2022; 600:4881-4895. [PMID: 36121348 DOI: 10.1113/jp283590] [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: 07/15/2022] [Accepted: 09/09/2022] [Indexed: 12/24/2022] Open
Abstract
The activity of dopamine neurons is dependent on both intrinsic properties and afferent projections. One potent form of inhibition is mediated by the activation of two inhibitory G protein-coupled receptors, D2 and GABAB receptors. Each of these receptors activates G protein-coupled inwardly rectifying potassium (GIRK) channels. Recordings in brain slices have shown that co-activation using saturating concentrations of agonists results in occlusion of the GIRK current. The present study examined the interaction between D2 and GABAB receptors using transient applications of sub-saturating concentrations of agonists where the co-application of one agonist resulted in both facilitation and inhibition (desensitization) of the other. The heterologous facilitation was modelled based on the known cooperative interaction between the G protein βγ subunits and GIRK channels. The results indicate that a low tonic level of G βγ results in facilitation of GIRK current and a high level of G βγ results in occlusion. The kinetics of the current induced by transient receptor activation is prolonged in each case. The results suggest that the cooperative interaction between G βγ subunits and GIRK channels determines both the amplitude and kinetics of GPCR-dependent current. KEY POINTS: Inhibitory D2 and GABAB receptors modulate dopamine neuron activity through shared G protein-coupled inwardly rectifying potassium (GIRK) channels. This study reports robust bidirectional interactions between these two converging receptor pathways. Coincident activation of D2 and GABAB receptors leads to facilitation of GIRK channel currents, augmenting both amplitude and prolonging the duration of phasic responses. Activation of either D2 or GABAB receptors also acutely desensitized the GIRK channel current induced by D2 receptor activation that rapidly recovers following termination of desensitizing stimulus. Results demonstrate that the activity of either G protein-coupled receptor system must be considered in the context of other G protein-coupled receptors.
Collapse
Affiliation(s)
- Alec F Condon
- The Vollum Institute, Oregon Health Sciences University, Portland, USA
| | - Naeem Asad
- New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates
| | - Timothy M Dore
- New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates
| | - John T Williams
- The Vollum Institute, Oregon Health Sciences University, Portland, USA
| |
Collapse
|
22
|
Evans R. Dendritic involvement in inhibition and disinhibition of vulnerable dopaminergic neurons in healthy and pathological conditions. Neurobiol Dis 2022; 172:105815. [PMID: 35820645 PMCID: PMC9851599 DOI: 10.1016/j.nbd.2022.105815] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/14/2022] [Accepted: 07/07/2022] [Indexed: 01/21/2023] Open
Abstract
Dopaminergic neurons in the substantia nigra pars compacta (SNc) differentially degenerate in Parkinson's Disease, with the ventral region degenerating more severely than the dorsal region. Compared with the dorsal neurons, the ventral neurons in the SNc have distinct dendritic morphology, electrophysiological characteristics, and circuit connections with the basal ganglia. These characteristics shape information processing in the ventral SNc and structure the balance of inhibition and disinhibition in the striatonigral circuitry. In this paper, I review foundational studies and recent work comparing the circuitry of the ventral and dorsal SNc neurons and discuss how loss of the ventral neurons early in Parkinson's Disease could affect the overall balance of inhibition and disinhibition of dopamine signals.
Collapse
Affiliation(s)
- R.C. Evans
- Georgetown University Medical Center, Department of Neuroscience, United States of America
| |
Collapse
|
23
|
Ambrosi P, Lerner TN. Striatonigrostriatal circuit architecture for disinhibition of dopamine signaling. Cell Rep 2022; 40:111228. [PMID: 35977498 PMCID: PMC9425427 DOI: 10.1016/j.celrep.2022.111228] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 07/11/2022] [Accepted: 07/26/2022] [Indexed: 11/16/2022] Open
Abstract
The basal ganglia operate largely in closed parallel loops, including an associative circuit for goal-directed behavior originating from the dorsomedial striatum (DMS) and a somatosensory circuit important for habit formation originating from the dorsolateral striatum (DLS). An exception to this parallel circuit organization has been proposed to explain how information might be transferred between striatal subregions, for example, from the DMS to the DLS during habit formation. The "ascending spiral hypothesis" proposes that the DMS disinhibits dopamine signaling in the DLS through a tri-synaptic, open-loop striatonigrostriatal circuit. Here, we use transsynaptic and intersectional genetic tools to investigate both closed- and open-loop striatonigrostriatal circuits. We find strong evidence for closed loops, which would allow striatal subregions to regulate their own dopamine release. We also find evidence for functional synapses in open loops. However, these synapses are unable to modulate tonic dopamine neuron firing, questioning the prominence of their role in mediating crosstalk between striatal subregions.
Collapse
Affiliation(s)
- Priscilla Ambrosi
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Northwestern University Interdepartmental Neuroscience Program (NUIN), Evanston, IL 60208, USA
| | - Talia N Lerner
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
24
|
Neurotensin Release from Dopamine Neurons Drives Long-Term Depression of Substantia Nigra Dopamine Signaling. J Neurosci 2022; 42:6186-6194. [PMID: 35794014 PMCID: PMC9374153 DOI: 10.1523/jneurosci.1395-20.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 05/11/2022] [Accepted: 06/12/2022] [Indexed: 11/21/2022] Open
Abstract
Midbrain dopamine neurons play central physiological roles in voluntary movement, reward learning, and motivated behavior. Inhibitory signaling at somatodendritic dopamine D2 receptor (D2R) synapses modulates excitability of dopamine neurons. The neuropeptide neurotensin is expressed by many inputs to the midbrain and induces LTD of D2R synaptic currents (LTDDA); however, the source of neurotensin that is responsible for LTDDA is not known. Here we show, in brain slices from male and female mice, that LTDDA is driven by neurotensin released by dopamine neurons themselves. Optogenetic stimulation of dopamine neurons was sufficient to induce LTDDA in the substantia nigra, but not the VTA, and was dependent on neurotensin receptor signaling, postsynaptic calcium, and vacuolar-type H+-ATPase activity in the postsynaptic cell. These findings reveal a novel form of signaling between dopamine neurons involving release of the peptide neurotensin, which may act as a feedforward mechanism to increase dopamine neuron excitability.SIGNIFICANCE STATEMENT Dopamine neurons in the midbrain play a critical role in reward learning and the initiation of movement. Aberrant dopamine neuron function is implicated in a range of diseases and disorders, including Parkinson's disease, schizophrenia, obesity, and substance use disorders. D2 receptor-mediated PSCs are produced by a rare form of dendrodendritic synaptic transmission between dopamine neurons. These D2 receptor-mediated PSCs undergo LTD following application of the neuropeptide neurotensin. Here we show that release of neurotensin by dopamine neurons themselves is sufficient to induce LTD of dopamine transmission in the substantia nigra. Neurotensin signaling therefore mediates a second form of interdopamine neuron communication and may provide a mechanism by which dopamine neurons maintain excitability when nigral dopamine is elevated.
Collapse
|
25
|
Kosillo P, Ahmed KM, Aisenberg EE, Karalis V, Roberts BM, Cragg SJ, Bateup HS. Dopamine neuron morphology and output are differentially controlled by mTORC1 and mTORC2. eLife 2022; 11:e75398. [PMID: 35881440 PMCID: PMC9328766 DOI: 10.7554/elife.75398] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 07/08/2022] [Indexed: 02/07/2023] Open
Abstract
The mTOR pathway is an essential regulator of cell growth and metabolism. Midbrain dopamine neurons are particularly sensitive to mTOR signaling status as activation or inhibition of mTOR alters their morphology and physiology. mTOR exists in two distinct multiprotein complexes termed mTORC1 and mTORC2. How each of these complexes affect dopamine neuron properties, and whether they have similar or distinct functions is unknown. Here, we investigated this in mice with dopamine neuron-specific deletion of Rptor or Rictor, which encode obligatory components of mTORC1 or mTORC2, respectively. We find that inhibition of mTORC1 strongly and broadly impacts dopamine neuron structure and function causing somatodendritic and axonal hypotrophy, increased intrinsic excitability, decreased dopamine production, and impaired dopamine release. In contrast, inhibition of mTORC2 has more subtle effects, with selective alterations to the output of ventral tegmental area dopamine neurons. Disruption of both mTOR complexes leads to pronounced deficits in dopamine release demonstrating the importance of balanced mTORC1 and mTORC2 signaling for dopaminergic function.
