1
|
Lin YE, Jaimon E, Tonelli F, Pfeffer SR. Pathogenic LRRK2 mutations cause loss of primary cilia and Neurturin in striatal parvalbumin interneurons. Life Sci Alliance 2025; 8:e202402922. [PMID: 39537338 PMCID: PMC11561259 DOI: 10.26508/lsa.202402922] [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: 07/02/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Parkinson's disease-associated, activating mutations in the LRRK2 kinase block primary cilium formation in cell culture and in specific cell types in the brain. In the striatum that is important for movement control, about half of astrocytes and cholinergic interneurons, but not the predominant medium spiny neurons, lose their primary cilia. Here, we show that mouse and human striatal parvalbumin interneurons that are inhibitory regulators of movement also lose primary cilia. Without cilia, these neurons are not able to respond to Sonic hedgehog signals that normally induce the expression of Patched RNA, and their numbers decrease. In addition, in mouse, glial cell line-derived neurotrophic factor-related Neurturin RNA is significantly decreased. These experiments highlight the importance of parvalbumin neurons in cilium-dependent, neuroprotective signaling pathways and show that LRRK2 activation correlates with decreased Neurturin production, resulting in less neuroprotection for dopamine neurons.
Collapse
Affiliation(s)
- Yu-En Lin
- https://ror.org/00f54p054 Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Bethesda, MD, USA
| | - Ebsy Jaimon
- https://ror.org/00f54p054 Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Bethesda, MD, USA
| | - Francesca Tonelli
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Bethesda, MD, USA
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Scotland, UK
| | - Suzanne R Pfeffer
- https://ror.org/00f54p054 Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Bethesda, MD, USA
| |
Collapse
|
2
|
Cepeda C, Holley SM, Barry J, Oikonomou KD, Yazon VW, Peng A, Argueta D, Levine MS. Corticostriatal maldevelopment in the R6/2 mouse model of juvenile Huntington's disease. Neurobiol Dis 2024; 204:106752. [PMID: 39644979 DOI: 10.1016/j.nbd.2024.106752] [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: 10/07/2024] [Revised: 11/22/2024] [Accepted: 11/24/2024] [Indexed: 12/09/2024] Open
Abstract
There is a growing consensus that brain development in Huntington's disease (HD) is abnormal, leading to the idea that HD is not only a neurodegenerative but also a neurodevelopmental disorder. Indeed, structural and functional abnormalities have been observed during brain development in both humans and animal models of HD. However, a concurrent study of cortical and striatal development in a genetic model of HD is still lacking. Here we report significant alterations of corticostriatal development in the R6/2 mouse model of juvenile HD. We examined wildtype (WT) and R6/2 mice at postnatal (P) days 7, 14, and 21. Morphological examination demonstrated early structural and cellular alterations reminiscent of malformations of cortical development, and ex vivo electrophysiological recordings of cortical pyramidal neurons (CPNs) demonstrated significant age- and genotype-dependent changes of intrinsic membrane and synaptic properties. In general, R6/2 CPNs had reduced cell membrane capacitance and increased input resistance (P7 and P14), along with reduced frequency of spontaneous excitatory and inhibitory synaptic events during early development (P7), suggesting delayed cortical maturation. This was confirmed by increased occurrence of GABAA receptor-mediated giant depolarizing potentials at P7. At P14, the rheobase of CPNs was significantly reduced, along with increased excitability. Altered membrane and synaptic properties of R6/2 CPNs recovered progressively, and by P21 they were similar to WT CPNs. In striatal medium-sized spiny neurons (MSNs), a different picture emerged. Intrinsic membrane properties were relatively normal throughout development, except for a transient increase in membrane capacitance at P14. The first alterations in MSNs synaptic activity were observed at P14 and consisted of significant deficits in GABAergic inputs, however, these also were normalized by P21. In contrast, excitatory inputs began to decrease at this age. We conclude that the developing HD brain is capable of compensating for early developmental abnormalities and that cortical alterations precede and are a main contributor of striatal changes. Addressing cortical maldevelopment could help prevent or delay disease manifestations.
Collapse
Affiliation(s)
- Carlos Cepeda
- IDDRC, Jane and Terry Semel Institute for Neuroscience & Human Behavior, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA.
| | - Sandra M Holley
- IDDRC, Jane and Terry Semel Institute for Neuroscience & Human Behavior, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| | - Joshua Barry
- IDDRC, Jane and Terry Semel Institute for Neuroscience & Human Behavior, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| | - Katerina D Oikonomou
- IDDRC, Jane and Terry Semel Institute for Neuroscience & Human Behavior, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| | - Vannah-Wila Yazon
- IDDRC, Jane and Terry Semel Institute for Neuroscience & Human Behavior, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| | - Allison Peng
- IDDRC, Jane and Terry Semel Institute for Neuroscience & Human Behavior, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| | - Deneen Argueta
- IDDRC, Jane and Terry Semel Institute for Neuroscience & Human Behavior, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| | - Michael S Levine
- IDDRC, Jane and Terry Semel Institute for Neuroscience & Human Behavior, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
3
|
Ibáñez-Sandoval DN, Hidalgo-Balbuena AE, Velázquez Contreras R, Saderi N, Flores G, Rueda-Orozco PE, Ibáñez-Sandoval O. Striatal Interneuron Imbalance in a Valproic Acid-Induced Model of Autism in Rodents Is Accompanied by Atypical Somatosensory Processing. eNeuro 2024; 11:ENEURO.0326-24.2024. [PMID: 39572246 DOI: 10.1523/eneuro.0326-24.2024] [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: 07/21/2024] [Revised: 10/29/2024] [Accepted: 11/14/2024] [Indexed: 12/16/2024] Open
Abstract
Autism spectrum disorder (ASD) is characterized by deficits in social interaction and communication, cognitive rigidity, and atypical sensory processing. Recent studies suggest that the basal ganglia, specifically the striatum (NSt), plays an important role in ASD. While striatal interneurons, including cholinergic (ChAT+) and parvalbumin-positive (PV+) GABAergic neurons, have been described to be altered in animal models of ASD, their specific contribution remains elusive. Here, we combined behavioral, anatomical, and electrophysiological quantifications to explore if interneuron balance could be implicated in atypical sensory processing in cortical and striatal somatosensory regions of rats subjected to a valproic acid (VPA) model of ASD. We found that VPA animals showed a significant decrease in the number of ChAT+ and PV+ cells in multiple regions (including the sensorimotor region) of the NSt. We also observed significantly different sensory-evoked responses at the single-neuron and population levels in both striatal and cortical regions, as well as corticostriatal interactions. Therefore, selective elimination of striatal PV+ neurons only partially recapitulated the effects of VPA, indicating that the mechanisms behind the VPA phenotype are much more complex than the elimination of a particular neural subpopulation. Our results indicate that VPA exposure induced significant histological changes in ChAT+ and PV+ cells accompanied by atypical sensory-evoked corticostriatal population dynamics that could partially explain the sensory processing differences associated with ASD.
Collapse
Affiliation(s)
- Dayna N Ibáñez-Sandoval
- Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí 78210, México
| | - Ana E Hidalgo-Balbuena
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, UNAM, Querétaro 76230, México
| | | | - Nadia Saderi
- Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí 78295, México
| | - Gonzalo Flores
- Instituto de Fisiología, Universidad Autónoma de Puebla, Puebla CP 72570, México
| | - Pavel E Rueda-Orozco
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, UNAM, Querétaro 76230, México
| | - Osvaldo Ibáñez-Sandoval
- Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí 78210, México
| |
Collapse
|
4
|
Yonk AJ, Linares-García I, Pasternak L, Juliani SE, Gradwell MA, George AJ, Margolis DJ. Role of Posterior Medial Thalamus in the Modulation of Striatal Circuitry and Choice Behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.21.586152. [PMID: 38585753 PMCID: PMC10996534 DOI: 10.1101/2024.03.21.586152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
The posterior medial (POm) thalamus is heavily interconnected with sensory and motor circuitry and is likely involved in behavioral modulation and sensorimotor integration. POm provides axonal projections to the dorsal striatum, a hotspot of sensorimotor processing, yet the role of POm-striatal projections has remained undetermined. Using optogenetics with slice electrophysiology, we found that POm provides robust synaptic input to direct and indirect pathway striatal spiny projection neurons (D1- and D2-SPNs, respectively) and parvalbumin-expressing fast spiking interneurons (PVs). During the performance of a whisker-based tactile discrimination task, POm-striatal projections displayed learning-related activation correlating with anticipatory, but not reward-related, pupil dilation. Inhibition of POm-striatal axons across learning caused slower reaction times and an increase in the number of training sessions for expert performance. Our data indicate that POm-striatal inputs provide a behaviorally relevant arousal-related signal, which may prime striatal circuitry for efficient integration of subsequent choice-related inputs.
Collapse
Affiliation(s)
- Alex J. Yonk
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, 08854, USA
| | - Ivan Linares-García
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, 08854, USA
| | - Logan Pasternak
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, 08854, USA
| | - Sofia E. Juliani
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, 08854, USA
| | - Mark A. Gradwell
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, 08854, USA
| | - Arlene J. George
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, 08854, USA
| | - David J. Margolis
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, 08854, USA
| |
Collapse
|
5
|
Twedell EL, Bair-Marshall CJ, Girasole AE, Scaria LK, Sridhar S, Nelson AB. Striatal lateral inhibition regulates action selection in a mouse model of levodopa-induced dyskinesia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.11.617939. [PMID: 39416118 PMCID: PMC11482940 DOI: 10.1101/2024.10.11.617939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Striatal medium spiny neurons (MSNs) integrate multiple external inputs to shape motor output. In addition, MSNs form local inhibitory synaptic connections with one another. The function of striatal lateral inhibition is unknown, but one possibility is in selecting an intended action while suppressing alternatives. Action selection is disrupted in several movement disorders, including levodopa-induced dyskinesia (LID), a complication of Parkinson's disease (PD) therapy characterized by involuntary movements. Here, we identify chronic changes in the strength of striatal lateral inhibitory synapses in a mouse model of PD/LID. These synapses are also modulated by acute dopamine signaling. Chemogenetic suppression of lateral inhibition originating from dopamine D2 receptor-expressing MSNs lowers the threshold to develop involuntary movements in vivo, supporting a role in motor control. By examining the role of lateral inhibition in basal ganglia function and dysfunction, we expand the framework surrounding the role of striatal microcircuitry in action selection.
Collapse
Affiliation(s)
- Emily L Twedell
- Neuroscience Graduate Program, UCSF, San Francisco, CA 94158, USA
- Kavli Institute for Fundamental Neuroscience, UCSF, San Francisco, CA 94158, USA
- Weill Institute for Neurosciences, UCSF, San Francisco, CA 94158, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Chloe J Bair-Marshall
- Kavli Institute for Fundamental Neuroscience, UCSF, San Francisco, CA 94158, USA
- Weill Institute for Neurosciences, UCSF, San Francisco, CA 94158, USA
- Department of Neurology, UCSF, San Francisco, CA 94158, USA
| | - Allison E Girasole
- Neuroscience Graduate Program, UCSF, San Francisco, CA 94158, USA
- Kavli Institute for Fundamental Neuroscience, UCSF, San Francisco, CA 94158, USA
- Weill Institute for Neurosciences, UCSF, San Francisco, CA 94158, USA
- Department of Neurology, UCSF, San Francisco, CA 94158, USA
| | - Lara K Scaria
- Kavli Institute for Fundamental Neuroscience, UCSF, San Francisco, CA 94158, USA
- Weill Institute for Neurosciences, UCSF, San Francisco, CA 94158, USA
- Department of Neurology, UCSF, San Francisco, CA 94158, USA
| | - Sadhana Sridhar
- Kavli Institute for Fundamental Neuroscience, UCSF, San Francisco, CA 94158, USA
- Weill Institute for Neurosciences, UCSF, San Francisco, CA 94158, USA
- Department of Neurology, UCSF, San Francisco, CA 94158, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Alexandra B Nelson
- Neuroscience Graduate Program, UCSF, San Francisco, CA 94158, USA
- Kavli Institute for Fundamental Neuroscience, UCSF, San Francisco, CA 94158, USA
- Weill Institute for Neurosciences, UCSF, San Francisco, CA 94158, USA
- Department of Neurology, UCSF, San Francisco, CA 94158, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| |
Collapse
|
6
|
Cepeda C, Holley SM, Barry J, Oikonomou KD, Yazon VW, Peng A, Argueta D, Levine MS. Corticostriatal Maldevelopment in the R6/2 Mouse Model of Juvenile Huntington's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.15.618500. [PMID: 39464124 PMCID: PMC11507867 DOI: 10.1101/2024.10.15.618500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
There is a growing consensus that brain development in Huntington's disease (HD) is abnormal, leading to the idea that HD is not only a neurodegenerative but also a neurodevelopmental disorder. Indeed, structural and functional abnormalities have been observed during brain development in both humans and animal models of HD. However, a concurrent study of cortical and striatal development in a genetic model of HD is still lacking. Here we report significant alterations of corticostriatal development in the R6/2 mouse model of juvenile HD. We examined wildtype (WT) and R6/2 mice at postnatal (P) days 7, 14, and 21. Morphological examination demonstrated early structural and cellular alterations reminiscent of malformations of cortical development, and ex vivo electrophysiological recordings of cortical pyramidal neurons (CPNs) demonstrated significant age- and genotype-dependent changes of intrinsic membrane and synaptic properties. In general, R6/2 CPNs had reduced cell membrane capacitance and increased input resistance (P7 and P14), along with reduced frequency of spontaneous excitatory and inhibitory synaptic events during early development (P7), suggesting delayed cortical maturation. This was confirmed by increased occurrence of GABA A receptor-mediated giant depolarizing potentials at P7. At P14, the rheobase of CPNs was significantly reduced, along with increased excitability. Altered membrane and synaptic properties of R6/2 CPNs recovered progressively, and by P21 they were similar to WT CPNs. In striatal medium-sized spiny neurons (MSNs), a different picture emerged. Intrinsic membrane properties were relatively normal throughout development, except for a transient increase in membrane capacitance at P14. The first alterations in MSNs synaptic activity were observed at P14 and consisted of significant deficits in GABAergic inputs, however, these also were normalized by P21. In contrast, excitatory inputs began to decrease at this age. We conclude that the developing HD brain is capable of compensating for early developmental abnormalities and that cortical alterations precede and are a main contributor of striatal changes. Addressing cortical maldevelopment could help prevent or delay disease manifestations.
Collapse
|
7
|
Sampathkumar V, Koster KP, Carroll BJ, Sherman SM, Kasthuri N. Synaptic integration of somatosensory and motor cortical inputs onto spiny projection neurons of mice caudoputamen. Eur J Neurosci 2024; 60:6107-6122. [PMID: 39315531 PMCID: PMC11483202 DOI: 10.1111/ejn.16538] [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: 03/29/2024] [Accepted: 08/29/2024] [Indexed: 09/25/2024]
Abstract
The basal ganglia play pivotal roles in motor control and cognitive functioning. These nuclei are embedded in an anatomical loop: cortex to basal ganglia to thalamus back to cortex. We focus here on an essential synapse for descending control, from cortical layer 5 (L5) onto the GABAergic spiny projection neurons (SPNs) of the caudoputamen (CP). We employed genetic labeling to distinguish L5 neurons from somatosensory (S1) and motor (M1) cortices in large volume serial electron microscopy and electrophysiology datasets to better detail these inputs. First, M1 and S1 synapses showed a strong preference to innervate the spines of SPNs and rarely contacted aspiny cells, which are likely to be interneurons. Second, L5 inputs commonly converge from both areas onto single SPNs. Third, compared to unlabeled terminals in CP, those labeled from M1 and S1 show ultrastructural hallmarks of strong driver synapses: They innervate larger spines that were more likely to contain a spine apparatus, more often had embedded mitochondria, and more often contacted multiple targets. Finally, these inputs also demonstrated driver-like functional properties: SPNs responded to optogenetic activation from S1 and M1 with large EPSP/Cs that depressed and were dependent on ionotropic but not metabotropic receptors. Together, our findings suggest that individual SPNs integrate driver input from multiple cortical areas with implications for how the basal ganglia relay cortical input to provide inhibitory innervation of motor thalamus.
Collapse
Affiliation(s)
- Vandana Sampathkumar
- Department of Neurobiology, University of Chicago, Chicago, IL 60637
- Argonne National Laboratory
| | - Kevin P Koster
- Department of Neurobiology, University of Chicago, Chicago, IL 60637
| | - Briana J Carroll
- Department of Neurobiology, University of Chicago, Chicago, IL 60637
| | - S Murray Sherman
- Department of Neurobiology, University of Chicago, Chicago, IL 60637
| | - Narayanan Kasthuri
- Department of Neurobiology, University of Chicago, Chicago, IL 60637
- Argonne National Laboratory
| |
Collapse
|
8
|
Hunker AC, Wirthlin ME, Gill G, Johansen NJ, Hooper M, Omstead V, Taskin N, Weed N, Vargas S, Bendrick JL, Gore B, Ben-Simon Y, Bishaw Y, Opitz-Araya X, Martinez RA, Way S, Thyagarajan B, Lerma MN, Laird W, Sven O, Sanchez REA, Alexander JR, Amaya A, Amster A, Ayala A, Baker PM, Barcelli T, Barta S, Bertagnolli D, Bielstein C, Bishwakarma P, Bowlus J, Boyer G, Brouner K, Casian B, Casper T, Chakka AB, Chakrabarty R, Clark M, Colbert K, Daniel S, Dawe T, Departee M, DiValentin P, Donadio NP, Dotson NI, Dwivedi D, Egdorf T, Fliss T, Gary A, Goldy J, Grasso C, Groce EL, Gudsnuk K, Han W, Haradon Z, Hastings S, Helback O, Ho WV, Huang C, Johnson T, Jones DL, Juneau Z, Kenney J, Leibly M, Li S, Liang E, Loeffler H, Lusk NA, Madigan Z, Malloy J, Malone J, McCue R, Melchor J, Mich JK, Moosman S, Morin E, Naidoo R, Newman D, Ngo K, Nguyen K, Oster AL, Ouellette B, Oyama AA, Pena N, Pham T, Phillips E, Pom C, Potekhina L, Ransford S, Reding M, Rette DF, Reynoldson C, Rimorin C, Rios Sigler A, Rocha DB, Ronellenfitch K, Ruiz A, Sawyer L, Sevigny J, Shapovalova NV, Shepard N, Shulga L, Soliman S, Staats B, Taormina MJ, Tieu M, Wang Y, Wilkes J, Wood T, Zhou T, Williford A, Dee N, Mollenkopf T, Ng L, Esposito L, Kalmbach B, Yao S, Ariza J, Mufti S, Smith K, Waters J, Ersing I, Patrick M, Zeng H, Lein ES, Kojima Y, Horwitz G, Owen SF, Levi BP, Daigle TL, Tasic B, Bakken TE, Ting JT. Enhancer AAV toolbox for accessing and perturbing striatal cell types and circuits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.27.615553. [PMID: 39386678 PMCID: PMC11463465 DOI: 10.1101/2024.09.27.615553] [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: 10/12/2024]
Abstract
We present an enhancer AAV toolbox for accessing and perturbing striatal cell types and circuits. Best-in-class vectors were curated for accessing major striatal neuron populations including medium spiny neurons (MSNs), direct and indirect pathway MSNs, as well as Sst-Chodl, Pvalb-Pthlh, and cholinergic interneurons. Specificity was evaluated by multiple modes of molecular validation, three different routes of virus delivery, and with diverse transgene cargos. Importantly, we provide detailed information necessary to achieve reliable cell type specific labeling under different experimental contexts. We demonstrate direct pathway circuit-selective optogenetic perturbation of behavior and multiplex labeling of striatal interneuron types for targeted analysis of cellular features. Lastly, we show conserved in vivo activity for exemplary MSN enhancers in rat and macaque. This collection of striatal enhancer AAVs offers greater versatility compared to available transgenic lines and can readily be applied for cell type and circuit studies in diverse mammalian species beyond the mouse model.
Collapse
Affiliation(s)
| | | | - Gursajan Gill
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA
| | | | | | | | - Naz Taskin
- Allen Institute for Brain Science, Seattle, WA
| | | | - Sara Vargas
- Allen Institute for Brain Science, Seattle, WA
| | | | - Bryan Gore
- Allen Institute for Brain Science, Seattle, WA
| | | | - Yeme Bishaw
- Allen Institute for Brain Science, Seattle, WA
| | | | | | - Sharon Way
- Allen Institute for Brain Science, Seattle, WA
| | | | | | - Will Laird
- Allen Institute for Brain Science, Seattle, WA
| | - Otto Sven
- Allen Institute for Brain Science, Seattle, WA
| | | | | | - Avalon Amaya
- Allen Institute for Neural Dynamics, Seattle, WA
| | - Adam Amster
- Allen Institute for Brain Science, Seattle, WA
| | | | - Pam M Baker
- Allen Institute for Brain Science, Seattle, WA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Tim Dawe
- Allen Institute for Brain Science, Seattle, WA
| | | | | | | | | | | | - Tom Egdorf
- Allen Institute for Brain Science, Seattle, WA
| | - Tim Fliss
- Allen Institute for Brain Science, Seattle, WA
| | - Amanda Gary
- Allen Institute for Brain Science, Seattle, WA
| | - Jeff Goldy
- Allen Institute for Brain Science, Seattle, WA
| | - Conor Grasso
- Allen Institute for Neural Dynamics, Seattle, WA
| | | | | | - Warren Han
- Allen Institute for Neural Dynamics, Seattle, WA
| | - Zeb Haradon
- Allen Institute for Brain Science, Seattle, WA
| | | | | | - Windy V Ho
- Allen Institute for Brain Science, Seattle, WA
| | - Cindy Huang
- Allen Institute for Brain Science, Seattle, WA
| | - Tye Johnson
- Allen Institute for Neural Dynamics, Seattle, WA
| | | | - Zoe Juneau
- Allen Institute for Brain Science, Seattle, WA
| | | | | | - Su Li
- Allen Institute for Brain Science, Seattle, WA
| | | | | | | | | | | | | | | | | | - John K Mich
- Allen Institute for Brain Science, Seattle, WA
| | | | - Elyse Morin
- Allen Institute for Brain Science, Seattle, WA
| | - Robyn Naidoo
- Allen Institute for Neural Dynamics, Seattle, WA
| | | | - Kiet Ngo
- Allen Institute for Brain Science, Seattle, WA
| | | | | | | | | | - Nick Pena
- Allen Institute for Brain Science, Seattle, WA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Lane Sawyer
- Allen Institute for Brain Science, Seattle, WA
| | | | | | | | | | | | | | | | | | - Yimin Wang
- Allen Institute for Brain Science, Seattle, WA
| | - Josh Wilkes
- Allen Institute for Neural Dynamics, Seattle, WA
| | - Toren Wood
- Allen Institute for Brain Science, Seattle, WA
| | - Thomas Zhou
- Allen Institute for Brain Science, Seattle, WA
| | | | - Nick Dee
- Allen Institute for Brain Science, Seattle, WA
| | | | - Lydia Ng
- Allen Institute for Brain Science, Seattle, WA
| | | | - Brian Kalmbach
- Allen Institute for Brain Science, Seattle, WA
- Department of Neurobiology & Biophysics, University of Washington, Seattle, WA
| | - Shenqin Yao
- Allen Institute for Brain Science, Seattle, WA
| | | | | | | | - Jack Waters
- Allen Institute for Brain Science, Seattle, WA
| | | | | | | | - Ed S Lein
- Allen Institute for Brain Science, Seattle, WA
- Department of Neurological Surgery, University of Washington, Seattle, WA
| | - Yoshiko Kojima
- Department of Otolaryngology, Head and Neck Surgery, University of Washington, Seattle, WA
- Washington National Primate Research Center, Seattle, WA
| | - Greg Horwitz
- Department of Neurobiology & Biophysics, University of Washington, Seattle, WA
- Washington National Primate Research Center, Seattle, WA
| | - Scott F Owen
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA
| | - Boaz P Levi
- Allen Institute for Brain Science, Seattle, WA
| | - Tanya L Daigle
- Allen Institute for Brain Science, Seattle, WA
- Department of Neurobiology & Biophysics, University of Washington, Seattle, WA
| | | | | | - Jonathan T Ting
- Allen Institute for Brain Science, Seattle, WA
- Department of Neurobiology & Biophysics, University of Washington, Seattle, WA
- Washington National Primate Research Center, Seattle, WA
| |
Collapse
|
9
|
Gu SM, Hong E, Seo S, Kim S, Yoon SS, Cha HJ, Yun J. Different development patterns of reward behaviors induced by ketamine and JWH-018 in striatal GAD67 knockdown mice. J Vet Sci 2024; 25:e63. [PMID: 39231788 PMCID: PMC11450393 DOI: 10.4142/jvs.23325] [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: 12/28/2023] [Revised: 06/25/2024] [Accepted: 07/15/2024] [Indexed: 09/06/2024] Open
Abstract
IMPORTANCE Glutamic acid decarboxylase 67 (GAD67) is a gamma-aminobutyric acid (GABA) synthesis enzyme associated with the function of other neurotransmitter receptors, such as the N-methyl-D-aspartate (NMDA) receptor and cannabinoid receptor 1. However, the role of GAD67 in the development of different abused drug-induced reward behaviors remains unknown. In order to elucidate the mechanisms of substance use disorder, it is crucial to study changes in biomarkers within the brain's reward circuit induced by drug use. OBJECTIVE The study was designed to examine the effects of the downregulation of GAD67 expression in the dorsal striatum on reward behavior development. METHODS We evaluated the effects of GAD67 knockdown on depression-like behavior and anxiety using the forced swim test and elevated plus maze test in a mouse model. We further determined the effects of GAD67 knockdown on ketamine- and JWH-018-induced conditioned place preference (CPP). RESULTS Knockdown of GAD67 in the dorsal striatum of mice increased depression-like behavior, but it decreased anxiety. Moreover, the CPP score on the NMDA receptor antagonist ketamine was increased by GAD67 knockdown, whereas the administration of JWH-018, a cannabinoid receptor agonist, did not affect the CPP score in the GAD67 knockdown mice group compared with the control group. CONCLUSIONS AND RELEVANCE These results suggest that striatal GAD67 reduces GABAergic neuronal activity and may cause ketamine-induced NMDA receptor inhibition. Consequently, GAD67 downregulation induces vulnerability to the drug reward behavior of ketamine.
Collapse
Affiliation(s)
- Sun Mi Gu
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Korea
| | - Eunchong Hong
- Non-Clinical Center, Osong Medical Innovation Foundation, Cheongju 28160, Korea
| | - Sowoon Seo
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Korea
| | - Sanghyeon Kim
- Stanley Brain Research Laboratory, Stanley Medical Research Institute, Rockville, MD 20850, USA
| | - Seong Shoon Yoon
- College of Korean Medicine, Daegu Haany University, Daegu 42158, Korea
| | - Hye Jin Cha
- College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea.
| | - Jaesuk Yun
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Korea.
| |
Collapse
|
10
|
Yarotskyy V, Nass SR, Hahn YK, Contois L, McQuiston AR, Knapp PE, Hauser KF. Sustained fentanyl exposure inhibits neuronal activity in dissociated striatal neuronal-glial cocultures through actions independent of opioid receptors. J Neurophysiol 2024; 132:1056-1073. [PMID: 39110896 PMCID: PMC11427067 DOI: 10.1152/jn.00444.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 06/26/2024] [Accepted: 07/31/2024] [Indexed: 09/19/2024] Open
Abstract
Besides having high potency and efficacy at the µ-opioid (MOR) and other opioid receptor types, fentanyl has some affinity for some adrenergic receptor types, which may underlie its unique pathophysiological differences from typical opioids. To better understand the unique actions of fentanyl, we assessed the extent to which fentanyl alters striatal medium spiny neuron (MSN) activity via opioid receptors or α1-adrenoceptors in dopamine type 1 or type 2 receptor (D1 or D2)-expressing MSNs. In neuronal and mixed-glial cocultures from the striatum, acute fentanyl (100 nM) exposure decreased the frequency of spontaneous action potentials. Overnight exposure of cocultures to 100 nM fentanyl severely reduced the proportion of MSNs with spontaneous action potentials, which was unaffected by coexposure to the opioid receptor antagonist naloxone (10 µM) but fully negated by coadministering the pan-α1-adrenoceptor inverse agonist prazosin (100 nM) and partially reversed by the selective α1A-adrenoceptor antagonist RS 100329 (300 nM). Acute fentanyl (100 nM) exposure modestly reduced the frequency of action potentials and caused firing rate adaptations in D2, but not D1, MSNs. Prolonged (2-5 h) fentanyl (100 nM) application dramatically attenuated firing rates in both D1 and D2 MSNs. To identify possible cellular sites of α1-adrenoceptor action, α1-adrenoceptors were localized in subpopulations of striatal astroglia and neurons by immunocytochemistry and Adra1a mRNA by in situ hybridization in astrocytes. Thus, sustained fentanyl exposure can inhibit striatal MSN activity via a nonopioid receptor-dependent pathway, which may be modulated via complex actions in α1-adrenoceptor-expressing striatal neurons and/or glia.NEW & NOTEWORTHY Acute fentanyl exposure attenuated the activity of striatal medium spiny neurons (MSNs) in vitro and in dopamine D2, but not D1, receptor-expressing MSNs in ex vivo slices. By contrast, sustained fentanyl exposure suppressed the spontaneous activity of MSNs cocultured with glia through a nonopioid receptor-dependent mechanism modulated, in part, by α1-adrenoceptors. Fentanyl exposure can affect striatal function via a nonopioid receptor mechanism of action that appears mediated by α1-adrenoreceptor-expressing striatal neurons and/or astroglia.