Collapse
Affiliation(s)
- Polina Kosillo
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Kamran M Ahmed
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Erin E Aisenberg
- Helen Wills Neuroscience Institute, University of California, BerkeleyBerkeleyUnited States
| | - Vasiliki Karalis
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Bradley M Roberts
- Department of Physiology, Physiology, Anatomy and Genetics, University of OxfordOxfordUnited Kingdom
| | - Stephanie J Cragg
- Department of Physiology, Physiology, Anatomy and Genetics, University of OxfordOxfordUnited Kingdom
| | - Helen S Bateup
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
- Helen Wills Neuroscience Institute, University of California, BerkeleyBerkeleyUnited States
- Chan Zuckerberg Biohub, San FranciscoSan FranciscoUnited States
| |
Collapse
|
26
|
Siller A, Hofer NT, Tomagra G, Burkert N, Hess S, Benkert J, Gaifullina A, Spaich D, Duda J, Poetschke C, Vilusic K, Fritz EM, Schneider T, Kloppenburg P, Liss B, Carabelli V, Carbone E, Ortner NJ, Striessnig J. β2-subunit alternative splicing stabilizes Cav2.3 Ca 2+ channel activity during continuous midbrain dopamine neuron-like activity. eLife 2022; 11:e67464. [PMID: 35792082 PMCID: PMC9307272 DOI: 10.7554/elife.67464] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
In dopaminergic (DA) Substantia nigra (SN) neurons Cav2.3 R-type Ca2+-currents contribute to somatodendritic Ca2+-oscillations. This activity may contribute to the selective degeneration of these neurons in Parkinson's disease (PD) since Cav2.3-knockout is neuroprotective in a PD mouse model. Here, we show that in tsA-201-cells the membrane-anchored β2-splice variants β2a and β2e are required to stabilize Cav2.3 gating properties allowing sustained Cav2.3 availability during simulated pacemaking and enhanced Ca2+-currents during bursts. We confirmed the expression of β2a- and β2e-subunit transcripts in the mouse SN and in identified SN DA neurons. Patch-clamp recordings of mouse DA midbrain neurons in culture and SN DA neurons in brain slices revealed SNX-482-sensitive R-type Ca2+-currents with voltage-dependent gating properties that suggest modulation by β2a- and/or β2e-subunits. Thus, β-subunit alternative splicing may prevent a fraction of Cav2.3 channels from inactivation in continuously active, highly vulnerable SN DA neurons, thereby also supporting Ca2+ signals contributing to the (patho)physiological role of Cav2.3 channels in PD.
Collapse
Affiliation(s)
- Anita Siller
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of InnsbruckInnsbruckAustria
| | - Nadja T Hofer
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of InnsbruckInnsbruckAustria
| | - Giulia Tomagra
- Department of Drug Science, NIS Centre, University of TorinoTorinoItaly
| | - Nicole Burkert
- Institute of Applied Physiology, University of Ulm, Ulm, GermanyUlmGermany
| | - Simon Hess
- Institute for Zoology, Biocenter, University of CologneCologneGermany
| | - Julia Benkert
- Institute of Applied Physiology, University of Ulm, Ulm, GermanyUlmGermany
| | - Aisylu Gaifullina
- Institute of Applied Physiology, University of Ulm, Ulm, GermanyUlmGermany
| | - Desiree Spaich
- Institute of Applied Physiology, University of Ulm, Ulm, GermanyUlmGermany
| | - Johanna Duda
- Institute of Applied Physiology, University of Ulm, Ulm, GermanyUlmGermany
| | | | - Kristina Vilusic
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of InnsbruckInnsbruckAustria
| | - Eva Maria Fritz
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of InnsbruckInnsbruckAustria
| | - Toni Schneider
- Institute of Neurophysiology, University of CologneCologneGermany
| | - Peter Kloppenburg
- Institute for Zoology, Biocenter, University of CologneCologneGermany
| | - Birgit Liss
- Institute of Applied Physiology, University of Ulm, Ulm, GermanyUlmGermany
- Linacre College & New College, University of OxfordOxfordUnited Kingdom
| | | | - Emilio Carbone
- Department of Drug Science, NIS Centre, University of TorinoTorinoItaly
| | - Nadine Jasmin Ortner
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of InnsbruckInnsbruckAustria
| | - Jörg Striessnig
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of InnsbruckInnsbruckAustria
| |
Collapse
|
27
|
Liu Z, Yang N, Dong J, Tian W, Chang L, Ma J, Guo J, Tan J, Dong A, He K, Zhou J, Cinar R, Wu J, Salinas AG, Sun L, Kumar M, Sullivan BT, Oldham BB, Pitz V, Makarious MB, Ding J, Kung J, Xie C, Hawes SL, Wang L, Wang T, Chan P, Zhang Z, Le W, Chen S, Lovinger DM, Blauwendraat C, Singleton AB, Cui G, Li Y, Cai H, Tang B. Deficiency in endocannabinoid synthase DAGLB contributes to early onset Parkinsonism and murine nigral dopaminergic neuron dysfunction. Nat Commun 2022; 13:3490. [PMID: 35715418 PMCID: PMC9205912 DOI: 10.1038/s41467-022-31168-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 06/07/2022] [Indexed: 11/09/2022] Open
Abstract
Endocannabinoid (eCB), 2-arachidonoyl-glycerol (2-AG), the most abundant eCB in the brain, regulates diverse neural functions. Here we linked multiple homozygous loss-of-function mutations in 2-AG synthase diacylglycerol lipase β (DAGLB) to an early onset autosomal recessive Parkinsonism. DAGLB is the main 2-AG synthase in human and mouse substantia nigra (SN) dopaminergic neurons (DANs). In mice, the SN 2-AG levels were markedly correlated with motor performance during locomotor skill acquisition. Genetic knockdown of Daglb in nigral DANs substantially reduced SN 2-AG levels and impaired locomotor skill learning, particularly the across-session learning. Conversely, pharmacological inhibition of 2-AG degradation increased nigral 2-AG levels, DAN activity and dopamine release and rescued the locomotor skill learning deficits. Together, we demonstrate that DAGLB-deficiency contributes to the pathogenesis of Parkinsonism, reveal the importance of DAGLB-mediated 2-AG biosynthesis in nigral DANs in regulating neuronal activity and dopamine release, and suggest potential benefits of 2-AG augmentation in alleviating Parkinsonism.
Collapse
Affiliation(s)
- Zhenhua Liu
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
- Department of Neurology, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Nannan Yang
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
- Department of Neurology, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Jie Dong
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
- Clinical Research Center on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, 116011, Dalian, Liaoning, China
| | - Wotu Tian
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
- Department of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 20025, Shanghai, China
| | - Lisa Chang
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jinghong Ma
- Department of Neurology, Xuanwu Hospital of Capital Medical University, 100053, Beijing, China
| | - Jifeng Guo
- Department of Neurology, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Jieqiong Tan
- Centre for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, 410008, Changsha, Hunan, China
| | - Ao Dong
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, 100871, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, 100871, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, 100871, Beijing, China
| | - Kaikai He
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, 100871, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, 100871, Beijing, China
| | - Jingheng Zhou
- In Vivo Neurobiology Group, Neurobiology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA
| | - Resat Cinar
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Junbing Wu
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Armando G Salinas
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD, 20852, USA
| | - Lixin Sun
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Mantosh Kumar
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Breanna T Sullivan
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Braden B Oldham
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Vanessa Pitz
- Molecular Genetics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Mary B Makarious
- Molecular Genetics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jinhui Ding
- Computational Biology Group, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Justin Kung
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Chengsong Xie
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Sarah L Hawes
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Lupeng Wang
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Tao Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, Hubei, China
| | - Piu Chan
- Department of Neurology, Xuanwu Hospital of Capital Medical University, 100053, Beijing, China
| | - Zhuohua Zhang
- Centre for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, 410008, Changsha, Hunan, China
- Department of Neurosciences, University of South China Medical School, 421200, Hengyang, Hunan, China
| | - Weidong Le
- Clinical Research Center on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, 116011, Dalian, Liaoning, China
- Institute of Neurology, Sichuan Academy of Medical Sciences-Sichuan Provincial Hospital, Medical School of University of Electronics & Technology of China, 610045, Chengdu, Sichuan, China
| | - Shengdi Chen
- Department of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 20025, Shanghai, China
| | - David M Lovinger
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD, 20852, USA
| | - Cornelis Blauwendraat
- Integrative Neurogenomics Unit, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Andrew B Singleton
- Molecular Genetics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Guohong Cui
- In Vivo Neurobiology Group, Neurobiology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, 100871, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, 100871, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, 100871, Beijing, China
- Chinese Institute for Brain Research, 102206, Beijing, China
| | - Huaibin Cai
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China.