Collapse
Affiliation(s)
- Viktor Yarotskyy
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Sara R Nass
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Yun-Kyung Hahn
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Liangru Contois
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States
| | - A Rory McQuiston
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Pamela E Knapp
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States
- Institute for Drug and Alcohol Studies, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Kurt F Hauser
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States
- Institute for Drug and Alcohol Studies, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States
| |
Collapse
|
11
|
Ryan MB, Girasole AE, Flores AJ, Twedell EL, McGregor MM, Brakaj R, Paletzki RF, Hnasko TS, Gerfen CR, Nelson AB. Excessive firing of dyskinesia-associated striatal direct pathway neurons is gated by dopamine and excitatory synaptic input. Cell Rep 2024; 43:114483. [PMID: 39024096 DOI: 10.1016/j.celrep.2024.114483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 04/19/2024] [Accepted: 06/25/2024] [Indexed: 07/20/2024] Open
Abstract
The striatum integrates dopaminergic and glutamatergic inputs to select preferred versus alternative actions. However, the precise mechanisms underlying this process remain unclear. One way to study action selection is to understand how it breaks down in pathological states. Here, we explored the cellular and synaptic mechanisms of levodopa-induced dyskinesia (LID), a complication of Parkinson's disease therapy characterized by involuntary movements. We used an activity-dependent tool (FosTRAP) in conjunction with a mouse model of LID to investigate functionally distinct subsets of striatal direct pathway medium spiny neurons (dMSNs). In vivo, levodopa differentially activates dyskinesia-associated (TRAPed) dMSNs compared to other dMSNs. We found this differential activation of TRAPed dMSNs is likely to be driven by higher dopamine receptor expression, dopamine-dependent excitability, and excitatory input from the motor cortex and thalamus. Together, these findings suggest how the intrinsic and synaptic properties of heterogeneous dMSN subpopulations integrate to support action selection.
Collapse
Affiliation(s)
- Michael B Ryan
- Neuroscience Graduate Program, UCSF, San Francisco, CA 94158, USA; Kavli Institute for Fundamental Neuroscience, UCSF, San Francisco, CA 94158, USA; Weill Institute for Neurosciences, UCSF, San Francisco, CA 94158, USA
| | - Allison E Girasole
- Neuroscience Graduate Program, UCSF, San Francisco, CA 94158, USA; Kavli Institute for Fundamental Neuroscience, UCSF, San Francisco, CA 94158, USA; Weill Institute for Neurosciences, UCSF, San Francisco, CA 94158, USA
| | - Andrew J Flores
- Department of Neurosciences, UCSD, La Jolla, CA 92093, USA; Veterans Affairs San Diego Healthcare System, San Diego, CA 92161, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Emily L Twedell
- Neuroscience Graduate Program, UCSF, San Francisco, CA 94158, USA; Kavli Institute for Fundamental Neuroscience, UCSF, San Francisco, CA 94158, USA; Weill Institute for Neurosciences, UCSF, San Francisco, CA 94158, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Matthew M McGregor
- Neuroscience Graduate Program, UCSF, San Francisco, CA 94158, USA; Kavli Institute for Fundamental Neuroscience, UCSF, San Francisco, CA 94158, USA; Weill Institute for Neurosciences, UCSF, San Francisco, CA 94158, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Rea Brakaj
- Department of Neurology, UCSF, San Francisco, CA 94158, USA
| | - Ronald F Paletzki
- Laboratory of Systems Neuroscience, National Institute of Mental Health, Bethesda, MD 20892, USA
| | - Thomas S Hnasko
- Department of Neurosciences, UCSD, La Jolla, CA 92093, USA; Veterans Affairs San Diego Healthcare System, San Diego, CA 92161, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Charles R Gerfen
- Laboratory of Systems Neuroscience, National Institute of Mental Health, Bethesda, MD 20892, USA
| | - Alexandra B Nelson
- Neuroscience Graduate Program, UCSF, San Francisco, CA 94158, USA; Kavli Institute for Fundamental Neuroscience, UCSF, San Francisco, CA 94158, USA; Weill Institute for Neurosciences, UCSF, San Francisco, CA 94158, USA; Department of Neurology, UCSF, San Francisco, CA 94158, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA.
| |
Collapse
|
12
|
Eckenwiler EA, Ingebretson AE, Stolley JJ, Fusaro MA, Romportl AM, Ross JM, Petersen CL, Kale EM, Clark MS, Schattauer SS, Zweifel LS, Lemos JC. Corticotropin-Releasing Factor Release From a Unique Subpopulation of Accumbal Neurons Constrains Action-Outcome Acquisition in Reward Learning. Biol Psychiatry 2024:S0006-3223(24)01534-8. [PMID: 39181385 DOI: 10.1016/j.biopsych.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 08/01/2024] [Accepted: 08/07/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND The nucleus accumbens (NAc) mediates reward learning and motivation. Despite an abundance of neuropeptides, peptidergic neurotransmission from the NAc has not been integrated into current models of reward learning. The existence of a sparse population of neurons containing corticotropin-releasing factor (CRF) has been previously documented. Here, we provide a comprehensive analysis of their identity and functional role in shaping reward learning. METHODS Our multidisciplinary approach included fluorescent in situ hybridization (n = ≥3 mice), tract tracing (n = 5 mice), ex vivo electrophysiology (n = ≥30 cells), in vivo calcium imaging with fiber photometry (n = ≥4 mice), and use of viral strategies in transgenic lines to selectively delete CRF peptide from NAc neurons (n = ≥4 mice). Behaviors used were instrumental learning, sucrose preference, and spontaneous exploration in an open field. RESULTS We showed that the vast majority of NAc CRF-containing neurons are spiny projection neurons (SPNs) comprising dopamine D1-, D2-, or D1/D2-containing SPNs that primarily project and connect to the ventral pallidum and to a lesser extent the ventral midbrain. As a population, they display mature and immature SPN firing properties. We demonstrated that NAc CRF-containing neurons track reward outcomes during operant reward learning and that CRF release from these neurons acts to constrain initial acquisition of action-outcome learning and at the same time facilitates flexibility in the face of changing contingencies. CONCLUSIONS CRF release from this sparse population of SPNs is critical for reward learning under normal conditions.
Collapse
Affiliation(s)
- Elizabeth A Eckenwiler
- Department of Neuroscience, University of Minnesota Twin Cities, Minneapolis, Minnesota; Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota
| | - Anna E Ingebretson
- Department of Neuroscience, University of Minnesota Twin Cities, Minneapolis, Minnesota; Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota
| | - Jeffrey J Stolley
- Department of Neuroscience, University of Minnesota Twin Cities, Minneapolis, Minnesota; Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota
| | - Maxine A Fusaro
- Department of Neuroscience, University of Minnesota Twin Cities, Minneapolis, Minnesota; Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota
| | - Alyssa M Romportl
- Department of Neuroscience, University of Minnesota Twin Cities, Minneapolis, Minnesota; Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota
| | - Jack M Ross
- Department of Neuroscience, University of Minnesota Twin Cities, Minneapolis, Minnesota; Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota
| | - Christopher L Petersen
- Department of Neuroscience, University of Minnesota Twin Cities, Minneapolis, Minnesota; Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota
| | - Eera M Kale
- Department of Neuroscience, University of Minnesota Twin Cities, Minneapolis, Minnesota; Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota
| | - Michael S Clark
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, Washington
| | - Selena S Schattauer
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, Washington; Department of Pharmacology, University of Washington, Seattle, Washington
| | - Larry S Zweifel
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, Washington; Department of Pharmacology, University of Washington, Seattle, Washington
| | - Julia C Lemos
- Department of Neuroscience, University of Minnesota Twin Cities, Minneapolis, Minnesota; Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
13
|
Xu Y, Lin Y, Yu M, Zhou K. The nucleus accumbens in reward and aversion processing: insights and implications. Front Behav Neurosci 2024; 18:1420028. [PMID: 39184934 PMCID: PMC11341389 DOI: 10.3389/fnbeh.2024.1420028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/26/2024] [Indexed: 08/27/2024] Open
Abstract
The nucleus accumbens (NAc), a central component of the brain's reward circuitry, has been implicated in a wide range of behaviors and emotional states. Emerging evidence, primarily drawing from recent rodent studies, suggests that the function of the NAc in reward and aversion processing is multifaceted. Prolonged stress or drug use induces maladaptive neuronal function in the NAc circuitry, which results in pathological conditions. This review aims to provide comprehensive and up-to-date insights on the role of the NAc in motivated behavior regulation and highlights areas that demand further in-depth analysis. It synthesizes the latest findings on how distinct NAc neuronal populations and pathways contribute to the processing of opposite valences. The review examines how a range of neuromodulators, especially monoamines, influence the NAc's control over various motivational states. Furthermore, it delves into the complex underlying mechanisms of psychiatric disorders such as addiction and depression and evaluates prospective interventions to restore NAc functionality.
Collapse
Affiliation(s)
| | | | | | - Kuikui Zhou
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China
| |
Collapse
|
14
|
Chuhma N, Rayport S. Regional heterogeneity in the membrane properties of mouse striatal neurons. Front Cell Neurosci 2024; 18:1412897. [PMID: 39144155 PMCID: PMC11321984 DOI: 10.3389/fncel.2024.1412897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 06/19/2024] [Indexed: 08/16/2024] Open
Abstract
The cytoarchitecture of the striatum is remarkably homogeneous, in contrast to the regional variation in striatal functions. Whether differences in the intrinsic membrane properties of striatal neurons contribute to regional heterogeneity has not been addressed systematically. We made recordings throughout the young adult mouse striatum under identical conditions, with synaptic input blocked, from four major striatal neuron types, namely, the two subtypes of spiny projection neurons (SPNs), cholinergic interneurons (ChIs), and fast-spiking GABAergic interneurons (FSIs), sampling at least 100 cells per cell type. Regional variation manifested across all cell types. All cell types in the nucleus accumbens (NAc) shell had higher input impedance and increased excitability. Cells in the NAc core were differentiated from the caudate-putamen (CPu) for both SPN subtypes by smaller action potentials and increased excitability. Similarity between the two SPN subtypes showed regional variation, differing more in the NAc than in the CPu. So, in the Str, both the intrinsic properties of interneurons and projection neurons are regionally heterogeneous, with the greatest difference between the NAc and CPu; greater excitability of NAc shell neurons may make the region more susceptible to activity-dependent plasticity.
Collapse
Affiliation(s)
- Nao Chuhma
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
- Department of Psychiatry, Columbia University, New York, NY, United States
| | - Stephen Rayport
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
- Department of Psychiatry, Columbia University, New York, NY, United States
| |
Collapse
|
15
|
Corrigan EK, DeBerardine M, Poddar A, Turrero García M, Schmitz MT, Harwell CC, Paredes MF, Krienen FM, Pollen AA. Conservation, alteration, and redistribution of mammalian striatal interneurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.29.605664. [PMID: 39131311 PMCID: PMC11312536 DOI: 10.1101/2024.07.29.605664] [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
Mammalian brains vary in size, structure, and function, but the extent to which evolutionarily novel cell types contribute to this variation remains unresolved1-4. Recent studies suggest there is a primate-specific population of striatal inhibitory interneurons, the TAC3 interneurons5. However, there has not yet been a detailed analysis of the spatial and phylogenetic distribution of this population. Here, we profile single cell gene expression in the developing pig (an ungulate) and ferret (a carnivore), representing 94 million years divergence from primates, and assign newborn inhibitory neurons to initial classes first specified during development6. We find that the initial class of TAC3 interneurons represents an ancestral striatal population that is also deployed towards the cortex in pig and ferret. In adult mouse, we uncover a rare population expressing Tac2, the ortholog of TAC3, in ventromedial striatum, prompting a reexamination of developing mouse striatal interneuron initial classes by targeted enrichment of their precursors. We conclude that the TAC3 interneuron initial class is conserved across Boreoeutherian mammals, with the mouse population representing Th striatal interneurons, a subset of which expresses Tac2. This study suggests that initial classes of telencephalic inhibitory neurons are largely conserved and that during evolution, neuronal types in the mammalian brain change through redistribution and fate refinement, rather than by derivation of novel precursors early in development.
Collapse
Affiliation(s)
- Emily K. Corrigan
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | | | - Aunoy Poddar
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Miguel Turrero García
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | | | - Corey C. Harwell
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub San Francisco, San Francisco, CA
| | - Mercedes F. Paredes
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Fenna M. Krienen
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Alex A. Pollen
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
16
|
Garma LD, Harder L, Barba-Reyes JM, Marco Salas S, Díez-Salguero M, Nilsson M, Serrano-Pozo A, Hyman BT, Muñoz-Manchado AB. Interneuron diversity in the human dorsal striatum. Nat Commun 2024; 15:6164. [PMID: 39039043 PMCID: PMC11263574 DOI: 10.1038/s41467-024-50414-w] [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: 05/16/2023] [Accepted: 07/01/2024] [Indexed: 07/24/2024] Open
Abstract
Deciphering the striatal interneuron diversity is key to understanding the basal ganglia circuit and to untangling the complex neurological and psychiatric diseases affecting this brain structure. We performed snRNA-seq and spatial transcriptomics of postmortem human caudate nucleus and putamen samples to elucidate the diversity and abundance of interneuron populations and their inherent transcriptional structure in the human dorsal striatum. We propose a comprehensive taxonomy of striatal interneurons with eight main classes and fourteen subclasses, providing their full transcriptomic identity and spatial expression profile as well as additional quantitative FISH validation for specific populations. We have also delineated the correspondence of our taxonomy with previous standardized classifications and shown the main transcriptomic and class abundance differences between caudate nucleus and putamen. Notably, based on key functional genes such as ion channels and synaptic receptors, we found matching known mouse interneuron populations for the most abundant populations, the recently described PTHLH and TAC3 interneurons. Finally, we were able to integrate other published datasets with ours, supporting the generalizability of this harmonized taxonomy.
Collapse
Affiliation(s)
- Leonardo D Garma
- Karolinska Institutet, Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Stockholm, Sweden
| | - Lisbeth Harder
- Karolinska Institutet, Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Stockholm, Sweden
| | - Juan M Barba-Reyes
- Departamento de Anatomía Patológica, Biología Celular, Histología, Historia de la Ciencia, Medicina Legal y Forense y Toxicología. Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA). University of Cádiz, Cádiz, Spain
| | - Sergio Marco Salas
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Mónica Díez-Salguero
- Departamento de Anatomía Patológica, Biología Celular, Histología, Historia de la Ciencia, Medicina Legal y Forense y Toxicología. Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA). University of Cádiz, Cádiz, Spain
| | - Mats Nilsson
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Alberto Serrano-Pozo
- Massachusetts General Hospital, Neurology Department, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Bradley T Hyman
- Massachusetts General Hospital, Neurology Department, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Ana B Muñoz-Manchado
- Karolinska Institutet, Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Stockholm, Sweden.
- Departamento de Anatomía Patológica, Biología Celular, Histología, Historia de la Ciencia, Medicina Legal y Forense y Toxicología. Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA). University of Cádiz, Cádiz, Spain.
| |
Collapse
|
17
|
Eckenwiler EA, Ingebretson AE, Stolley JJ, Fusaro MA, Romportl AM, Ross JM, Petersen CL, Kale EM, Clark MS, Schattauer SS, Zweifel LS, Lemos JC. CRF release from a unique subpopulation of accumbal neurons constrains action-outcome acquisition in reward learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.16.567495. [PMID: 39005420 PMCID: PMC11244858 DOI: 10.1101/2023.11.16.567495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Background The nucleus accumbens (NAc) mediates reward learning and motivation. Despite an abundance of neuropeptides, peptidergic neurotransmission from the NAc has not been integrated into current models of reward learning. The existence of a sparse population of neurons containing corticotropin releasing factor (CRF) has been previously documented. Here we provide a comprehensive analysis of their identity and functional role in shaping reward learning. Methods To do this, we took a multidisciplinary approach that included florescent in situ hybridization (N mice ≥ 3), tract tracing (N mice = 5), ex vivo electrophysiology (N cells ≥ 30), in vivo calcium imaging with fiber photometry (N mice ≥ 4) and use of viral strategies in transgenic lines to selectively delete CRF peptide from NAc neurons (N mice ≥ 4). Behaviors used were instrumental learning, sucrose preference and spontaneous exploration in an open field. Results Here we show that the vast majority of NAc CRF-containing (NAc CRF ) neurons are spiny projection neurons (SPNs) comprised of dopamine D1-, D2- or D1/D2-containing SPNs that primarily project and connect to the ventral pallidum and to a lesser extent the ventral midbrain. As a population, they display mature and immature SPN firing properties. We demonstrate that NAc CRF neurons track reward outcomes during operant reward learning and that CRF release from these neurons acts to constrain initial acquisition of action-outcome learning, and at the same time facilitates flexibility in the face of changing contingencies. Conclusion We conclude that CRF release from this sparse population of SPNs is critical for reward learning under normal conditions.
Collapse
|
18
|
Brill-Weil SG, Kramer PF, Yanez A, Clever FH, Zhang R, Khaliq ZM. Presynaptic GABA A receptors control integration of nicotinic input onto dopaminergic axons in the striatum. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.25.600616. [PMID: 39372741 PMCID: PMC11451734 DOI: 10.1101/2024.06.25.600616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Axons of dopaminergic neurons express gamma-aminobutyric acid type-A receptors (GABAARs) and nicotinic acetylcholine receptors (nAChRs) which are both independently positioned to shape striatal dopamine release. Using electrophysiology and calcium imaging, we investigated how interactions between GABAARs and nAChRs influence dopaminergic axon excitability. Direct axonal recordings showed that benzodiazepine application suppresses subthreshold axonal input from cholinergic interneurons (CINs). In imaging experiments, we used the first temporal derivative of presynaptic calcium signals to distinguish between direct- and nAChR-evoked activity in dopaminergic axons. We found that GABAAR antagonism with gabazine selectively enhanced nAChR-evoked axonal signals. Acetylcholine release was unchanged in gabazine suggesting that GABAARs located on dopaminergic axons, but not CINs, mediated this enhancement. Unexpectedly, we found that a widely used GABAAR antagonist, picrotoxin, inhibits axonal nAChRs and should be used cautiously for striatal circuit analysis. Overall, we demonstrate that GABAARs on dopaminergic axons regulate integration of nicotinic input to shape presynaptic excitability.
Collapse
Affiliation(s)
- Samuel G. Brill-Weil
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Paul F. Kramer
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Anthony Yanez
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Faye H. Clever
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Renshu Zhang
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Zayd M. Khaliq
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| |
Collapse
|
19
|
van Velthoven CTJ, Gao Y, Kunst M, Lee C, McMillen D, Chakka AB, Casper T, Clark M, Chakrabarty R, Daniel S, Dolbeare T, Ferrer R, Gloe J, Goldy J, Guzman J, Halterman C, Ho W, Huang M, James K, Nguy B, Pham T, Ronellenfitch K, Thomas ED, Torkelson A, Pagan CM, Kruse L, Dee N, Ng L, Waters J, Smith KA, Tasic B, Yao Z, Zeng H. The transcriptomic and spatial organization of telencephalic GABAergic neuronal types. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.18.599583. [PMID: 38948843 PMCID: PMC11212977 DOI: 10.1101/2024.06.18.599583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
The telencephalon of the mammalian brain comprises multiple regions and circuit pathways that play adaptive and integrative roles in a variety of brain functions. There is a wide array of GABAergic neurons in the telencephalon; they play a multitude of circuit functions, and dysfunction of these neurons has been implicated in diverse brain disorders. In this study, we conducted a systematic and in-depth analysis of the transcriptomic and spatial organization of GABAergic neuronal types in all regions of the mouse telencephalon and their developmental origins. This was accomplished by utilizing 611,423 single-cell transcriptomes from the comprehensive and high-resolution transcriptomic and spatial cell type atlas for the adult whole mouse brain we have generated, supplemented with an additional single-cell RNA-sequencing dataset containing 99,438 high-quality single-cell transcriptomes collected from the pre- and postnatal developing mouse brain. We present a hierarchically organized adult telencephalic GABAergic neuronal cell type taxonomy of 7 classes, 52 subclasses, 284 supertypes, and 1,051 clusters, as well as a corresponding developmental taxonomy of 450 clusters across different ages. Detailed charting efforts reveal extraordinary complexity where relationships among cell types reflect both spatial locations and developmental origins. Transcriptomically and developmentally related cell types can often be found in distant and diverse brain regions indicating that long-distance migration and dispersion is a common characteristic of nearly all classes of telencephalic GABAergic neurons. Additionally, we find various spatial dimensions of both discrete and continuous variations among related cell types that are correlated with gene expression gradients. Lastly, we find that cortical, striatal and some pallidal GABAergic neurons undergo extensive postnatal diversification, whereas septal and most pallidal GABAergic neuronal types emerge simultaneously during the embryonic stage with limited postnatal diversification. Overall, the telencephalic GABAergic cell type taxonomy can serve as a foundational reference for molecular, structural and functional studies of cell types and circuits by the entire community.
Collapse
Affiliation(s)
| | - Yuan Gao
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Changkyu Lee
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | | | - Scott Daniel
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Tim Dolbeare
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Jessica Gloe
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Jeff Goldy
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Windy Ho
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Mike Huang
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Beagan Nguy
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | | | - Lauren Kruse
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Nick Dee
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Lydia Ng
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Jack Waters
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Zizhen Yao
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA, USA
| |
Collapse
|
20
|
Iacino MC, Stowe TA, Pitts EG, Sexton LL, Macauley SL, Ferris MJ. A unique multi-synaptic mechanism involving acetylcholine and GABA regulates dopamine release in the nucleus accumbens through early adolescence in male rats. eLife 2024; 13:e62999. [PMID: 38860652 PMCID: PMC11281780 DOI: 10.7554/elife.62999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 06/10/2024] [Indexed: 06/12/2024] Open
Abstract
Adolescence is characterized by changes in reward-related behaviors, social behaviors, and decision-making. These behavioral changes are necessary for the transition into adulthood, but they also increase vulnerability to the development of a range of psychiatric disorders. Major reorganization of the dopamine system during adolescence is thought to underlie, in part, the associated behavioral changes and increased vulnerability. Here, we utilized fast scan cyclic voltammetry and microdialysis to examine differences in dopamine release as well as mechanisms that underlie differential dopamine signaling in the nucleus accumbens (NAc) core of adolescent (P28-35) and adult (P70-90) male rats. We show baseline differences between adult and adolescent-stimulated dopamine release in male rats, as well as opposite effects of the α6 nicotinic acetylcholine receptor (nAChR) on modulating dopamine release. The α6-selective blocker, α-conotoxin, increased dopamine release in early adolescent rats, but decreased dopamine release in rats beginning in middle adolescence and extending through adulthood. Strikingly, blockade of GABAA and GABAB receptors revealed that this α6-mediated increase in adolescent dopamine release requires NAc GABA signaling to occur. We confirm the role of α6 nAChRs and GABA in mediating this effect in vivo using microdialysis. Results herein suggest a multisynaptic mechanism potentially unique to the period of development that includes early adolescence, involving acetylcholine acting at α6-containing nAChRs to drive inhibitory GABA tone on dopamine release.
Collapse
Affiliation(s)
- Melody C Iacino
- Department of Translational Neuroscience, Wake Forest University School of MedicineWinston-SalemUnited States
| | - Taylor A Stowe
- Department of Translational Neuroscience, Wake Forest University School of MedicineWinston-SalemUnited States
| | - Elizabeth G Pitts
- Department of Translational Neuroscience, Wake Forest University School of MedicineWinston-SalemUnited States
| | - Lacey L Sexton
- Department of Translational Neuroscience, Wake Forest University School of MedicineWinston-SalemUnited States
| | - Shannon L Macauley
- Department of Translational Neuroscience, Wake Forest University School of MedicineWinston-SalemUnited States
- Department of Physiology, University of Kentucky College of MedicineLexingtonUnited States
| | - Mark J Ferris
- Department of Translational Neuroscience, Wake Forest University School of MedicineWinston-SalemUnited States
| |
Collapse
|
21
|
Oriol L, Chao M, Kollman GJ, Dowlat DS, Singhal SM, Steinkellner T, Hnasko TS. Ventral tegmental area interneurons revisited: GABA and glutamate projection neurons make local synapses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.07.597996. [PMID: 38895464 PMCID: PMC11185768 DOI: 10.1101/2024.06.07.597996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
The ventral tegmental area (VTA) contains projection neurons that release the neurotransmitters dopamine, GABA, and/or glutamate from distal synapses. VTA also contains GABA neurons that synapse locally on to VTA dopamine neurons, synapses widely credited to a population of so-called VTA interneurons. Interneurons in cortex, striatum, and elsewhere have well-defined morphological features, physiological properties, and molecular markers, but such features have not been clearly described in VTA. Indeed, there is scant evidence that local and distal synapses originate from separate populations of VTA GABA neurons. In this study we tested whether several markers expressed in non-dopamine VTA neurons are selective markers of interneurons, defined as neurons that synapse locally but not distally. Challenging previous assumptions, we found that VTA neurons genetically defined by expression of parvalbumin, somatostatin, neurotensin, or mu-opioid receptor project to known VTA targets including nucleus accumbens, ventral pallidum, lateral habenula, and prefrontal cortex. Moreover, we provide evidence that VTA GABA and glutamate projection neurons make functional inhibitory or excitatory synapses locally within VTA. These findings suggest that local collaterals of VTA projection neurons could mediate functions prior attributed to VTA interneurons. This study underscores the need for a refined understanding of VTA connectivity to explain how heterogeneous VTA circuits mediate diverse functions related to reward, motivation, or addiction.
Collapse
Affiliation(s)
- Lucie Oriol
- Department of Neurosciences, University of California, San Diego, La Jolla, United States
| | - Melody Chao
- Department of Neurosciences, University of California, San Diego, La Jolla, United States
| | - Grace J Kollman
- Department of Neurosciences, University of California, San Diego, La Jolla, United States
| | - Dina S Dowlat
- Department of Neurosciences, University of California, San Diego, La Jolla, United States
| | - Sarthak M Singhal
- Department of Neurosciences, University of California, San Diego, La Jolla, United States
| | - Thomas Steinkellner
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Austria
| | - Thomas S Hnasko
- Department of Neurosciences, University of California, San Diego, La Jolla, United States
- Research Service VA San Diego Healthcare System, San Diego, United States
| |
Collapse
|
22
|
Wegman E, Wosiski-Kuhn M, Luo Y. The dual role of striatal interneurons: circuit modulation and trophic support for the basal ganglia. Neural Regen Res 2024; 19:1277-1283. [PMID: 37905876 PMCID: PMC11467944 DOI: 10.4103/1673-5374.382987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 06/26/2023] [Accepted: 07/30/2023] [Indexed: 11/02/2023] Open
Abstract
ABSTRACT Striatal interneurons play a key role in modulating striatal-dependent behaviors, including motor activity and reward and emotional processing. Interneurons not only provide modulation to the basal ganglia circuitry under homeostasis but are also involved in changes to plasticity and adaptation during disease conditions such as Parkinson's or Huntington's disease. This review aims to summarize recent findings regarding the role of striatal cholinergic and GABAergic interneurons in providing circuit modulation to the basal ganglia in both homeostatic and disease conditions. In addition to direct circuit modulation, striatal interneurons have also been shown to provide trophic support to maintain neuron populations in adulthood. We discuss this interesting and novel role of striatal interneurons, with a focus on the maintenance of adult dopaminergic neurons from interneuron-derived sonic-hedgehog.
Collapse
Affiliation(s)
- Elliot Wegman
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH, USA
| | - Marlena Wosiski-Kuhn
- Department of Emergency Medicine at the School of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Yu Luo
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH, USA
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
23
|
Jiang T, Liang S, Zhang X, Dong S, Zhu H, Wang Y, Sun Y. Parvalbumin neurons in the nucleus accumbens shell modulate seizure in temporal lobe epilepsy. Neurobiol Dis 2024; 194:106482. [PMID: 38522590 DOI: 10.1016/j.nbd.2024.106482] [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: 12/10/2023] [Revised: 03/02/2024] [Accepted: 03/22/2024] [Indexed: 03/26/2024] Open
Abstract
A growing number of clinical and animal studies suggest that the nucleus accumbens (NAc), especially the shell, is involved in the pathogenesis of temporal lobe epilepsy (TLE). However, the role of parvalbumin (PV) GABAergic neurons in the NAc shell involved in TLE is still unclear. In this study, we induced a spontaneous TLE model by intrahippocampal administration of kainic acid (KA), which generally induce acute seizures in first 2 h (acute phase) and then lead to spontaneous recurrent seizures after two months (chronic phase). We found that chemogenetic activation of NAc shell PV neurons could alleviate TLE seizures by reducing the number and period of focal seizures (FSs) and secondary generalized seizures (sGSs), while selective inhibition of PV exacerbated seizure activity. Ruby-virus mapping results identified that the hippocampus (ventral and dorsal) is one of the projection targets of NAc shell PV neurons. Chemogenetic activation of the NAc-Hip PV projection fibers can mitigate seizures while inhibition has no effect on seizure ictogenesis. In summary, our findings reveal that PV neurons in the NAc shell could modulate the seizures in TLE via a long-range NAc-Hip circuit. All of these results enriched the investigation between NAc and epilepsy, offering new targets for future epileptogenesis research and precision therapy.