- Centre for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, 410008, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China.
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, 410008, Changsha, Hunan, China.
| |
Collapse
|
28
|
Xu P, He H, Gao Q, Zhou Y, Wu Z, Zhang X, Sun L, Hu G, Guan Q, You Z, Zhang X, Zheng W, Xiong M, Chen Y. Human midbrain dopaminergic neuronal differentiation markers predict cell therapy outcome in a Parkinson's disease model. J Clin Invest 2022; 132:156768. [PMID: 35700056 PMCID: PMC9282930 DOI: 10.1172/jci156768] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 06/07/2022] [Indexed: 11/17/2022] Open
Abstract
Human pluripotent stem cell (hPSC)-based replacement therapy holds great promise in treating Parkinson's disease (PD). However, the heterogeneity of hPSC-derived donor cells and the low yield of midbrain dopaminergic (mDA) neurons after transplantation hinder its broad clinical application. Here, we depicted the single-cell molecular landscape during mDA neuron differentiation. We found that this process recapitulated the development of multiple but adjacent fetal brain regions including ventral midbrain, isthmus, and ventral hindbrain, resulting in heterogenous donor cell population. We reconstructed the differentiation trajectory of mDA lineage and identified CLSTN2 and PTPRO as specific surface markers of mDA progenitors, which were predictive of mDA neuron differentiation and could facilitate highly enriched mDA neurons (up to 80%) following progenitor sorting and transplantation. Marker sorted progenitors exhibited higher therapeutic potency in correcting motor deficits of PD mice. Different marker sorted grafts had a strikingly consistent cellular composition, in which mDA neurons were enriched, while off-target neuron types were mostly depleted, suggesting stable graft outcomes. Our study provides a better understanding of cellular heterogeneity during mDA neuron differentiation, and establishes a strategy to generate highly purified donor cells to achieve stable and predictable therapeutic outcomes, raising the prospect of hPSC-based PD cell replacement therapies.
Collapse
Affiliation(s)
- Peibo Xu
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Hui He
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Qinqin Gao
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yingying Zhou
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Ziyan Wu
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Xiao Zhang
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Linyu Sun
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Gang Hu
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Qian Guan
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Zhiwen You
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Xinyue Zhang
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Wenping Zheng
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Man Xiong
- Institute State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Yuejun Chen
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
29
|
Shin J, Kovacheva L, Thomas D, Stojanovic S, Knowlton CJ, Mankel J, Boehm J, Farassat N, Paladini C, Striessnig J, Canavier CC, Geisslinger G, Roeper J. Ca v1.3 calcium channels are full-range linear amplifiers of firing frequencies in lateral DA SN neurons. SCIENCE ADVANCES 2022; 8:eabm4560. [PMID: 35675413 PMCID: PMC9177074 DOI: 10.1126/sciadv.abm4560] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 04/22/2022] [Indexed: 05/12/2023]
Abstract
The low-threshold L-type calcium channel Cav1.3 accelerates the pacemaker rate in the heart, but its functional role for the extended dynamic range of neuronal firing is still unresolved. Here, we show that Cav1.3 calcium channels act as unexpectedly simple, full-range linear amplifiers of firing rates for lateral dopamine substantia nigra (DA SN) neurons in mice. This means that they boost in vitro or in vivo firing frequencies between 2 and 50 hertz by about 30%. Furthermore, we demonstrate that clinically relevant, low nanomolar concentrations of the L-type channel inhibitor isradipine selectively reduce the in vivo firing activity of these nigrostriatal DA SN neurons at therapeutic plasma concentrations. Thus, our study identifies the pacemaker function of neuronal Cav1.3 channels and provides direct evidence that repurposing dihydropyridines such as isradipine is feasible to selectively modulate the in vivo activity of highly vulnerable DA SN subpopulations in Parkinson's disease.
Collapse
Affiliation(s)
- Josef Shin
- Goethe University, Institute of Neurophysiology, Neuroscience Center, Frankfurt am Main, Germany
| | - Lora Kovacheva
- Goethe University, Institute of Neurophysiology, Neuroscience Center, Frankfurt am Main, Germany
| | - Dominique Thomas
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Frankfurt am Main, Germany
| | - Strahinja Stojanovic
- Goethe University, Institute of Neurophysiology, Neuroscience Center, Frankfurt am Main, Germany
| | - Christopher J. Knowlton
- Department of Cell Biology and Anatomy, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Johanna Mankel
- Goethe University, Institute of Neurophysiology, Neuroscience Center, Frankfurt am Main, Germany
| | - Johannes Boehm
- Goethe University, Institute of Neurophysiology, Neuroscience Center, Frankfurt am Main, Germany
| | - Navid Farassat
- Goethe University, Institute of Neurophysiology, Neuroscience Center, Frankfurt am Main, Germany
| | - Carlos Paladini
- UTSA Neuroscience Institute, University of Texas at San Antonio, San Antonio, TX, USA
| | - Jörg Striessnig
- University of Innsbruck, Department of Pharmacology and Toxicology, Center for Molecular Biosciences, Innsbruck, Austria
| | - Carmen C. Canavier
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Frankfurt am Main, Germany
| | - Gerd Geisslinger
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Frankfurt am Main, Germany
| | - Jochen Roeper
- Goethe University, Institute of Neurophysiology, Neuroscience Center, Frankfurt am Main, Germany
| |
Collapse
|
30
|
Radulovic J, Ivkovic S, Adzic M. From chronic stress and anxiety to neurodegeneration: Focus on neuromodulation of the axon initial segment. HANDBOOK OF CLINICAL NEUROLOGY 2022; 184:481-495. [PMID: 35034756 DOI: 10.1016/b978-0-12-819410-2.00025-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
To adapt to the sustained demands of chronic stress, discrete brain circuits undergo structural and functional changes often resulting in anxiety disorders. In some individuals, anxiety disorders precede the development of motor symptoms of Parkinson's disease (PD) caused by degeneration of neurons in the substantia nigra (SN). Here, we present a circuit framework for probing a causal link between chronic stress, anxiety, and PD, which postulates a central role of abnormal neuromodulation of the SN's axon initial segment by brainstem inputs. It is grounded in findings demonstrating that the earliest PD pathologies occur in the stress-responsive, emotion regulation network of the brainstem, which provides the SN with dense aminergic and cholinergic innervation. SN's axon initial segment (AIS) has unique features that support the sustained and bidirectional propagation of activity in response to synaptic inputs. It is therefore, especially sensitive to circuit-mediated stress-induced imbalance of neuromodulation, and thus a plausible initiating site of neurodegeneration. This could explain why, although secondary to pathophysiologies in other brainstem nuclei, SN degeneration is the most extensive. Consequently, the cardinal symptom of PD, severe motor deficits, arise from degeneration of the nigrostriatal pathway rather than other brainstem nuclei. Understanding when and how circuit dysfunctions underlying anxiety can progress to neurodegeneration, raises the prospect of timed interventions for reversing, or at least impeding, the early pathophysiologies that lead to PD and possibly other neurodegenerative disorders.