Collapse
Affiliation(s)
- Tong Jiang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| | - Shuyu Liang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| | - Xiaohan Zhang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| | - Shasha Dong
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| | - HaiFang Zhu
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| | - Ying Wang
- Institute of Neuropsychiatric Diseases, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266000, China.
| | - Yanping Sun
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| |
Collapse
|
24
|
Wadsworth HA, Warnecke AMP, Barlow JC, Robinson JK, Steimle E, Ronström JW, Williams PE, Galbraith CJ, Baldridge J, Jakowec MW, Davies DL, Yorgason JT. Ivermectin increases striatal cholinergic activity to facilitate dopamine terminal function. Cell Biosci 2024; 14:50. [PMID: 38632622 PMCID: PMC11025261 DOI: 10.1186/s13578-024-01228-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 04/01/2024] [Indexed: 04/19/2024] Open
Abstract
Ivermectin (IVM) is a commonly prescribed antiparasitic treatment with pharmacological effects on invertebrate glutamate ion channels resulting in paralysis and death of invertebrates. However, it can also act as a modulator of some vertebrate ion channels and has shown promise in facilitating L-DOPA treatment in preclinical models of Parkinson's disease. The pharmacological effects of IVM on dopamine terminal function were tested, focusing on the role of two of IVM's potential targets: purinergic P2X4 and nicotinic acetylcholine receptors. Ivermectin enhanced electrochemical detection of dorsal striatum dopamine release. Although striatal P2X4 receptors were observed, IVM effects on dopamine release were not blocked by P2X4 receptor inactivation. In contrast, IVM attenuated nicotine effects on dopamine release, and antagonizing nicotinic receptors prevented IVM effects on dopamine release. IVM also enhanced striatal cholinergic interneuron firing. L-DOPA enhances dopamine release by increasing vesicular content. L-DOPA and IVM co-application further enhanced release but resulted in a reduction in the ratio between high and low frequency stimulations, suggesting that IVM is enhancing release largely through changes in terminal excitability and not vesicular content. Thus, IVM is increasing striatal dopamine release through enhanced cholinergic activity on dopamine terminals.
Collapse
Affiliation(s)
- Hillary A Wadsworth
- Department of Cellular Biology and Physiology, and Neuroscience Program, Brigham Young University, 4005 LSB, Provo, UT, 84602, USA
| | - Alicia M P Warnecke
- Titus Family Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA, 90089, USA
| | - Joshua C Barlow
- Department of Cellular Biology and Physiology, and Neuroscience Program, Brigham Young University, 4005 LSB, Provo, UT, 84602, USA
| | - J Kayden Robinson
- Department of Cellular Biology and Physiology, and Neuroscience Program, Brigham Young University, 4005 LSB, Provo, UT, 84602, USA
| | - Emma Steimle
- Department of Cellular Biology and Physiology, and Neuroscience Program, Brigham Young University, 4005 LSB, Provo, UT, 84602, USA
| | - Joakim W Ronström
- Department of Cellular Biology and Physiology, and Neuroscience Program, Brigham Young University, 4005 LSB, Provo, UT, 84602, USA
| | - Pacen E Williams
- Department of Cellular Biology and Physiology, and Neuroscience Program, Brigham Young University, 4005 LSB, Provo, UT, 84602, USA
| | - Christopher J Galbraith
- Department of Cellular Biology and Physiology, and Neuroscience Program, Brigham Young University, 4005 LSB, Provo, UT, 84602, USA
| | - Jared Baldridge
- Department of Cellular Biology and Physiology, and Neuroscience Program, Brigham Young University, 4005 LSB, Provo, UT, 84602, USA
| | - Michael W Jakowec
- Titus Family Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA, 90089, USA
| | - Daryl L Davies
- Department of Neurology, Keck School of Medicine, University of Southern California, 1333 San Pablo Street, Los Angeles, CA, 90033, USA
| | - Jordan T Yorgason
- Department of Cellular Biology and Physiology, and Neuroscience Program, Brigham Young University, 4005 LSB, Provo, UT, 84602, USA.
| |
Collapse
|
25
|
Thabault M, Fernandes-Gomes C, Huot AL, Francheteau M, Balbous-Gautier A, Fernagut PO, Galvan L. Dysfunction of striatal parvalbumin interneurons drives motor stereotypies in Cntnap2-/- mouse model of autism spectrum disorders. PNAS NEXUS 2024; 3:pgae132. [PMID: 38617583 PMCID: PMC11010650 DOI: 10.1093/pnasnexus/pgae132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 03/18/2024] [Indexed: 04/16/2024]
Abstract
The involvement of parvalbumin (PV) interneurons in autism spectrum disorders (ASD) pathophysiology has been widely described without clearly elucidating how their dysfunctions could lead to ASD symptoms. The Cntnap2-/- mice, an ASD mouse model deficient for a major ASD susceptibility gene, display core ASD symptoms including motor stereotypies, which are directly linked to striatal dysfunction. This study reveals that striatal PV interneurons display hyperexcitability and hyperactivity in Cntnap2-/- mice, along with a reduced response in medium spiny neurons. We also provide evidence for a crucial role of striatal PV interneurons in motor stereotypies by demonstrating that their selective inhibition rescued a wild type-like phenotype. Our study identifies how PV interneuron dysfunctions disrupt striatal circuitry and drive the motor stereotypies in ASD.
Collapse
Affiliation(s)
- Mathieu Thabault
- Laboratoire de Neurosciences Expérimentales et Cliniques, Inserm U1084, Université de Poitiers, 86073, Poitiers, France
| | - Cloé Fernandes-Gomes
- Laboratoire de Neurosciences Expérimentales et Cliniques, Inserm U1084, Université de Poitiers, 86073, Poitiers, France
| | - Anne-Lise Huot
- Prébios Animal Facility, Université de Poitiers, 86073, Poitiers, France
| | - Maureen Francheteau
- Laboratoire de Neurosciences Expérimentales et Cliniques, Inserm U1084, Université de Poitiers, 86073, Poitiers, France
| | - Anaïs Balbous-Gautier
- Laboratoire de Neurosciences Expérimentales et Cliniques, Inserm U1084, Université de Poitiers, 86073, Poitiers, France
- Centre Hospitalier Universitaire, 86021, Poitiers, France
| | - Pierre-Olivier Fernagut
- Laboratoire de Neurosciences Expérimentales et Cliniques, Inserm U1084, Université de Poitiers, 86073, Poitiers, France
| | - Laurie Galvan
- Laboratoire de Neurosciences Expérimentales et Cliniques, Inserm U1084, Université de Poitiers, 86073, Poitiers, France
| |
Collapse
|
26
|
Ford CL, McDonough AA, Horie K, Young LJ. Melanocortin agonism in a social context selectively activates nucleus accumbens in an oxytocin-dependent manner. Neuropharmacology 2024; 247:109848. [PMID: 38253222 PMCID: PMC10923148 DOI: 10.1016/j.neuropharm.2024.109848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 10/18/2023] [Accepted: 01/13/2024] [Indexed: 01/24/2024]
Abstract
Social deficits are debilitating features of many psychiatric disorders, including autism. While time-intensive behavioral therapy is moderately effective, there are no pharmacological interventions for social deficits in autism. Many studies have attempted to treat social deficits using the neuropeptide oxytocin for its powerful neuromodulatory abilities and influence on social behaviors and cognition. However, clinical trials utilizing supplementation paradigms in which exogenous oxytocin is chronically administered independent of context have failed. An alternative treatment paradigm suggests pharmacologically activating the endogenous oxytocin system during behavioral therapy to enhance the efficacy of therapy by facilitating social learning. To this end, melanocortin receptor agonists like Melanotan II (MTII), which induces central oxytocin release and accelerates formation of partner preference, a form of social learning, in prairie voles, are promising pharmacological tools. To model pharmacological activation of the endogenous oxytocin system during behavioral therapy, we administered MTII prior to social interactions between male and female voles. We assessed its effect on oxytocin-dependent activity in brain regions subserving social learning using Fos expression as a proxy for neuronal activation. In non-social contexts, MTII only activated hypothalamic paraventricular nucleus, a primary site of oxytocin synthesis. However, during social interactions, MTII selectively increased oxytocin-dependent activation of nucleus accumbens, a site critical for social learning. These results suggest a mechanism for the MTII-induced acceleration of partner preference formation observed in previous studies. Moreover, they are consistent with the hypothesis that pharmacologically activating the endogenous oxytocin system with a melanocortin agonist during behavioral therapy has potential to facilitate social learning.
Collapse
Affiliation(s)
- Charles L Ford
- Center for Translational Social Neuroscience, Silvio O. Conte Center for Oxytocin and Social Cognition, Emory National Primate Research Center, Atlanta, GA, 30329, USA.
| | - Anna A McDonough
- Center for Translational Social Neuroscience, Silvio O. Conte Center for Oxytocin and Social Cognition, Emory National Primate Research Center, Atlanta, GA, 30329, USA
| | - Kengo Horie
- Center for Translational Social Neuroscience, Silvio O. Conte Center for Oxytocin and Social Cognition, Emory National Primate Research Center, Atlanta, GA, 30329, USA
| | - Larry J Young
- Center for Translational Social Neuroscience, Silvio O. Conte Center for Oxytocin and Social Cognition, Emory National Primate Research Center, Atlanta, GA, 30329, USA; Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| |
Collapse
|
27
|
Patel JC, Sherpa AD, Melani R, Witkovsky P, Wiseman MR, O'Neill B, Aoki C, Tritsch NX, Rice ME. GABA co-released from striatal dopamine axons dampens phasic dopamine release through autoregulatory GABA A receptors. Cell Rep 2024; 43:113834. [PMID: 38431842 PMCID: PMC11089423 DOI: 10.1016/j.celrep.2024.113834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/29/2023] [Accepted: 02/05/2024] [Indexed: 03/05/2024] Open
Abstract
Striatal dopamine axons co-release dopamine and gamma-aminobutyric acid (GABA), using GABA provided by uptake via GABA transporter-1 (GAT1). Functions of GABA co-release are poorly understood. We asked whether co-released GABA autoinhibits dopamine release via axonal GABA type A receptors (GABAARs), complementing established inhibition by dopamine acting at axonal D2 autoreceptors. We show that dopamine axons express α3-GABAAR subunits in mouse striatum. Enhanced dopamine release evoked by single-pulse optical stimulation in striatal slices with GABAAR antagonism confirms that an endogenous GABA tone limits dopamine release. Strikingly, an additional inhibitory component is seen when multiple pulses are used to mimic phasic axonal activity, revealing the role of GABAAR-mediated autoinhibition of dopamine release. This autoregulation is lost in conditional GAT1-knockout mice lacking GABA co-release. Given the faster kinetics of ionotropic GABAARs than G-protein-coupled D2 autoreceptors, our data reveal a mechanism whereby co-released GABA acts as a first responder to dampen phasic-to-tonic dopamine signaling.
Collapse
Affiliation(s)
- Jyoti C Patel
- Department of Neurosurgery, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA.
| | - Ang D Sherpa
- Department of Neurosurgery, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA; Center for Neural Science New York University, 4 Washington Place, New York, NY 10003, USA
| | - Riccardo Melani
- NYU Neuroscience Institute, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Paul Witkovsky
- Department of Neurosurgery, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Madeline R Wiseman
- Department of Neurosurgery, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Brian O'Neill
- Department of Neurosurgery, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Chiye Aoki
- NYU Neuroscience Institute, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA; Center for Neural Science New York University, 4 Washington Place, New York, NY 10003, USA
| | - Nicolas X Tritsch
- NYU Neuroscience Institute, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Margaret E Rice
- Department of Neurosurgery, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA; NYU Neuroscience Institute, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA.
| |
Collapse
|
28
|
Riley B, Gould E, Lloyd J, Hallum LE, Vlajkovic S, Todd K, Freestone PS. Dopamine transmission in the tail striatum: Regional variation and contribution of dopamine clearance mechanisms. J Neurochem 2024; 168:251-268. [PMID: 38308566 DOI: 10.1111/jnc.16052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 12/05/2023] [Accepted: 01/05/2024] [Indexed: 02/05/2024]
Abstract
The striatum can be divided into four anatomically and functionally distinct domains: the dorsolateral, dorsomedial, ventral and the more recently identified caudolateral (tail) striatum. Dopamine transmission in these striatal domains underlies many important behaviours, yet little is known about this phenomenon in the tail striatum. Furthermore, the tail is divided anatomically into four divisions (dorsal, medial, intermediate and lateral) based on the profile of D1 and D2 dopamine receptor-expressing medium spiny neurons, something that is not seen elsewhere in the striatum. Considering this organisation, how dopamine transmission occurs in the tail striatum is of great interest. We recorded evoked dopamine release in the four tail divisions, with comparison to the dorsolateral striatum, using fast-scan cyclic voltammetry in rat brain slices. Contributions of clearance mechanisms were investigated using dopamine transporter knockout (DAT-KO) rats, pharmacological transporter inhibitors and dextran. Evoked dopamine release in all tail divisions was smaller in amplitude than in the dorsolateral striatum and, importantly, regional variation was observed: dorsolateral ≈ lateral > medial > dorsal ≈ intermediate. Release amplitudes in the lateral division were 300% of that in the intermediate division, which also exhibited uniquely slow peak dopamine clearance velocity. Dopamine clearance in the intermediate division was most dependent on DAT, and no alternative dopamine transporters investigated (organic cation transporter-3, norepinephrine transporter and serotonin transporter) contributed significantly to dopamine clearance in any tail division. Our findings confirm that the tail striatum is not only a distinct dopamine domain but also that each tail division has unique dopamine transmission characteristics. This supports that the divisions are not only anatomically but also functionally distinct. How this segregation relates to the overall function of the tail striatum, particularly the processing of multisensory information, is yet to be determined.
Collapse
Affiliation(s)
- Bronwyn Riley
- Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Emily Gould
- Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Jordan Lloyd
- Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Luke E Hallum
- Department of Mechanical and Mechatronics Engineering, University of Auckland, Auckland, New Zealand
| | - Srdjan Vlajkovic
- Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Kathryn Todd
- Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Faculty of Physiology, Anatomy and Genetics, Oxford University, Oxford, UK
| | - Peter S Freestone
- Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
29
|
Plasil SL, Farris SP, Blednov Y, Mayfield RD, Mangieri RA, Nwokeji UJ, Aziz HC, Lambeth PS, Harris RA, Homanics GE. Mutation of novel ethanol-responsive lncRNA Gm41261 impacts ethanol-related behavioral responses in mice. GENES, BRAIN, AND BEHAVIOR 2024; 23:e12886. [PMID: 38373108 PMCID: PMC10876150 DOI: 10.1111/gbb.12886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/05/2024] [Accepted: 01/26/2024] [Indexed: 02/21/2024]
Abstract
Chronic alcohol exposure results in widespread dysregulation of gene expression that contributes to the pathogenesis of Alcohol Use Disorder (AUD). Long noncoding RNAs are key regulators of the transcriptome that we hypothesize coordinate alcohol-induced transcriptome dysregulation and contribute to AUD. Based on RNA-Sequencing data of human prefrontal cortex, basolateral amygdala and nucleus accumbens of AUD versus non-AUD brain, the human LINC01265 and its predicted murine homolog Gm41261 (i.e., TX2) were selected for functional interrogation. We tested the hypothesis that TX2 contributes to ethanol drinking and behavioral responses to ethanol. CRISPR/Cas9 mutagenesis was used to create a TX2 mutant mouse line in which 306 base-pairs were deleted from the locus. RNA analysis revealed that an abnormal TX2 transcript was produced at an unchanged level in mutant animals. Behaviorally, mutant mice had reduced ethanol, gaboxadol and zolpidem-induced loss of the righting response and reduced tolerance to ethanol in both sexes. In addition, a male-specific reduction in two-bottle choice every-other-day ethanol drinking was observed. Male TX2 mutants exhibited evidence of enhanced GABA release and altered GABAA receptor subunit composition in neurons of the nucleus accumbens shell. In C57BL6/J mice, TX2 within the cortex was cytoplasmic and largely present in Rbfox3+ neurons and IBA1+ microglia, but not in Olig2+ oligodendrocytes or in the majority of GFAP+ astrocytes. These data support the hypothesis that TX2 mutagenesis and dysregulation impacts ethanol drinking behavior and ethanol-induced behavioral responses in mice, likely through alterations in the GABAergic system.