Collapse
Affiliation(s)
- Jelena Radulovic
- Department of Neuroscience, Albert Einstein Medical College, Bronx, NY, United States; Department of Psychiatry and Behavioral Sciences, Albert Einstein Medical College, Bronx, NY, United States.
| | - Sanja Ivkovic
- Department of Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Miroslav Adzic
- Department of Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
31
|
Montero T, Gatica RI, Farassat N, Meza R, González-Cabrera C, Roeper J, Henny P. Dendritic Architecture Predicts in vivo Firing Pattern in Mouse Ventral Tegmental Area and Substantia Nigra Dopaminergic Neurons. Front Neural Circuits 2021; 15:769342. [PMID: 34867214 PMCID: PMC8640462 DOI: 10.3389/fncir.2021.769342] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/12/2021] [Indexed: 11/13/2022] Open
Abstract
The firing activity of ventral tegmental area (VTA) and substantia nigra pars compacta (SNc) dopaminergic (DA) neurons is an important factor in shaping DA release and its role in motivated behavior. Dendrites in DA neurons are the main postsynaptic compartment and, along with cell body and axon initial segment, contribute to action potential generation and firing pattern. In this study, the organization of the dendritic domain in individual VTA and SNc DA neurons of adult male mice, and their relationship to in vivo spontaneous firing, are described. In comparison with dorsal VTA DA neurons, ventrally located VTA neurons (as measured by cell body location) possess a shorter total dendritic length and simpler dendritic architecture, and exhibit the most irregular in vivo firing patterns among DA neurons. In contrast, for DA neurons in the SNc, the higher irregularity of firing was related to a smaller dendritic domain, as measured by convex hull volumes. However, firing properties were also related to the specific regional distribution of the dendritic tree. Thus, VTA DA neurons with a larger extension of their dendritic tree within the parabrachial pigmented (PBP) nucleus fired more regularly compared with those with relatively more dendrites extending outside the PBP. For DA neurons in the SNc, enhanced firing irregularity was associated with a smaller proportion of dendrites penetrating the substantia nigra pars reticulata. These results suggest that differences in dendritic morphology contribute to the in vivo firing properties of individual DA neurons, and that the existence of region-specific synaptic connectivity rules that shape firing diversity.
Collapse
Affiliation(s)
- Trinidad Montero
- Laboratorio de Neuroanatomía, Departamento de Anatomía, and Centro Interdisciplinario de Neurociencia, NeuroUC, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rafael Ignacio Gatica
- Laboratorio de Neuroanatomía, Departamento de Anatomía, and Centro Interdisciplinario de Neurociencia, NeuroUC, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Navid Farassat
- Institute of Neurophysiology, Goethe University, Frankfurt, Germany
| | - Rodrigo Meza
- Laboratorio de Neuroanatomía, Departamento de Anatomía, and Centro Interdisciplinario de Neurociencia, NeuroUC, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cristian González-Cabrera
- Laboratorio de Neuroanatomía, Departamento de Anatomía, and Centro Interdisciplinario de Neurociencia, NeuroUC, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jochen Roeper
- Institute of Neurophysiology, Goethe University, Frankfurt, Germany
| | - Pablo Henny
- Laboratorio de Neuroanatomía, Departamento de Anatomía, and Centro Interdisciplinario de Neurociencia, NeuroUC, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
32
|
Pereira Luppi M, Azcorra M, Caronia-Brown G, Poulin JF, Gaertner Z, Gatica S, Moreno-Ramos OA, Nouri N, Dubois M, Ma YC, Ramakrishnan C, Fenno L, Kim YS, Deisseroth K, Cicchetti F, Dombeck DA, Awatramani R. Sox6 expression distinguishes dorsally and ventrally biased dopamine neurons in the substantia nigra with distinctive properties and embryonic origins. Cell Rep 2021; 37:109975. [PMID: 34758317 PMCID: PMC8607753 DOI: 10.1016/j.celrep.2021.109975] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 09/15/2021] [Accepted: 10/19/2021] [Indexed: 12/12/2022] Open
Abstract
Dopamine (DA) neurons in the ventral tier of the substantia nigra pars compacta (SNc) degenerate prominently in Parkinson's disease, while those in the dorsal tier are relatively spared. Defining the molecular, functional, and developmental characteristics of each SNc tier is crucial to understand their distinct susceptibility. We demonstrate that Sox6 expression distinguishes ventrally and dorsally biased DA neuron populations in the SNc. The Sox6+ population in the ventral SNc includes an Aldh1a1+ subset and is enriched in gene pathways that underpin vulnerability. Sox6+ neurons project to the dorsal striatum and show activity correlated with acceleration. Sox6- neurons project to the medial, ventral, and caudal striatum and respond to rewards. Moreover, we show that this adult division is encoded early in development. Overall, our work demonstrates a dual origin of the SNc that results in DA neuron cohorts with distinct molecular profiles, projections, and functions.
Collapse
Affiliation(s)
- Milagros Pereira Luppi
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Maite Azcorra
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Giuliana Caronia-Brown
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jean-Francois Poulin
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Zachary Gaertner
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Serafin Gatica
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | - Navid Nouri
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Marilyn Dubois
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Yongchao C Ma
- Department of Pediatrics, Feinberg School of Medicine, 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
| | - 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
| | - Francesca Cicchetti
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Daniel A Dombeck
- Department of Neurobiology, Northwestern University, Evanston, IL, USA.
| | - Rajeshwar Awatramani
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
33
|
Daniel NH, Aravind A, Thakur P. Are ion channels potential therapeutic targets for Parkinson's disease? Neurotoxicology 2021; 87:243-257. [PMID: 34699791 DOI: 10.1016/j.neuro.2021.10.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/15/2021] [Accepted: 10/21/2021] [Indexed: 01/31/2023]
Abstract
Parkinson's disease (PD) is primarily associated with the progressive neurodegeneration of the dopaminergic neurons in the substantia nigra region of the brain. The resulting motor symptoms are managed with the help of dopamine replacement therapies. However, these therapeutics do not prevent the neurodegeneration underlying the disease and therefore lose their effectiveness in managing disease symptoms over time. Thus, there is an urgent need to develop newer therapeutics for the benefit of patients. The release of dopamine and the firing activity of substantia nigra neurons is regulated by several ion channels that act in concert. Dysregulations of these channels cause the aberrant movement of various ions in the intracellular milieu. This eventually leads to disruption of intracellular signalling cascades, alterations in cellular homeostasis, and bioenergetic deficits. Therefore, ion channels play a central role in driving the high vulnerability of dopaminergic neurons to degenerate during PD. Targeting ion channels offers an attractive mechanistic strategy to combat the process of neurodegeneration. In this review, we highlight the evidence pointing to the role of various ion channels in driving the PD processes. In addition, we also discuss the various drugs or compounds that target the ion channels and have shown neuroprotective potential in the in-vitro and in-vivo models of PD. We also discuss the current clinical status of various drugs targeting the ion channels in the context of PD.
Collapse
Affiliation(s)
- Neha Hanna Daniel
- School of Biology, Indian Institute of Science Education and Research (IISER)-Thiruvananthapuram, Kerala, 695551, India
| | - Ananya Aravind
- School of Biology, Indian Institute of Science Education and Research (IISER)-Thiruvananthapuram, Kerala, 695551, India
| | - Poonam Thakur
- School of Biology, Indian Institute of Science Education and Research (IISER)-Thiruvananthapuram, Kerala, 695551, India.
| |
Collapse
|
34
|
Lane AR, Cogdell IC, Jessell TM, Bikoff JB, Alvarez FJ. Genetic targeting of adult Renshaw cells using a Calbindin 1 destabilized Cre allele for intersection with Parvalbumin or Engrailed1. Sci Rep 2021; 11:19861. [PMID: 34615947 PMCID: PMC8494874 DOI: 10.1038/s41598-021-99333-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 09/16/2021] [Indexed: 11/09/2022] Open
Abstract
Renshaw cells (RCs) are one of the most studied spinal interneurons; however, their roles in motor control remain enigmatic in part due to the lack of experimental models to interfere with RC function, specifically in adults. To overcome this limitation, we leveraged the distinct temporal regulation of Calbindin (Calb1) expression in RCs to create genetic models for timed RC manipulation. We used a Calb1 allele expressing a destabilized Cre (dgCre) theoretically active only upon trimethoprim (TMP) administration. TMP timing and dose influenced RC targeting efficiency, which was highest within the first three postnatal weeks, but specificity was low with many other spinal neurons also targeted. In addition, dgCre showed TMP-independent activity resulting in spontaneous recombination events that accumulated with age. Combining Calb1-dgCre with Parvalbumin (Pvalb) or Engrailed1 (En1) Flpo alleles in dual conditional systems increased cellular and timing specificity. Under optimal conditions, Calb1-dgCre/Pvalb-Flpo mice targeted 90% of RCs and few dorsal horn neurons; Calb1-dgCre/En1-Flpo mice showed higher specificity, but only a maximum of 70% of RCs targeted. Both models targeted neurons throughout the brain. Restricted spinal expression was obtained by injecting intraspinally AAVs carrying dual conditional genes. These results describe the first models to genetically target RCs bypassing development.
Collapse
Affiliation(s)
- Alicia R Lane
- Department of Physiology, Emory University, Atlanta, GA, 30322, USA
| | | | - Thomas M Jessell
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, 10032, USA
| | - Jay B Bikoff
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | | |
Collapse
|
35
|
Carmichael K, Sullivan B, Lopez E, Sun L, Cai H. Diverse midbrain dopaminergic neuron subtypes and implications for complex clinical symptoms of Parkinson's disease. AGEING AND NEURODEGENERATIVE DISEASES 2021; 1. [PMID: 34532720 PMCID: PMC8442626 DOI: 10.20517/and.2021.07] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Parkinson’s disease (PD), the most common degenerative movement disorder, is clinically manifested with various motor and non-motor symptoms. Degeneration of midbrain substantia nigra pas compacta (SNc) dopaminergic neurons (DANs) is generally attributed to the motor syndrome. The underlying neuronal mechanisms of non-motor syndrome are largely unexplored. Besides SNc, midbrain ventral tegmental area (VTA) DANs also produce and release dopamine and modulate movement, reward, motivation, and memory. Degeneration of VTA DANs also occurs in postmortem brains of PD patients, implying an involvement of VTA DANs in PD-associated non-motor symptoms. However, it remains to be established that there is a distinct segregation of different SNc and VTA DAN subtypes in regulating different motor and non-motor functions, and that different DAN subpopulations are differentially affected by normal ageing or PD. Traditionally, the distinction among different DAN subtypes was mainly based on the location of cell bodies and axon terminals. With the recent advance of single cell RNA sequencing technology, DANs can be readily classified based on unique gene expression profiles. A combination of specific anatomic and molecular markers shows great promise to facilitate the identification of DAN subpopulations corresponding to different behavior modules under normal and disease conditions. In this review, we first summarize the recent progress in characterizing genetically, anatomically, and functionally diverse midbrain DAN subtypes. Then, we provide perspectives on how the preclinical research on the connectivity and functionality of DAN subpopulations improves our current understanding of cell-type and circuit specific mechanisms of the disease, which could be critically informative for designing new mechanistic treatments.
Collapse
Affiliation(s)
- Kathleen Carmichael
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA.,The Graduate Partnership Program of NIH and Brown University, National Institutes of Health, Bethesda, MD 20892, USA
| | - Breanna Sullivan
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Elena Lopez
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lixin Sun
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Huaibin Cai
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
36
|
Knowlton CJ, Ziouziou TI, Hammer N, Roeper J, Canavier CC. Inactivation mode of sodium channels defines the different maximal firing rates of conventional versus atypical midbrain dopamine neurons. PLoS Comput Biol 2021; 17:e1009371. [PMID: 34534209 PMCID: PMC8480832 DOI: 10.1371/journal.pcbi.1009371] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 09/29/2021] [Accepted: 08/23/2021] [Indexed: 12/21/2022] Open
Abstract
Two subpopulations of midbrain dopamine (DA) neurons are known to have different dynamic firing ranges in vitro that correspond to distinct projection targets: the originally identified conventional DA neurons project to the dorsal striatum and the lateral shell of the nucleus accumbens, whereas an atypical DA population with higher maximum firing frequencies projects to prefrontal regions and other limbic regions including the medial shell of nucleus accumbens. Using a computational model, we show that previously identified differences in biophysical properties do not fully account for the larger dynamic range of the atypical population and predict that the major difference is that originally identified conventional cells have larger occupancy of voltage-gated sodium channels in a long-term inactivated state that recovers slowly; stronger sodium and potassium conductances during action potential firing are also predicted for the conventional compared to the atypical DA population. These differences in sodium channel gating imply that longer intervals between spikes are required in the conventional population for full recovery from long-term inactivation induced by the preceding spike, hence the lower maximum frequency. These same differences can also change the bifurcation structure to account for distinct modes of entry into depolarization block: abrupt versus gradual. The model predicted that in cells that have entered depolarization block, it is much more likely that an additional depolarization can evoke an action potential in conventional DA population. New experiments comparing lateral to medial shell projecting neurons confirmed this model prediction, with implications for differential synaptic integration in the two populations. We developed a theoretical and mathematical framework that could explain the major electrophysiological differences between the conventional midbrain dopamine (DA) neurons with a low maximum firing rate, and the more recently identified atypical DA neurons. Testable predictions from this framework were then verified with in vitro patch-clamp recordings from DA neurons with identified phenotypes and projection targets. Since different subpopulations of DA neurons participate in different circuits, and these circuits are likely differentially dysregulated in diseases such as addiction, Parkinson disease, and schizophrenia, it is important to identify the differences of their intrinsic electrophysiological properties as a prelude to developing more precisely targeted therapies.
Collapse
Affiliation(s)
- Christopher J. Knowlton
- Department of Cell Biology and Anatomy, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
| | | | - Niklas Hammer
- Institut für Neurophysiologie, Goethe University, Frankfurt, Germany
| | - Jochen Roeper
- Institut für Neurophysiologie, Goethe University, Frankfurt, Germany
| | - Carmen C. Canavier
- Department of Cell Biology and Anatomy, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
- * E-mail:
| |
Collapse
|
37
|
The gating pore blocker 1-(2,4-xylyl)guanidinium selectively inhibits pacemaking of midbrain dopaminergic neurons. Neuropharmacology 2021; 197:108722. [PMID: 34273387 DOI: 10.1016/j.neuropharm.2021.108722] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 07/08/2021] [Accepted: 07/13/2021] [Indexed: 11/22/2022]
Abstract
Although several ionic mechanisms are known to control rate and regularity of the slow pacemaker in dopamine (DA) neurons, the core mechanism of pacing is controversial. Here we tested the hypothesis that pacemaking of SNc DA neurons is enabled by an unconventional conductance. We found that 1-(2,4-xylyl)guanidinium (XG), an established blocker of gating pore currents, selectively inhibits pacemaking of DA neurons. The compound inhibited all slow pacemaking DA neurons that were tested, both in the substantia nigra pars compacta, and in the ventral tegmental area. Interestingly, bursting behavior was not affected by XG. Furthermore, the drug did not affect fast pacemaking of GABAergic neurons from substantia nigra pars reticulata neurons or slow pacemaking of noradrenergic neurons. In DA neurons, current-clamp analysis revealed that XG did not appear to affect ion channels involved in the action potential. Its inhibitory effect persisted during blockade of all ion channels previously suggested to contribute to pacemaking. RNA sequencing and voltage-clamp recordings yielded no evidence for a gating pore current to underlie the conductance. However, we could isolate a small subthreshold XG-sensitive current, which was carried by both Na+ and Cl- ions. Although the molecular target of XG remains to be defined, these observations represent a step towards understanding pacemaking in DA neurons.
Collapse
|
38
|
Miller DR, Guenther DT, Maurer AP, Hansen CA, Zalesky A, Khoshbouei H. Dopamine Transporter Is a Master Regulator of Dopaminergic Neural Network Connectivity. J Neurosci 2021; 41:5453-5470. [PMID: 33980544 PMCID: PMC8221606 DOI: 10.1523/jneurosci.0223-21.2021] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/19/2021] [Accepted: 05/01/2021] [Indexed: 12/13/2022] Open
Abstract
Dopaminergic neurons of the substantia nigra pars compacta (SNC) and ventral tegmental area (VTA) exhibit spontaneous firing activity. The dopaminergic neurons in these regions have been shown to exhibit differential sensitivity to neuronal loss and psychostimulants targeting dopamine transporter. However, it remains unclear whether these regional differences scale beyond individual neuronal activity to regional neuronal networks. Here, we used live-cell calcium imaging to show that network connectivity greatly differs between SNC and VTA regions with higher incidence of hub-like neurons in the VTA. Specifically, the frequency of hub-like neurons was significantly lower in SNC than in the adjacent VTA, consistent with the interpretation of a lower network resilience to SNC neuronal loss. We tested this hypothesis, in DAT-cre/loxP-GCaMP6f mice of either sex, when activity of an individual dopaminergic neuron is suppressed, through whole-cell patch clamp electrophysiology, in either SNC or VTA networks. Neuronal loss in the SNC increased network clustering, whereas the larger number of hub-neurons in the VTA overcompensated by decreasing network clustering in the VTA. We further show that network properties are regulatable via a dopamine transporter but not a D2 receptor dependent mechanism. Our results demonstrate novel regulatory mechanisms of functional network topology in dopaminergic brain regions.SIGNIFICANCE STATEMENT In this work, we begin to untangle the differences in complex network properties between the substantia nigra pars compacta (SNC) and VTA, that may underlie differential sensitivity between regions. The methods and analysis employed provide a springboard for investigations of network topology in multiple deep brain structures and disorders.