Collapse
Affiliation(s)
- S. L. Plasil
- Department of Pharmacology and Chemical BiologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - S. P. Farris
- Department of Anesthesiology and Perioperative MedicineUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of Biomedical InformaticsUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- The Waggoner Center for Alcohol and Addiction ResearchThe University of Texas at AustinAustinTexasUSA
| | - Y. Blednov
- The Waggoner Center for Alcohol and Addiction ResearchThe University of Texas at AustinAustinTexasUSA
| | - R. D. Mayfield
- The Waggoner Center for Alcohol and Addiction ResearchThe University of Texas at AustinAustinTexasUSA
- Department of NeuroscienceThe University of Texas at AustinAustinTexasUSA
| | - R. A. Mangieri
- The Waggoner Center for Alcohol and Addiction ResearchThe University of Texas at AustinAustinTexasUSA
- Division of Pharmacology and Toxicology, College of PharmacyThe University of Texas at AustinAustinTexasUSA
| | - U. J. Nwokeji
- Department of Pharmacology and Chemical BiologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - H. C. Aziz
- The Waggoner Center for Alcohol and Addiction ResearchThe University of Texas at AustinAustinTexasUSA
- Division of Pharmacology and Toxicology, College of PharmacyThe University of Texas at AustinAustinTexasUSA
| | - P. S. Lambeth
- The Waggoner Center for Alcohol and Addiction ResearchThe University of Texas at AustinAustinTexasUSA
- Department of NeuroscienceThe University of Texas at AustinAustinTexasUSA
| | - R. A. Harris
- The Waggoner Center for Alcohol and Addiction ResearchThe University of Texas at AustinAustinTexasUSA
| | - G. E. Homanics
- Department of Pharmacology and Chemical BiologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of Anesthesiology and Perioperative MedicineUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of NeurobiologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| |
Collapse
|
30
|
Sanabria BD, Baskar SS, Yonk AJ, Linares-Garcia I, Abraira VE, Lee CR, Margolis DJ. Cell-Type Specific Connectivity of Whisker-Related Sensory and Motor Cortical Input to Dorsal Striatum. eNeuro 2024; 11:ENEURO.0503-23.2023. [PMID: 38164611 PMCID: PMC10849041 DOI: 10.1523/eneuro.0503-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 01/03/2024] Open
Abstract
The anterior dorsolateral striatum (DLS) is heavily innervated by convergent excitatory projections from the primary motor (M1) and sensory cortex (S1) and considered an important site of sensorimotor integration. M1 and S1 corticostriatal synapses have functional differences in their connection strength with striatal spiny projection neurons (SPNs) and fast-spiking interneurons (FSIs) in the DLS and, as a result, exert distinct influences on sensory-guided behaviors. In the present study, we tested whether M1 and S1 inputs exhibit differences in the subcellular anatomical distribution of striatal neurons. We injected adeno-associated viral vectors encoding spaghetti monster fluorescent proteins (sm.FPs) into M1 and S1 in male and female mice and used confocal microscopy to generate 3D reconstructions of corticostriatal inputs to single identified SPNs and FSIs obtained through ex vivo patch clamp electrophysiology. We found that M1 and S1 dually innervate SPNs and FSIs; however, there is a consistent bias towards the M1 input in SPNs that is not found in FSIs. In addition, M1 and S1 inputs were distributed similarly across the proximal, medial, and distal regions of SPN and FSI dendrites. Notably, closely localized M1 and S1 clusters of inputs were more prevalent in SPNs than FSIs, suggesting that cortical inputs are integrated through cell-type specific mechanisms. Our results suggest that the stronger functional connectivity from M1 to SPNs compared to S1, as previously observed, is due to a higher quantity of synaptic inputs. Our results have implications for how sensorimotor integration is performed in the striatum through cell-specific differences in corticostriatal connections.
Collapse
Affiliation(s)
- Branden D Sanabria
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway 08854, New Jersey
| | - Sindhuja S Baskar
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway 08854, New Jersey
| | - Alex J Yonk
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway 08854, New Jersey
| | - Iván Linares-Garcia
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway 08854, New Jersey
| | - Victoria E Abraira
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway 08854, New Jersey
| | - Christian R Lee
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway 08854, New Jersey
| | - David J Margolis
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway 08854, New Jersey
| |
Collapse
|
31
|
Fang LZ, Creed MC. Updating the striatal-pallidal wiring diagram. Nat Neurosci 2024; 27:15-27. [PMID: 38057614 DOI: 10.1038/s41593-023-01518-x] [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: 08/15/2021] [Accepted: 11/06/2023] [Indexed: 12/08/2023]
Abstract
The striatal and pallidal complexes are basal ganglia structures that orchestrate learning and execution of flexible behavior. Models of how the basal ganglia subserve these functions have evolved considerably, and the advent of optogenetic and molecular tools has shed light on the heterogeneity of subcircuits within these pathways. However, a synthesis of how molecularly diverse neurons integrate into existing models of basal ganglia function is lacking. Here, we provide an overview of the neurochemical and molecular diversity of striatal and pallidal neurons and synthesize recent circuit connectivity studies in rodents that takes this diversity into account. We also highlight anatomical organizational principles that distinguish the dorsal and ventral basal ganglia pathways in rodents. Future work integrating the molecular and anatomical properties of striatal and pallidal subpopulations may resolve controversies regarding basal ganglia network function.
Collapse
Affiliation(s)
- Lisa Z Fang
- Washington University Pain Center, Department of Anesthesiology, St. Louis, MO, USA
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, Newfoundland and Labrador, Canada
| | - Meaghan C Creed
- Washington University Pain Center, Department of Anesthesiology, St. Louis, MO, USA.
- Departments of Psychiatry, Neuroscience and Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
32
|
Day M, Belal M, Surmeier WC, Melendez A, Wokosin D, Tkatch T, Clarke VRJ, Surmeier DJ. GABAergic regulation of striatal spiny projection neurons depends upon their activity state. PLoS Biol 2024; 22:e3002483. [PMID: 38295323 PMCID: PMC10830145 DOI: 10.1371/journal.pbio.3002483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 12/26/2023] [Indexed: 02/02/2024] Open
Abstract
Synaptic transmission mediated by GABAA receptors (GABAARs) in adult, principal striatal spiny projection neurons (SPNs) can suppress ongoing spiking, but its effect on synaptic integration at subthreshold membrane potentials is less well characterized, particularly those near the resting down-state. To fill this gap, a combination of molecular, optogenetic, optical, and electrophysiological approaches were used to study SPNs in mouse ex vivo brain slices, and computational tools were used to model somatodendritic synaptic integration. In perforated patch recordings, activation of GABAARs, either by uncaging of GABA or by optogenetic stimulation of GABAergic synapses, evoked currents with a reversal potential near -60 mV in both juvenile and adult SPNs. Transcriptomic analysis and pharmacological work suggested that this relatively positive GABAAR reversal potential was not attributable to NKCC1 expression, but rather to HCO3- permeability. Regardless, from down-state potentials, optogenetic activation of dendritic GABAergic synapses depolarized SPNs. This GABAAR-mediated depolarization summed with trailing ionotropic glutamate receptor (iGluR) stimulation, promoting dendritic spikes and increasing somatic depolarization. Simulations revealed that a diffuse dendritic GABAergic input to SPNs effectively enhanced the response to dendritic iGluR signaling and promoted dendritic spikes. Taken together, our results demonstrate that GABAARs can work in concert with iGluRs to excite adult SPNs when they are in the resting down-state, suggesting that their inhibitory role is limited to brief periods near spike threshold. This state-dependence calls for a reformulation for the role of intrastriatal GABAergic circuits.
Collapse
Affiliation(s)
- Michelle Day
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Marziyeh Belal
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - William C. Surmeier
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Alexandria Melendez
- Department of Neurology, Baylor College of Medicine, Houston, Texas, United States of America
| | - David Wokosin
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Tatiana Tkatch
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, Maryland, United States of America
| | - Vernon R. J. Clarke
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, Maryland, United States of America
| | - D. James Surmeier
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, Maryland, United States of America
| |
Collapse
|
33
|
Holley SM, Reidling JC, Cepeda C, Wu J, Lim RG, Lau A, Moore C, Miramontes R, Fury B, Orellana I, Neel M, Coleal-Bergum D, Monuki ES, Bauer G, Meshul CK, Levine MS, Thompson LM. Transplanted human neural stem cells rescue phenotypes in zQ175 Huntington's disease mice and innervate the striatum. Mol Ther 2023; 31:3545-3563. [PMID: 37807512 PMCID: PMC10727970 DOI: 10.1016/j.ymthe.2023.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 08/28/2023] [Accepted: 10/04/2023] [Indexed: 10/10/2023] Open
Abstract
Huntington's disease (HD), a genetic neurodegenerative disorder, primarily affects the striatum and cortex with progressive loss of medium-sized spiny neurons (MSNs) and pyramidal neurons, disrupting cortico-striatal circuitry. A promising regenerative therapeutic strategy of transplanting human neural stem cells (hNSCs) is challenged by the need for long-term functional integration. We previously described that, with short-term hNSC transplantation into the striatum of HD R6/2 mice, human cells differentiated into electrophysiologically active immature neurons, improving behavior and biochemical deficits. Here, we show that long-term (8 months) implantation of hNSCs into the striatum of HD zQ175 mice ameliorates behavioral deficits, increases brain-derived neurotrophic factor (BDNF) levels, and reduces mutant huntingtin (mHTT) accumulation. Patch clamp recordings, immunohistochemistry, single-nucleus RNA sequencing (RNA-seq), and electron microscopy demonstrate that hNSCs differentiate into diverse neuronal populations, including MSN- and interneuron-like cells, and form connections. Single-nucleus RNA-seq analysis also shows restoration of several mHTT-mediated transcriptional changes of endogenous striatal HD mouse cells. Remarkably, engrafted cells receive synaptic inputs, innervate host neurons, and improve membrane and synaptic properties. Overall, the findings support hNSC transplantation for further evaluation and clinical development for HD.
Collapse
Affiliation(s)
- Sandra M Holley
- Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience & Human Behavior, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Jack C Reidling
- Institute for Memory Impairment and Neurological Disorders, University of California Irvine, Irvine, CA 92697, USA
| | - Carlos Cepeda
- Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience & Human Behavior, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Jie Wu
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Ryan G Lim
- Institute for Memory Impairment and Neurological Disorders, University of California Irvine, Irvine, CA 92697, USA
| | - Alice Lau
- Psychiatry & Human Behavior, University of California Irvine, Irvine, CA 92697, USA
| | - Cindy Moore
- Portland VA Medical Center, Portland, OR 97239, USA
| | - Ricardo Miramontes
- Institute for Memory Impairment and Neurological Disorders, University of California Irvine, Irvine, CA 92697, USA
| | - Brian Fury
- Institute for Regenerative Cures, University of California Davis, Sacramento, CA 95817, USA
| | - Iliana Orellana
- Institute for Memory Impairment and Neurological Disorders, University of California Irvine, Irvine, CA 92697, USA
| | - Michael Neel
- Department of Pathology & Laboratory Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Dane Coleal-Bergum
- Institute for Regenerative Cures, University of California Davis, Sacramento, CA 95817, USA
| | - Edwin S Monuki
- Department of Pathology & Laboratory Medicine, University of California, Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Center, University of California Irvine, Irvine, CA 92697, USA
| | - Gerhard Bauer
- Institute for Regenerative Cures, University of California Davis, Sacramento, CA 95817, USA
| | - Charles K Meshul
- Portland VA Medical Center, Portland, OR 97239, USA; Oregon Health & Science University, Department of Behavioral Neuroscience and Pathology, Portland, OR 97239, USA
| | - Michael S Levine
- Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience & Human Behavior, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA; Brain Research Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Leslie M Thompson
- Institute for Memory Impairment and Neurological Disorders, University of California Irvine, Irvine, CA 92697, USA; Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA; Psychiatry & Human Behavior, University of California Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Center, University of California Irvine, Irvine, CA 92697, USA; Department of Neurobiology & Behavior University of California Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
34
|
Zhai S, Cui Q, Simmons DV, Surmeier DJ. Distributed dopaminergic signaling in the basal ganglia and its relationship to motor disability in Parkinson's disease. Curr Opin Neurobiol 2023; 83:102798. [PMID: 37866012 PMCID: PMC10842063 DOI: 10.1016/j.conb.2023.102798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/19/2023] [Accepted: 09/20/2023] [Indexed: 10/24/2023]
Abstract
The degeneration of mesencephalic dopaminergic neurons that innervate the basal ganglia is responsible for the cardinal motor symptoms of Parkinson's disease (PD). It has been thought that loss of dopaminergic signaling in one basal ganglia region - the striatum - was solely responsible for the network pathophysiology causing PD motor symptoms. While our understanding of dopamine (DA)'s role in modulating striatal circuitry has deepened in recent years, it also has become clear that it acts in other regions of the basal ganglia to influence movement. Underscoring this point, examination of a new progressive mouse model of PD shows that striatal dopamine DA depletion alone is not sufficient to induce parkinsonism and that restoration of extra-striatal DA signaling attenuates parkinsonian motor deficits once they appear. This review summarizes recent advances in the effort to understand basal ganglia circuitry, its modulation by DA, and how its dysfunction drives PD motor symptoms.
Collapse
Affiliation(s)
- Shenyu Zhai
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Qiaoling Cui
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - DeNard V Simmons
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - D James Surmeier
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA.
| |
Collapse
|
35
|
Gómez-Ocádiz R, Silberberg G. Corticostriatal pathways for bilateral sensorimotor functions. Curr Opin Neurobiol 2023; 83:102781. [PMID: 37696188 DOI: 10.1016/j.conb.2023.102781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 08/15/2023] [Accepted: 08/16/2023] [Indexed: 09/13/2023]
Abstract
Corticostriatal pathways are essential for a multitude of motor, sensory, cognitive, and affective functions. They are mediated by cortical pyramidal neurons, roughly divided into two projection classes: the pyramidal tract (PT) and the intratelencephalic tract (IT). These pathways have been the focus of numerous studies in recent years, revealing their distinct structural and functional properties. Notably, their synaptic connectivity within ipsi- and contralateral cortical and striatal microcircuits is characterized by a high degree of target selectivity, providing a means to regulate the local neuromodulatory landscape in the striatum. Here, we discuss recent findings regarding the functional organization of the PT and IT corticostriatal pathways and its implications for bilateral sensorimotor functions.
Collapse
Affiliation(s)
- Ruy Gómez-Ocádiz
- Department of Neuroscience, Karolinska Institutet, Stockholm 17177, Sweden. https://twitter.com/@RuyGomezOcadiz
| | - Gilad Silberberg
- Department of Neuroscience, Karolinska Institutet, Stockholm 17177, Sweden.
| |
Collapse
|
36
|
Song C, Zhao Y, Zhang J, Dong Z, Kang X, Pan Y, Du J, Gao Y, Zhang H, Xi Y, Ding H, Kuang F, Wang W, Luo C, Zhang Z, Zhao Q, Yang J, Jiang W, Wu S, Gao F. Spatial Distribution of Parvalbumin-Positive Fibers in the Mouse Brain and Their Alterations in Mouse Models of Temporal Lobe Epilepsy and Parkinson's Disease. Neurosci Bull 2023; 39:1683-1702. [PMID: 37523099 PMCID: PMC10603013 DOI: 10.1007/s12264-023-01083-0] [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: 09/13/2022] [Accepted: 03/27/2023] [Indexed: 08/01/2023] Open
Abstract
Parvalbumin interneurons belong to the major types of GABAergic interneurons. Although the distribution and pathological alterations of parvalbumin interneuron somata have been widely studied, the distribution and vulnerability of the neurites and fibers extending from parvalbumin interneurons have not been detailly interrogated. Through the Cre recombinase-reporter system, we visualized parvalbumin-positive fibers and thoroughly investigated their spatial distribution in the mouse brain. We found that parvalbumin fibers are widely distributed in the brain with specific morphological characteristics in different regions, among which the cortex and thalamus exhibited the most intense parvalbumin signals. In regions such as the striatum and optic tract, even long-range thick parvalbumin projections were detected. Furthermore, in mouse models of temporal lobe epilepsy and Parkinson's disease, parvalbumin fibers suffered both massive and subtle morphological alterations. Our study provides an overview of parvalbumin fibers in the brain and emphasizes the potential pathological implications of parvalbumin fiber alterations.
Collapse
Affiliation(s)
- Changgeng Song
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yan Zhao
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Jiajia Zhang
- National Translational Science Center for Molecular Medicine, Department of Cell Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Ziyi Dong
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Xin Kang
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Yuqi Pan
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Jinle Du
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Yiting Gao
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Haifeng Zhang
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Ye Xi
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Hui Ding
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Fang Kuang
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Wenting Wang
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Ceng Luo
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Zhengping Zhang
- Department of Spinal Surgery, Honghui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, 710054, China
| | - Qinpeng Zhao
- Department of Spinal Surgery, Honghui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, 710054, China
| | - Jiazhou Yang
- The Medical College of Yan'an University, Yan'an, 716000, China
| | - Wen Jiang
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| | - Shengxi Wu
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| | - Fang Gao
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
37
|
Matityahu L, Gilin N, Sarpong GA, Atamna Y, Tiroshi L, Tritsch NX, Wickens JR, Goldberg JA. Acetylcholine waves and dopamine release in the striatum. Nat Commun 2023; 14:6852. [PMID: 37891198 PMCID: PMC10611775 DOI: 10.1038/s41467-023-42311-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
Striatal dopamine encodes reward, with recent work showing that dopamine release occurs in spatiotemporal waves. However, the mechanism of dopamine waves is unknown. Here we report that acetylcholine release in mouse striatum also exhibits wave activity, and that the spatial scale of striatal dopamine release is extended by nicotinic acetylcholine receptors. Based on these findings, and on our demonstration that single cholinergic interneurons can induce dopamine release, we hypothesized that the local reciprocal interaction between cholinergic interneurons and dopamine axons suffices to drive endogenous traveling waves. We show that the morphological and physiological properties of cholinergic interneuron - dopamine axon interactions can be modeled as a reaction-diffusion system that gives rise to traveling waves. Analytically-tractable versions of the model show that the structure and the nature of propagation of acetylcholine and dopamine traveling waves depend on their coupling, and that traveling waves can give rise to empirically observed correlations between these signals. Thus, our study provides evidence for striatal acetylcholine waves in vivo, and proposes a testable theoretical framework that predicts that the observed dopamine and acetylcholine waves are strongly coupled phenomena.
Collapse
Affiliation(s)
- Lior Matityahu
- Department of Medical Neurobiology, Institute of Medical Research Israel - Canada, The Faculty of Medicine, The Hebrew University of Jerusalem, 9112102, Jerusalem, Israel
| | - Naomi Gilin
- Department of Medical Neurobiology, Institute of Medical Research Israel - Canada, The Faculty of Medicine, The Hebrew University of Jerusalem, 9112102, Jerusalem, Israel
| | - Gideon A Sarpong
- Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Yara Atamna
- Department of Medical Neurobiology, Institute of Medical Research Israel - Canada, The Faculty of Medicine, The Hebrew University of Jerusalem, 9112102, Jerusalem, Israel
| | - Lior Tiroshi
- Department of Medical Neurobiology, Institute of Medical Research Israel - Canada, The Faculty of Medicine, The Hebrew University of Jerusalem, 9112102, Jerusalem, Israel
| | - Nicolas X Tritsch
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Jeffery R Wickens
- Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Joshua A Goldberg
- Department of Medical Neurobiology, Institute of Medical Research Israel - Canada, The Faculty of Medicine, The Hebrew University of Jerusalem, 9112102, Jerusalem, Israel.
| |
Collapse
|
38
|
Klavinskis-Whiting S, Bitzenhofer S, Hanganu-Opatz I, Ellender T. Generation and propagation of bursts of activity in the developing basal ganglia. Cereb Cortex 2023; 33:10595-10613. [PMID: 37615347 PMCID: PMC10560579 DOI: 10.1093/cercor/bhad307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/25/2023] Open
Abstract
The neonatal brain is characterized by intermittent bursts of oscillatory activity interspersed by relative silence. Although well-characterized for many cortical areas, to what extent these propagate and interact with subcortical brain areas is largely unknown. Here, early network activity was recorded from the developing basal ganglia, including motor/somatosensory cortex, dorsal striatum, and intralaminar thalamus, during the first postnatal weeks in mice. An unsupervised detection and classification method revealed two main classes of bursting activity, namely spindle bursts and nested gamma spindle bursts, characterized by oscillatory activity at ~ 10 and ~ 30 Hz frequencies, respectively. These were reliably identified across all three brain regions and exhibited region-specific differences in their structural, spectral, and developmental characteristics. Bursts of the same type often co-occurred in different brain regions and coherence and cross-correlation analyses reveal dynamic developmental changes in their interactions. The strongest interactions were seen for cortex and striatum, from the first postnatal week onwards, and cortex appeared to drive burst events in subcortical regions. Together, these results provide the first detailed description of early network activity within the developing basal ganglia and suggest that cortex is one of the main drivers of activity in downstream nuclei during this postnatal period.
Collapse
Affiliation(s)
| | - Sebastian Bitzenhofer
- Department of Biomedical Sciences, Institute of Developmental Neurophysiology, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Ileana Hanganu-Opatz
- Department of Biomedical Sciences, Institute of Developmental Neurophysiology, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Tommas Ellender
- Department of Pharmacology, University of Oxford, Mansfield Rd, Oxford, OX13QT, United Kingdom
- Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| |
Collapse
|
39
|
Franz D, Richter A, Köhling R. Electrophysiological insights into deep brain stimulation of the network disorder dystonia. Pflugers Arch 2023; 475:1133-1147. [PMID: 37530804 PMCID: PMC10499667 DOI: 10.1007/s00424-023-02845-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 06/02/2023] [Accepted: 07/24/2023] [Indexed: 08/03/2023]
Abstract
Deep brain stimulation (DBS), a treatment for modulating the abnormal central neuronal circuitry, has become the standard of care nowadays and is sometimes the only option to reduce symptoms of movement disorders such as dystonia. However, on the one hand, there are still open questions regarding the pathomechanisms of dystonia and, on the other hand, the mechanisms of DBS on neuronal circuitry. That lack of knowledge limits the therapeutic effect and makes it hard to predict the outcome of DBS for individual dystonia patients. Finding electrophysiological biomarkers seems to be a promising option to enable adapted individualised DBS treatment. However, biomarker search studies cannot be conducted on patients on a large scale and experimental approaches with animal models of dystonia are needed. In this review, physiological findings of deep brain stimulation studies in humans and animal models of dystonia are summarised and the current pathophysiological concepts of dystonia are discussed.
Collapse
Affiliation(s)
- Denise Franz
- Oscar Langendorff Institute of Physiology, University Medical Center Rostock, Rostock, Germany
| | - Angelika Richter
- Institute of Pharmacology, Pharmacy and Toxicology, University of Leipzig, Leipzig, Germany
| | - Rüdiger Köhling
- Oscar Langendorff Institute of Physiology, University Medical Center Rostock, Rostock, Germany.
| |
Collapse
|
40
|
Surmeier DJ, Zhai S, Cui Q, Simmons DV. Rethinking the network determinants of motor disability in Parkinson's disease. Front Synaptic Neurosci 2023; 15:1186484. [PMID: 37448451 PMCID: PMC10336242 DOI: 10.3389/fnsyn.2023.1186484] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 06/12/2023] [Indexed: 07/15/2023] Open
Abstract
For roughly the last 30 years, the notion that striatal dopamine (DA) depletion was the critical determinant of network pathophysiology underlying the motor symptoms of Parkinson's disease (PD) has dominated the field. While the basal ganglia circuit model underpinning this hypothesis has been of great heuristic value, the hypothesis itself has never been directly tested. Moreover, studies in the last couple of decades have made it clear that the network model underlying this hypothesis fails to incorporate key features of the basal ganglia, including the fact that DA acts throughout the basal ganglia, not just in the striatum. Underscoring this point, recent work using a progressive mouse model of PD has shown that striatal DA depletion alone is not sufficient to induce parkinsonism and that restoration of extra-striatal DA signaling attenuates parkinsonian motor deficits once they appear. Given the broad array of discoveries in the field, it is time for a new model of the network determinants of motor disability in PD.
Collapse
Affiliation(s)
- Dalton James Surmeier
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | | | | | | |
Collapse
|
41
|
Anderson AG, Kulkarni A, Konopka G. A single-cell trajectory atlas of striatal development. Sci Rep 2023; 13:9031. [PMID: 37270616 PMCID: PMC10239471 DOI: 10.1038/s41598-023-36255-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 05/31/2023] [Indexed: 06/05/2023] Open
Abstract
The striatum integrates dense neuromodulatory inputs from many brain regions to coordinate complex behaviors. This integration relies on the coordinated responses from distinct striatal cell types. While previous studies have characterized the cellular and molecular composition of the striatum using single-cell RNA-sequencing at distinct developmental timepoints, the molecular changes spanning embryonic through postnatal development at the single-cell level have not been examined. Here, we combine published mouse striatal single-cell datasets from both embryonic and postnatal timepoints to analyze the developmental trajectory patterns and transcription factor regulatory networks within striatal cell types. Using this integrated dataset, we found that dopamine receptor-1 expressing spiny projection neurons have an extended period of transcriptional dynamics and greater transcriptional complexity over postnatal development compared to dopamine receptor-2 expressing neurons. Moreover, we found the transcription factor, FOXP1, exerts indirect changes to oligodendrocytes. These data can be accessed and further analyzed through an interactive website ( https://mouse-striatal-dev.cells.ucsc.edu ).
Collapse
Affiliation(s)
- Ashley G Anderson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ashwinikumar Kulkarni
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, 75390-9111, USA
- Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Genevieve Konopka
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, 75390-9111, USA.
- Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
42
|
Giua G, Lassalle O, Makrini-Maleville L, Valjent E, Chavis P, Manzoni OJJ. Investigating cell-specific effects of FMRP deficiency on spiny projection neurons in a mouse model of Fragile X syndrome. Front Cell Neurosci 2023; 17:1146647. [PMID: 37323585 PMCID: PMC10264852 DOI: 10.3389/fncel.2023.1146647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 05/16/2023] [Indexed: 06/17/2023] Open
Abstract
Introduction Fragile X syndrome (FXS), resulting from a mutation in the Fmr1 gene, is the most common monogenic cause of autism and inherited intellectual disability. Fmr1 encodes the Fragile X Messenger Ribonucleoprotein (FMRP), and its absence leads to cognitive, emotional, and social deficits compatible with the nucleus accumbens (NAc) dysfunction. This structure is pivotal in social behavior control, consisting mainly of spiny projection neurons (SPNs), distinguished by dopamine D1 or D2 receptor expression, connectivity, and associated behavioral functions. This study aims to examine how FMRP absence differentially affects SPN cellular properties, which is crucial for categorizing FXS cellular endophenotypes. Methods We utilized a novel Fmr1-/y::Drd1a-tdTomato mouse model, which allows in-situ identification of SPN subtypes in FXS mice. Using RNA-sequencing, RNAScope and ex-vivo patch-clamp in adult male mice NAc, we comprehensively compared the intrinsic passive and active properties of SPN subtypes. Results Fmr1 transcripts and their gene product, FMRP, were found in both SPNs subtypes, indicating potential cell-specific functions for Fmr1. The study found that the distinguishing membrane properties and action potential kinetics typically separating D1- from D2-SPNs in wild-type mice were either reversed or abolished in Fmr1-/y::Drd1a-tdTomato mice. Interestingly, multivariate analysis highlighted the compound effects of Fmr1 ablation by disclosing how the phenotypic traits distinguishing each cell type in wild-type mice were altered in FXS. Discussion Our results suggest that the absence of FMRP disrupts the standard dichotomy characterizing NAc D1- and D2-SPNs, resulting in a homogenous phenotype. This shift in cellular properties could potentially underpin select aspects of the pathology observed in FXS. Therefore, understanding the nuanced effects of FMRP absence on SPN subtypes can offer valuable insights into the pathophysiology of FXS, opening avenues for potential therapeutic strategies.