Collapse
Affiliation(s)
- Douglas R Miller
- Department of Neuroscience, University of Florida, Gainesville, Florida
| | - Dylan T Guenther
- Department of Neuroscience, University of Florida, Gainesville, Florida
| | - Andrew P Maurer
- Department of Neuroscience, University of Florida, Gainesville, Florida
| | - Carissa A Hansen
- Department of Neuroscience, University of Florida, Gainesville, Florida
| | - Andrew Zalesky
- Melbourne Neuropsychiatry Centre, The University of Melbourne and Melbourne Health, Melbourne, Victoria 3010, Australia
- Department of Biomedical Engineering, Melbourne School of Engineering, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | | |
Collapse
|
39
|
Chen APF, Chen L, Kim TA, Xiong Q. Integrating the Roles of Midbrain Dopamine Circuits in Behavior and Neuropsychiatric Disease. Biomedicines 2021; 9:biomedicines9060647. [PMID: 34200134 PMCID: PMC8228225 DOI: 10.3390/biomedicines9060647] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/01/2021] [Accepted: 06/03/2021] [Indexed: 01/11/2023] Open
Abstract
Dopamine (DA) is a behaviorally and clinically diverse neuromodulator that controls CNS function. DA plays major roles in many behaviors including locomotion, learning, habit formation, perception, and memory processing. Reflecting this, DA dysregulation produces a wide variety of cognitive symptoms seen in neuropsychiatric diseases such as Parkinson’s, Schizophrenia, addiction, and Alzheimer’s disease. Here, we review recent advances in the DA systems neuroscience field and explore the advancing hypothesis that DA’s behavioral function is linked to disease deficits in a neural circuit-dependent manner. We survey different brain areas including the basal ganglia’s dorsomedial/dorsolateral striatum, the ventral striatum, the auditory striatum, and the hippocampus in rodent models. Each of these regions have different reported functions and, correspondingly, DA’s reflecting role in each of these regions also has support for being different. We then focus on DA dysregulation states in Parkinson’s disease, addiction, and Alzheimer’s Disease, emphasizing how these afflictions are linked to different DA pathways. We draw upon ideas such as selective vulnerability and region-dependent physiology. These bodies of work suggest that different channels of DA may be dysregulated in different sets of disease. While these are great advances, the fine and definitive segregation of such pathways in behavior and disease remains to be seen. Future studies will be required to define DA’s necessity and contribution to the functional plasticity of different striatal regions.
Collapse
Affiliation(s)
- Allen PF Chen
- Department of Neurobiology and Behavior, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794, USA; (A.P.C.); (L.C.); (T.A.K.)
- Medical Scientist Training Program, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794, USA
| | - Lu Chen
- Department of Neurobiology and Behavior, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794, USA; (A.P.C.); (L.C.); (T.A.K.)
| | - Thomas A. Kim
- Department of Neurobiology and Behavior, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794, USA; (A.P.C.); (L.C.); (T.A.K.)
- Medical Scientist Training Program, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794, USA
| | - Qiaojie Xiong
- Department of Neurobiology and Behavior, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794, USA; (A.P.C.); (L.C.); (T.A.K.)
- Correspondence:
| |
Collapse
|
40
|
Carmichael K, Evans RC, Lopez E, Sun L, Kumar M, Ding J, Khaliq ZM, Cai H. Function and Regulation of ALDH1A1-Positive Nigrostriatal Dopaminergic Neurons in Motor Control and Parkinson's Disease. Front Neural Circuits 2021; 15:644776. [PMID: 34079441 PMCID: PMC8165242 DOI: 10.3389/fncir.2021.644776] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 04/26/2021] [Indexed: 12/13/2022] Open
Abstract
Dopamine is an important chemical messenger in the brain, which modulates movement, reward, motivation, and memory. Different populations of neurons can produce and release dopamine in the brain and regulate different behaviors. Here we focus our discussion on a small but distinct group of dopamine-producing neurons, which display the most profound loss in the ventral substantia nigra pas compacta of patients with Parkinson's disease. This group of dopaminergic neurons can be readily identified by a selective expression of aldehyde dehydrogenase 1A1 (ALDH1A1) and accounts for 70% of total nigrostriatal dopaminergic neurons in both human and mouse brains. Recently, we presented the first whole-brain circuit map of these ALDH1A1-positive dopaminergic neurons and reveal an essential physiological function of these neurons in regulating the vigor of movement during the acquisition of motor skills. In this review, we first summarize previous findings of ALDH1A1-positive nigrostriatal dopaminergic neurons and their connectivity and functionality, and then provide perspectives on how the activity of ALDH1A1-positive nigrostriatal dopaminergic neurons is regulated through integrating diverse presynaptic inputs and its implications for potential Parkinson's disease treatment.
Collapse
Affiliation(s)
- Kathleen Carmichael
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, United States
- The Graduate Partnership Program of NIH and Brown University, National Institutes of Health, Bethesda, MD, United States
| | - Rebekah C. Evans
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, United States
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Elena Lopez
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, United States
| | - Lixin Sun
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, United States
| | - Mantosh Kumar
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, United States
| | - Jinhui Ding
- Computational Biology Group, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, United States
| | - Zayd M. Khaliq
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Huaibin Cai
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
41
|
Ortner NJ. Voltage-Gated Ca 2+ Channels in Dopaminergic Substantia Nigra Neurons: Therapeutic Targets for Neuroprotection in Parkinson's Disease? Front Synaptic Neurosci 2021; 13:636103. [PMID: 33716705 PMCID: PMC7952618 DOI: 10.3389/fnsyn.2021.636103] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/25/2021] [Indexed: 12/21/2022] Open
Abstract
The loss of dopamine (DA)-producing neurons in the substantia nigra pars compacta (SN) underlies the core motor symptoms of the progressive movement disorder Parkinson's disease (PD). To date, no treatment to prevent or slow SN DA neurodegeneration exists; thus, the identification of the underlying factors contributing to the high vulnerability of these neurons represents the basis for the development of novel therapies. Disrupted Ca2+ homeostasis and mitochondrial dysfunction seem to be key players in the pathophysiology of PD. The autonomous pacemaker activity of SN DA neurons, in combination with low cytosolic Ca2+ buffering, leads to large somatodendritic fluctuations of intracellular Ca2+ levels that are linked to elevated mitochondrial oxidant stress. L-type voltage-gated Ca2+ channels (LTCCs) contribute to these Ca2+ oscillations in dendrites, and LTCC inhibition was beneficial in cellular and in vivo animal models of PD. However, in a recently completed phase 3 clinical trial, the dihydropyridine (DHP) LTCC inhibitor isradipine failed to slow disease progression in early PD patients, questioning the feasibility of DHPs for PD therapy. Novel evidence also suggests that R- and T-type Ca2+ channels (RTCCs and TTCCs, respectively) represent potential PD drug targets. This short review aims to (re)evaluate the therapeutic potential of LTCC, RTCC, and TTCC inhibition in light of novel preclinical and clinical data and the feasibility of available Ca2+ channel blockers to modify PD disease progression. I also summarize their cell-specific roles for SN DA neuron function and describe how their gating properties allow activity (and thus their contribution to stressful Ca2+ oscillations) during pacemaking.
Collapse
Affiliation(s)
- Nadine J. Ortner
- Department of Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
42
|
Mannal N, Kleiner K, Fauler M, Dougalis A, Poetschke C, Liss B. Multi-Electrode Array Analysis Identifies Complex Dopamine Responses and Glucose Sensing Properties of Substantia Nigra Neurons in Mouse Brain Slices. Front Synaptic Neurosci 2021; 13:635050. [PMID: 33716704 PMCID: PMC7952765 DOI: 10.3389/fnsyn.2021.635050] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 01/08/2021] [Indexed: 12/16/2022] Open
Abstract
Dopaminergic (DA) midbrain neurons within the substantia nigra (SN) display an autonomous pacemaker activity that is crucial for dopamine release and voluntary movement control. Their progressive degeneration is a hallmark of Parkinson's disease. Their metabolically demanding activity-mode affects Ca2+ homeostasis, elevates metabolic stress, and renders SN DA neurons particularly vulnerable to degenerative stressors. Accordingly, their activity is regulated by complex mechanisms, notably by dopamine itself, via inhibitory D2-autoreceptors and the neuroprotective neuronal Ca2+ sensor NCS-1. Analyzing regulation of SN DA neuron activity-pattern is complicated by their high vulnerability. We studied this activity and its control by dopamine, NCS-1, and glucose with extracellular multi-electrode array (MEA) recordings from midbrain slices of juvenile and adult mice. Our tailored MEA- and spike sorting-protocols allowed high throughput and long recording times. According to individual dopamine-responses, we identified two distinct SN cell-types, in similar frequency: dopamine-inhibited and dopamine-excited neurons. Dopamine-excited neurons were either silent in the absence of dopamine, or they displayed pacemaker-activities, similar to that of dopamine-inhibited neurons. Inhibition of pacemaker-activity by dopamine is typical for SN DA neurons, and it can undergo prominent desensitization. We show for adult mice, that the number of SN DA neurons with desensitized dopamine-inhibition was increased (~60–100%) by a knockout of NCS-1, or by prevention of NCS-1 binding to D2-autoreceptors, while time-course and degrees of desensitization were not altered. The number of neurons with desensitized D2-responses was also higher (~65%) at high glucose-levels (25 mM), compared to lower glucose (2.5 mM), while again desensitization-kinetics were unaltered. However, spontaneous firing-rates were significantly higher at high glucose-levels (~20%). Moreover, transient glucose-deprivation (1 mM) induced a fast and fully-reversible pacemaker frequency reduction. To directly address and quantify glucose-sensing properties of SN DA neurons, we continuously monitored their electrical activity, while altering extracellular glucose concentrations stepwise from 0.5 mM up to 25 mM. SN DA neurons were excited by glucose, with EC50 values ranging from 0.35 to 2.3 mM. In conclusion, we identified a novel, common subtype of dopamine-excited SN neurons, and a complex, joint regulation of dopamine-inhibited neurons by dopamine and glucose, within the range of physiological brain glucose-levels.
Collapse
Affiliation(s)
- Nadja Mannal
- Institute of Applied Physiology, University of Ulm, Ulm, Germany
| | | | - Michael Fauler
- Institute of Applied Physiology, University of Ulm, Ulm, Germany
| | | | | | - Birgit Liss
- Institute of Applied Physiology, University of Ulm, Ulm, Germany.,Linacre and New College, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
43
|
Tsutsui-Kimura I, Matsumoto H, Akiti K, Yamada MM, Uchida N, Watabe-Uchida M. Distinct temporal difference error signals in dopamine axons in three regions of the striatum in a decision-making task. eLife 2020; 9:e62390. [PMID: 33345774 PMCID: PMC7771962 DOI: 10.7554/elife.62390] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 12/18/2020] [Indexed: 12/24/2022] Open
Abstract
Different regions of the striatum regulate different types of behavior. However, how dopamine signals differ across striatal regions and how dopamine regulates different behaviors remain unclear. Here, we compared dopamine axon activity in the ventral, dorsomedial, and dorsolateral striatum, while mice performed a perceptual and value-based decision task. Surprisingly, dopamine axon activity was similar across all three areas. At a glance, the activity multiplexed different variables such as stimulus-associated values, confidence, and reward feedback at different phases of the task. Our modeling demonstrates, however, that these modulations can be inclusively explained by moment-by-moment changes in the expected reward, that is the temporal difference error. A major difference between areas was the overall activity level of reward responses: reward responses in dorsolateral striatum were positively shifted, lacking inhibitory responses to negative prediction errors. The differences in dopamine signals put specific constraints on the properties of behaviors controlled by dopamine in these regions.
Collapse
Affiliation(s)
- Iku Tsutsui-Kimura
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard UniversityCambridgeUnited States
| | - Hideyuki Matsumoto
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard UniversityCambridgeUnited States
- Department of Physiology, Osaka City University Graduate School of MedicineOsakaJapan
| | - Korleki Akiti
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard UniversityCambridgeUnited States
| | - Melissa M Yamada
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard UniversityCambridgeUnited States
| | - Naoshige Uchida
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard UniversityCambridgeUnited States
| | - Mitsuko Watabe-Uchida
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard UniversityCambridgeUnited States
| |
Collapse
|
44
|
Otomo K, Perkins J, Kulkarni A, Stojanovic S, Roeper J, Paladini CA. In vivo patch-clamp recordings reveal distinct subthreshold signatures and threshold dynamics of midbrain dopamine neurons. Nat Commun 2020; 11:6286. [PMID: 33293613 PMCID: PMC7722714 DOI: 10.1038/s41467-020-20041-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 11/06/2020] [Indexed: 01/19/2023] Open
Abstract
The in vivo firing patterns of ventral midbrain dopamine neurons are controlled by afferent and intrinsic activity to generate sensory cue and prediction error signals that are essential for reward-based learning. Given the absence of in vivo intracellular recordings during the last three decades, the subthreshold membrane potential events that cause changes in dopamine neuron firing patterns remain unknown. To address this, we established in vivo whole-cell recordings and obtained over 100 spontaneously active, immunocytochemically-defined midbrain dopamine neurons in isoflurane-anaesthetized adult mice. We identified a repertoire of subthreshold membrane potential signatures associated with distinct in vivo firing patterns. Dopamine neuron activity in vivo deviated from single-spike pacemaking by phasic increases in firing rate via two qualitatively distinct biophysical mechanisms: 1) a prolonged hyperpolarization preceding rebound bursts, accompanied by a hyperpolarizing shift in action potential threshold; and 2) a transient depolarization leading to high-frequency plateau bursts, associated with a depolarizing shift in action potential threshold. Our findings define a mechanistic framework for the biophysical implementation of dopamine neuron firing patterns in the intact brain.
Collapse
Affiliation(s)
- Kanako Otomo
- Institute of Neurophysiology, Neuroscience Center, Goethe University, Frankfurt, Germany
| | - Jessica Perkins
- University of Texas at San Antonio Neurosciences Institute, San Antonio, USA
| | - Anand Kulkarni
- University of Texas at San Antonio Neurosciences Institute, San Antonio, USA
| | - Strahinja Stojanovic
- Institute of Neurophysiology, Neuroscience Center, Goethe University, Frankfurt, Germany
| | - Jochen Roeper
- Institute of Neurophysiology, Neuroscience Center, Goethe University, Frankfurt, Germany.
| | - Carlos A Paladini
- University of Texas at San Antonio Neurosciences Institute, San Antonio, USA.
| |
Collapse
|
45
|
Howell RD, Dominguez-Lopez S, Ocañas SR, Freeman WM, Beckstead MJ. Female mice are resilient to age-related decline of substantia nigra dopamine neuron firing parameters. Neurobiol Aging 2020; 95:195-204. [PMID: 32846275 PMCID: PMC7606778 DOI: 10.1016/j.neurobiolaging.2020.07.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 06/03/2020] [Accepted: 07/25/2020] [Indexed: 02/06/2023]
Abstract
Degeneration of substantia nigra pars compacta dopamine neurons is a central feature in the pathology of Parkinson's disease, which is characterized by progressive loss of motor and cognitive functions. The largest risk factors for Parkinson's disease are age and sex; most cases occur after age 60 and males have nearly twice the incidence as females. Preclinical work has scarcely considered the influence of these 2 factors to disease risk and presentation. Here, we observed a progressive decline in dopamine neuron firing activity in male C57BL/6 mice by 18 months of age, while dopamine neurons from females remained largely unaffected. This was accompanied by increased mRNA expression of PINK1 in both males and females, and PARK2 primarily in males, both of which have been linked to Parkinson's. Since the declining cell properties were accompanied by only slight decreases in locomotion in both sexes, it is likely that these age-related impairments in males represent a vulnerability to further insults that could predispose the neurons to neurodegenerative processes such as in Parkinson's.