Collapse
Affiliation(s)
- Gabriele Giua
- INMED, INSERM U1249, Marseille, France
- Aix-Marseille University, Marseille, France
- Cannalab “Cannabinoids Neuroscience Research International Associated Laboratory”, INSERM-Aix-Marseille University/Indiana University, Marseille, France
| | - Olivier Lassalle
- INMED, INSERM U1249, Marseille, France
- Aix-Marseille University, Marseille, France
- Cannalab “Cannabinoids Neuroscience Research International Associated Laboratory”, INSERM-Aix-Marseille University/Indiana University, Marseille, France
| | | | - Emmanuel Valjent
- IGF, University of Montpellier, INSERM, CNRS, Montpellier, France
| | - Pascale Chavis
- INMED, INSERM U1249, Marseille, France
- Aix-Marseille University, Marseille, France
- Cannalab “Cannabinoids Neuroscience Research International Associated Laboratory”, INSERM-Aix-Marseille University/Indiana University, Marseille, France
| | - Olivier J. J. Manzoni
- INMED, INSERM U1249, Marseille, France
- Aix-Marseille University, Marseille, France
- Cannalab “Cannabinoids Neuroscience Research International Associated Laboratory”, INSERM-Aix-Marseille University/Indiana University, Marseille, France
| |
Collapse
|
43
|
Garma L, Harder L, Barba-Reyes J, Diez-Salguero M, Serrano-Pozo A, Hyman B, Munoz-Manchado A. Interneuron diversity in the human dorsal striatum. RESEARCH SQUARE 2023:rs.3.rs-2921627. [PMID: 37292997 PMCID: PMC10246286 DOI: 10.21203/rs.3.rs-2921627/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Deciphering the striatal interneuron diversity is key to understanding the basal ganglia circuit and to untangle the complex neurological and psychiatric diseases affecting this brain structure. We performed snRNA-seq of postmortem human caudate nucleus and putamen samples to elucidate the diversity and abundance of interneuron populations and their transcriptional structure in the human dorsal striatum. We propose a new taxonomy of striatal interneurons with eight main classes and fourteen subclasses and provide their specific markers and some quantitative FISH validation, particularly for a novel PTHLH-expressing population. For the most abundant populations, PTHLH and TAC3, we found matching known mouse interneuron populations based on key functional genes such as ion channels and synaptic receptors. Remarkably, human TAC3 and mouse Th populations share important similarities including the expression of the neuropeptide tachykinin 3. Finally, we were able to integrate other published datasets supporting the generalizability of this new harmonized taxonomy.
Collapse
Affiliation(s)
| | | | | | | | | | - Bradley Hyman
- Massachusetts General Hospital, Harvard Medical School
| | | |
Collapse
|
44
|
Schulz A, Richter F, Richter A. In vivo optogenetic inhibition of striatal parvalbumin-reactive interneurons induced genotype-specific changes in neuronal activity without dystonic signs in male DYT1 knock-in mice. J Neurosci Res 2023; 101:448-463. [PMID: 36546658 DOI: 10.1002/jnr.25157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 10/30/2022] [Accepted: 12/10/2022] [Indexed: 12/24/2022]
Abstract
The pathophysiology of early-onset torsion dystonia (TOR1A/DYT1) remains unclear. Like 70% of human mutation carriers, rodent models with ΔGAG mutation such as DYT1 knock-in (KI) mice do not show overt dystonia but have subtle sensorimotor deficits and pattern of abnormal synaptic plasticity within the striatal microcircuits. There is evidence that dysfunction of striatal parvalbumin-reactive (Parv+) fast-spiking interneurons (FSIs) can be involved in dystonic signs. To elucidate the relevance of these GABAergic interneurons in the pathophysiology of DYT1 dystonia, we used in vivo optogenetics to specifically inhibit Parv+ and to detect changes in motor behavior and neuronal activity. Optogenetic fibers were bilaterally implanted into the dorsal striatum of male DYT1 KI mice and wild-type (WT) littermates expressing halorhodopsin (eNpHR3.0) in Parv+ interneurons. While stimulations with yellow light pulses for up to 60 min at different pulse durations and interval lengths did not induce abnormal movements, such as dystonic signs, immunohistochemical examinations revealed genotype-dependent differences. In contrast to WT mice, stimulated DYT1 KI showed decreased striatal neuronal activity, that is, less c-Fos reactive neurons, and increased activation of cholinergic interneurons after optogenetic inhibition of Parv+ interneurons. These findings suggest an involvement of Parv+ interneurons in an impaired striatal network in DYT1 KI mice, but at least short-term inhibition of these GABAergic interneurons is not sufficient to trigger a dystonic phenotype, similar to previously shown optogenetic activation of cholinergic interneurons.
Collapse
Affiliation(s)
- Anja Schulz
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| | - Franziska Richter
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, University of Leipzig, Leipzig, Germany.,Institute of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Angelika Richter
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| |
Collapse
|
45
|
Chuhma N, Oh SJ, Rayport S. The dopamine neuron synaptic map in the striatum. Cell Rep 2023; 42:112204. [PMID: 36867530 PMCID: PMC10657204 DOI: 10.1016/j.celrep.2023.112204] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 12/21/2022] [Accepted: 02/16/2023] [Indexed: 03/04/2023] Open
Abstract
Dopamine neurons project to the striatum to control movement, cognition, and motivation via slower volume transmission as well as faster dopamine, glutamate, and GABA synaptic actions capable of conveying the temporal information in dopamine neuron firing. To define the scope of these synaptic actions, recordings of dopamine-neuron-evoked synaptic currents were made in four major striatal neuron types, spanning the entire striatum. This revealed that inhibitory postsynaptic currents are widespread, while excitatory postsynaptic currents are localized to the medial nucleus accumbens and the anterolateral-dorsal striatum, and that all synaptic actions are weak in the posterior striatum. Synaptic actions in cholinergic interneurons are the strongest, variably mediating inhibition throughout the striatum and excitation in the medial accumbens, capable of controlling their activity. This mapping shows that dopamine neuron synaptic actions extend throughout the striatum, preferentially target cholinergic interneurons, and define distinct striatal subregions.
Collapse
Affiliation(s)
- Nao Chuhma
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA; Department of Psychiatry, Columbia University, New York, NY 10032, USA.
| | - Soo Jung Oh
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA; Department of Psychiatry, Columbia University, New York, NY 10032, USA
| | - Stephen Rayport
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA; Department of Psychiatry, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
46
|
Day M, Belal M, Surmeier WC, Melendez A, Wokosin D, Tkatch T, Clarke VRJ, Surmeier DJ. State-dependent GABAergic regulation of striatal spiny projection neuron excitability. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.14.532627. [PMID: 36993489 PMCID: PMC10055173 DOI: 10.1101/2023.03.14.532627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Synaptic transmission mediated by GABA A receptors (GABA A Rs) in adult, principal striatal spiny projection neurons (SPNs) can suppress ongoing spiking, but its effect on synaptic integration at sub-threshold membrane potentials is less well characterized, particularly those near the resting down-state. To fill this gap, a combination of molecular, optogenetic, optical and electrophysiological approaches were used to study SPNs in mouse ex vivo brain slices, and computational tools were used to model somatodendritic synaptic integration. Activation of GABA A Rs, either by uncaging of GABA or by optogenetic stimulation of GABAergic synapses, evoked currents with a reversal potential near -60 mV in perforated patch recordings from both juvenile and adult SPNs. Molecular profiling of SPNs suggested that this relatively positive reversal potential was not attributable to NKCC1 expression, but rather to a dynamic equilibrium between KCC2 and Cl-/HCO3-cotransporters. Regardless, from down-state potentials, optogenetic activation of dendritic GABAergic synapses depolarized SPNs. This GABAAR-mediated depolarization summed with trailing ionotropic glutamate receptor (iGluR) stimulation, promoting dendritic spikes and increasing somatic depolarization. Simulations revealed that a diffuse dendritic GABAergic input to SPNs effectively enhanced the response to coincident glutamatergic input. Taken together, our results demonstrate that GABA A Rs can work in concert with iGluRs to excite adult SPNs when they are in the resting down-state, suggesting that their inhibitory role is limited to brief periods near spike threshold. This state-dependence calls for a reformulation of the role intrastriatal GABAergic circuits.
Collapse
|
47
|
Jiang Y, Zou M, Wang Y, Wang Y. Nucleus accumbens in the pathogenesis of major depressive disorder: A brief review. Brain Res Bull 2023; 196:68-75. [PMID: 36889362 DOI: 10.1016/j.brainresbull.2023.03.004] [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: 11/09/2022] [Revised: 02/16/2023] [Accepted: 03/05/2023] [Indexed: 03/08/2023]
Abstract
Major depressive disorder (MDD) is the most prevalent mental disorder characterized by anhedonia, loss of motivation, avolition, behavioral despair and cognitive abnormalities. Despite substantial advancements in the pathophysiology of MDD in recent years, the pathogenesis of this disorder is not fully understood. Meanwhile,the treatment of MDD with currently available antidepressants is inadequate, highlighting the urgent need for clarifying the pathophysiology of MDD and developing novel therapeutics. Extensive studies have demonstrated the involvement of nuclei such as the prefrontal cortex (PFC), hippocampus (HIP), nucleus accumbens (NAc), hypothalamus, etc., in MDD. NAc,a region critical for reward and motivation,dysregulation of its activity seems to be a hallmark of this mood disorder. In this paper, we present a review of NAc related circuits, cellular and molecular mechanisms underlying MDD and share an analysis of the gaps in current research and possible future research directions.
Collapse
Affiliation(s)
- Yajie Jiang
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China; Hunan Key Laboratory of Traditional Chinese Medicine Prevention & Treatment of Depressive Diseases, Changsha, China
| | - Manshu Zou
- Hunan Key Laboratory of Traditional Chinese Medicine Prevention & Treatment of Depressive Diseases, Changsha, China
| | - Yeqing Wang
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Yuhong Wang
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China; Hunan Key Laboratory of Traditional Chinese Medicine Prevention & Treatment of Depressive Diseases, Changsha, China.
| |
Collapse
|
48
|
Sanabria BD, Baskar SS, Yonk AJ, Lee CR, Margolis DJ. Cell-Type Specific Connectivity of Whisker-Related Sensory and Motor Cortical Input to Dorsal Striatum. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.06.531405. [PMID: 36945420 PMCID: PMC10028946 DOI: 10.1101/2023.03.06.531405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
The anterior dorsolateral striatum (DLS) is heavily innervated by convergent excitatory projections from the primary motor (M1) and sensory cortex (S1) and is considered an important site of sensorimotor integration. M1 and S1 corticostriatal synapses have functional differences in the strength of their connections with striatal spiny projection neurons (SPNs) and fast-spiking interneurons (FSIs) in the DLS, and as a result exert an opposing influence on sensory-guided behaviors. In the present study, we tested whether M1 and S1 inputs exhibit differences in the subcellular anatomical distribution onto striatal neurons. We injected adeno-associated viral vectors encoding spaghetti monster fluorescent proteins (sm.FPs) into M1 and S1, and used confocal microscopy to generate 3D reconstructions of corticostriatal inputs to single identified SPNs and FSIs obtained through ex-vivo patch-clamp electrophysiology. We found that SPNs are less innervated by S1 compared to M1, but FSIs receive a similar number of inputs from both M1 and S1. In addition, M1 and S1 inputs were distributed similarly across the proximal, medial, and distal regions of SPNs and FSIs. Notably, clusters of inputs were prevalent in SPNs but not FSIs. Our results suggest that SPNs have stronger functional connectivity to M1 compared to S1 due to a higher density of synaptic inputs. The clustering of M1 and S1 inputs onto SPNs but not FSIs suggest that cortical inputs are integrated through cell-type specific mechanisms and more generally have implications for how sensorimotor integration is performed in the striatum. Significance Statement The dorsolateral striatum (DLS) is a key brain area involved in sensorimotor integration due to its dense innervation by the primary motor (M1) and sensory cortex (S1). However, the quantity and anatomical distribution of these inputs to the striatal cell population has not been well characterized. In this study we demonstrate that corticostriatal projections from M1 and S1 differentially innervate spiny projection neurons (SPNs) and fast-spiking interneurons (FSIs) in the DLS. S1 inputs innervate SPNs less than M1 and are likely to form synaptic clusters in SPNs but not in FSIs. These findings suggest that sensorimotor integration is partly achieved by differences in the synaptic organization of corticostriatal inputs to local striatal microcircuits.
Collapse
|
49
|
Sippy T, Tritsch NX. Unraveling the dynamics of dopamine release and its actions on target cells. Trends Neurosci 2023; 46:228-239. [PMID: 36635111 PMCID: PMC10204099 DOI: 10.1016/j.tins.2022.12.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/22/2022] [Accepted: 12/15/2022] [Indexed: 01/11/2023]
Abstract
The neuromodulator dopamine (DA) is essential for regulating learning, motivation, and movement. Despite its importance, however, the mechanisms by which DA influences the activity of target cells to alter behavior remain poorly understood. In this review, we describe recent methodological advances that are helping to overcome challenges that have historically hindered the field. We discuss how the employment of these methods is shedding light on the complex dynamics of extracellular DA in the brain, as well as how DA signaling alters the electrical, biochemical, and population activity of target neurons in vivo. These developments are generating novel hypotheses about the mechanisms through which DA release modifies behavior.
Collapse
Affiliation(s)
- Tanya Sippy
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA; Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA.
| | - Nicolas X Tritsch
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA; Fresco Institute for Parkinson's and Movement Disorders, New York University Langone Health, New York, NY, USA.
| |
Collapse
|
50
|
Xu J, Farsad HL, Hou Y, Barclay K, Lopez BA, Yamada S, Saliu IO, Shi Y, Knight WC, Bateman RJ, Benzinger TLS, Yi JJ, Li Q, Wang T, Perlmutter JS, Morris JC, Zhao G. Human striatal glia differentially contribute to AD- and PD-specific neurodegeneration. NATURE AGING 2023; 3:346-365. [PMID: 36993867 PMCID: PMC10046522 DOI: 10.1038/s43587-023-00363-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 01/09/2023] [Indexed: 02/11/2023]
Abstract
The commonalities and differences in cell-type-specific pathways that lead to Alzheimer disease (AD) and Parkinson disease (PD) remain unknown. Here, we performed a single-nucleus transcriptome comparison of control, AD and PD striata. We describe three astrocyte subpopulations shared across different brain regions and evolutionarily conserved between humans and mice. We reveal common features between AD and PD astrocytes and regional differences that contribute toward amyloid pathology and neurodegeneration. In contrast, we found that transcriptomic changes in microglia are largely unique to each disorder. Our analysis identified a population of activated microglia that shared molecular signatures with murine disease-associated microglia (DAM) as well as disease-associated and regional differences in microglia transcriptomic changes linking microglia to disease-specific amyloid pathology, tauopathy and neuronal death. Finally, we delineate undescribed subpopulations of medium spiny neurons (MSNs) in the striatum and provide neuronal transcriptomic profiles suggesting disease-specific changes and selective neuronal vulnerability.
Collapse
Affiliation(s)
- Jinbin Xu
- The Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Huifangjie L. Farsad
- The Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Yiran Hou
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
- Present address: Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA
| | - Kia Barclay
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, USA
| | - Ben Anthony Lopez
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, USA
- MD-PhD in Molecular Medicine Program, College of Medicine, University of the Philippines Manila, Manila, Philippines
| | - Shinnosuke Yamada
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Yiming Shi
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, USA
| | - William C. Knight
- The Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Randall J. Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie L. S. Benzinger
- The Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jason J. Yi
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, USA
| | - Qingyun Li
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, USA
| | - Ting Wang
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Joel S. Perlmutter
- The Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - John C. Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Guoyan Zhao
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|