Collapse
Affiliation(s)
- Rebecca D Howell
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK; Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Sergio Dominguez-Lopez
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Sarah R Ocañas
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK; Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Willard M Freeman
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Michael J Beckstead
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK.
| |
Collapse
|
46
|
Xiong M, Tao Y, Gao Q, Feng B, Yan W, Zhou Y, Kotsonis TA, Yuan T, You Z, Wu Z, Xi J, Haberman A, Graham J, Block J, Zhou W, Chen Y, Zhang SC. Human Stem Cell-Derived Neurons Repair Circuits and Restore Neural Function. Cell Stem Cell 2020; 28:112-126.e6. [PMID: 32966778 DOI: 10.1016/j.stem.2020.08.014] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 05/11/2020] [Accepted: 08/21/2020] [Indexed: 02/02/2023]
Abstract
Although cell transplantation can rescue motor defects in Parkinson's disease (PD) models, whether and how grafts functionally repair damaged neural circuitry in the adult brain is not known. We transplanted hESC-derived midbrain dopamine (mDA) or cortical glutamate neurons into the substantia nigra or striatum of a mouse PD model and found extensive graft integration with host circuitry. Axonal pathfinding toward the dorsal striatum was determined by the identity of the grafted neurons, and anatomical presynaptic inputs were largely dependent on graft location, whereas inhibitory versus excitatory input was dictated by the identity of grafted neurons. hESC-derived mDA neurons display A9 characteristics and restore functionality of the reconstructed nigrostriatal circuit to mediate improvements in motor function. These results indicate similarity in cell-type-specific pre- and post-synaptic integration between transplant-reconstructed circuit and endogenous neural networks, highlighting the capacity of hPSC-derived neuron subtypes for specific circuit repair and functional restoration in the adult brain.
Collapse
Affiliation(s)
- Man Xiong
- Institute of Pediatrics, Children's Hospital, Fudan University, 399 Wanyuan Road, Shanghai 201102, China
| | - Yezheng Tao
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Program in Neuroscience & Behavioral Disorders, Duke-NUS Medical School, 169857 Singapore, Singapore
| | - Qinqin Gao
- Institute of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ban Feng
- Institute of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wei Yan
- Institute of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Yingying Zhou
- Institute of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Thomas A Kotsonis
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Tingli Yuan
- Institute of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhiwen You
- Institute of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Ziyan Wu
- Institute of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jiajie Xi
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | | | - Julia Graham
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jasper Block
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Wenhao Zhou
- Institute of Pediatrics, Children's Hospital, Fudan University, 399 Wanyuan Road, Shanghai 201102, China
| | - Yuejun Chen
- Institute of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai 201210, China.
| | - Su-Chun Zhang
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neurology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; Program in Neuroscience & Behavioral Disorders, Duke-NUS Medical School, 169857 Singapore, Singapore
| |
Collapse
|
47
|
Evans RC, Twedell EL, Zhu M, Ascencio J, Zhang R, Khaliq ZM. Functional Dissection of Basal Ganglia Inhibitory Inputs onto Substantia Nigra Dopaminergic Neurons. Cell Rep 2020; 32:108156. [PMID: 32937133 PMCID: PMC9887718 DOI: 10.1016/j.celrep.2020.108156] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 07/11/2020] [Accepted: 08/25/2020] [Indexed: 02/02/2023] Open
Abstract
Substantia nigra (SNc) dopaminergic neurons respond to aversive stimuli with inhibitory pauses in firing followed by transient rebound activation. We tested integration of inhibitory synaptic inputs onto SNc neurons from genetically defined populations in dorsal striatum (striosome and matrix) and external globus pallidus (GPe; parvalbumin- and Lhx6-positive), and examined their contribution to pause-rebound firing. Activation of striosome projections, which target "dendron bouquets" in the pars reticulata (SNr), consistently quiets firing and relief from striosome inhibition triggers rebound activity. Striosomal inhibitory postsynaptic currents (IPSCs) display a prominent GABA-B receptor-mediated component that strengthens the impact of SNr dendrite synapses on somatic excitability and enables rebounding. By contrast, GPe projections activate GABA-A receptors on the soma and proximal dendrites but do not result in rebounding. Lastly, optical mapping shows that dorsal striatum selectively inhibits the ventral population of SNc neurons, which are intrinsically capable of rebounding. Therefore, we define a distinct striatonigral circuit for generating dopamine rebound.
Collapse
Affiliation(s)
- Rebekah C. Evans
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Emily L. Twedell
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Manhua Zhu
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jefferson Ascencio
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Renshu Zhang
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zayd M. Khaliq
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA,Lead Contact,Correspondence:
| |
Collapse
|
48
|
Zampese E, Surmeier DJ. Calcium, Bioenergetics, and Parkinson's Disease. Cells 2020; 9:cells9092045. [PMID: 32911641 PMCID: PMC7564460 DOI: 10.3390/cells9092045] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/04/2020] [Accepted: 09/07/2020] [Indexed: 12/12/2022] Open
Abstract
Degeneration of substantia nigra (SN) dopaminergic (DAergic) neurons is responsible for the core motor deficits of Parkinson’s disease (PD). These neurons are autonomous pacemakers that have large cytosolic Ca2+ oscillations that have been linked to basal mitochondrial oxidant stress and turnover. This review explores the origin of Ca2+ oscillations and their role in the control of mitochondrial respiration, bioenergetics, and mitochondrial oxidant stress.
Collapse
|
49
|
Matsushima A, Graybiel AM. Combinatorial Developmental Controls on Striatonigral Circuits. Cell Rep 2020; 31:107778. [PMID: 32553154 PMCID: PMC7433760 DOI: 10.1016/j.celrep.2020.107778] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 02/12/2020] [Accepted: 05/27/2020] [Indexed: 11/17/2022] Open
Abstract
Cortical pyramidal cells are generated locally, from pre-programmed progenitors, to form functionally distinct areas. By contrast, striatal projection neurons (SPNs) are generated remotely from a common source, undergo migration to form mosaics of striosomes and matrix, and become incorporated into functionally distinct sectors. Striatal circuits might thus have a unique logic of developmental organization, distinct from those of the neocortex. We explore this possibility in mice by mapping one set of SPNs, those in striosomes, with striatonigral projections to the dopamine-containing substantia nigra pars compacta (SNpc). Same-age SPNs exhibit topographic striatonigral projections, according to their resident sector. However, the different birth dates of resident SPNs within a given sector specify the destination of their axons within the SNpc. These findings highlight a logic intercalating birth date-dependent and birth date-independent factors in determining the trajectories of SPN axons and organizing specialized units of striatonigral circuitry that could influence behavioral expression and vulnerabilities to disease.
Collapse
Affiliation(s)
- Ayano Matsushima
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 20139, USA
| | - Ann M Graybiel
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 20139, USA.
| |
Collapse
|
50
|
Poulin JF, Gaertner Z, Moreno-Ramos OA, Awatramani R. Classification of Midbrain Dopamine Neurons Using Single-Cell Gene Expression Profiling Approaches. Trends Neurosci 2020; 43:155-169. [PMID: 32101709 PMCID: PMC7285906 DOI: 10.1016/j.tins.2020.01.004] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 12/13/2019] [Accepted: 01/11/2020] [Indexed: 01/31/2023]
Abstract
Dysfunctional dopamine (DA) signaling has been associated with a broad spectrum of neuropsychiatric disorders, prompting investigations into how midbrain DA neuron heterogeneity may underpin this variety of behavioral symptoms. Emerging literature indeed points to functional heterogeneity even within anatomically defined DA clusters. Recognizing the need for a systematic classification scheme, several groups have used single-cell profiling to catalog DA neurons based on their gene expression profiles. We aim here not only to synthesize points of congruence but also to highlight key differences between the molecular classification schemes derived from these studies. In doing so, we hope to provide a common framework that will facilitate investigations into the functions of DA neuron subtypes in the healthy and diseased brain.
Collapse
Affiliation(s)
- Jean-Francois Poulin
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Zachary Gaertner
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | - Rajeshwar Awatramani
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|