1
|
Caulfield ME, Vander Werp MJ, Stancati JA, Collier TJ, Sortwell CE, Sandoval IM, Kordower JH, Manfredsson FP, Steece-Collier K. Advancing age and sex modulate antidyskinetic efficacy of striatal Ca V1.3 gene therapy in a rat model of Parkinson's disease. Neurobiol Aging 2025; 149:54-66. [PMID: 40010015 PMCID: PMC12007665 DOI: 10.1016/j.neurobiolaging.2025.02.003] [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: 09/05/2024] [Revised: 02/09/2025] [Accepted: 02/10/2025] [Indexed: 02/28/2025]
Abstract
We previously demonstrated that viral vector-mediated striatal CaV1.3 calcium channel downregulation in young adult (3mo) male parkinsonian rats provides uniform, robust protection against levodopa-induced dyskinesias (LID). Acknowledging the association of PD with aging and incidence in male and female sexes, we have expanded our studies to include rats of advancing age of both sexes. The current study directly contrasts age and sex, determining their impact on efficacy of intrastriatal AAV-CaV1.3-shRNA to prevent LID induction, removing the variable of levodopa-priming. Considering both sexes together, late-middle-aged ('aged'; 15mo) parkinsonian rats receiving AAV-CaV1.3-shRNA developed significantly less severe LID compared control AAV-scramble(SCR)-shRNA rats, however therapeutic benefit was significantly less robust than observed in young males. When considered separately, females showed significantly less therapeutic benefit than males. Furthermore, aged non-cycling/proestrous-negative female rats were refractory to LID induction, regardless of vector. This study provides novel insight into the impact of age and sex on the variable antidyskinetic responses of CaV1.3-targeted gene therapy, highlighting the importance of including clinically relevant age and sex populations in PD studies.
Collapse
Affiliation(s)
- Margaret E Caulfield
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, 400 Monroe Ave NW, Grand Rapids, MI 49503, USA
| | - Molly J Vander Werp
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, 400 Monroe Ave NW, Grand Rapids, MI 49503, USA
| | - Jennifer A Stancati
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, 400 Monroe Ave NW, Grand Rapids, MI 49503, USA
| | - Timothy J Collier
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, 400 Monroe Ave NW, Grand Rapids, MI 49503, USA
| | - Caryl E Sortwell
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, 400 Monroe Ave NW, Grand Rapids, MI 49503, USA
| | - Ivette M Sandoval
- Parkinson's Disease Research Unit, Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Jeffrey H Kordower
- ASU-Banner Neurodegenerative Disease Research Center (NDRC), College of Liberal Arts and Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Fredric P Manfredsson
- Parkinson's Disease Research Unit, Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Kathy Steece-Collier
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, 400 Monroe Ave NW, Grand Rapids, MI 49503, USA.
| |
Collapse
|
2
|
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. eLife 2025; 13:RP100085. [PMID: 40238649 PMCID: PMC12002793 DOI: 10.7554/elife.100085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025] Open
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 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 DiegoSan DiegoUnited States
| | - Melody Chao
- Department of Neurosciences, University of California, San DiegoSan DiegoUnited States
| | - Grace J Kollman
- Department of Neurosciences, University of California, San DiegoSan DiegoUnited States
| | - Dina S Dowlat
- Department of Neurosciences, University of California, San DiegoSan DiegoUnited States
| | - Sarthak M Singhal
- Department of Neurosciences, University of California, San DiegoSan DiegoUnited States
| | - Thomas Steinkellner
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of ViennaViennaAustria
| | - Thomas S Hnasko
- Department of Neurosciences, University of California, San DiegoSan DiegoUnited States
- Research Service VA San Diego Healthcare SystemSan DiegoUnited States
| |
Collapse
|
3
|
Zuo Y, Formoli N, Libster A, Sun D, Turner A, Iemolo A, Telese F. Single-Nucleus Transcriptomics Identifies Neuroblast Migration Programs Sensitive to Reelin and Cannabis in the Adolescent Nucleus Accumbens. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.03.646846. [PMID: 40236084 PMCID: PMC11996521 DOI: 10.1101/2025.04.03.646846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
The interplay between cannabis exposure during adolescence and genetic predisposition has been linked to increased vulnerability to psychiatric disorders. To investigate the molecular underpinnings of this interaction, we performed single-nucleus RNA sequencing of the nucleus accumbens (NAc) in a mouse model of Reln haploinsufficiency, a genetic risk factor for psychiatric disorders, following adolescent exposure to tetrahydrocannabinol (THC), the primary psychoactive component of cannabis. We identified a gene co-expression network influenced by both Reln genotype and THC, enriched in genes associated with human psychiatric disorders and predominantly expressed in a GABAergic neuroblast subpopulation. We showed that neuroblasts actively migrated in the adolescent NAc, but declined with age. Cell-to-cell communication analysis further revealed that these neuroblasts receive migratory cues from cholecystokinin interneurons, which express high levels of cannabinoid receptors. Together, these findings provide mechanistic insights into how adolescent THC exposure and genetic risk factors may impair GABAergic circuit maturation.
Collapse
|
4
|
Al Massadi O, Labarchède M, de Pins B, Longueville S, Giralt A, Irinopoulou T, Savariradjane M, Subashi E, Ginés S, Caboche J, Mariani LL, Betuing S, Girault JA. PYK2 in the dorsal striatum of Huntington's disease R6/2 mouse model. Neurobiol Dis 2025; 207:106840. [PMID: 39971200 DOI: 10.1016/j.nbd.2025.106840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 02/11/2025] [Indexed: 02/21/2025] Open
Abstract
Huntington's disease (HD) is a devastating disease due to autosomal dominant mutation in the HTT gene. Its pathophysiology involves multiple molecular alterations including transcriptional defects. We previously showed that in HD patients and mouse model, the protein levels of the non-receptor tyrosine kinase PYK2 were decreased in the hippocampus and that viral expression of PYK2 improved the hippocampal phenotype. Here, we investigated the possible contribution of PYK2 in the striatum, a brain region particularly altered in HD. PYK2 mRNA levels were decreased in the striatum and hippocampus of R6/2 mice, a severe HD model. Striatal PYK2 protein levels were also decreased in R6/2 mice and human patients. PYK2 knockout by itself did not result in motor symptoms observed in HD mouse models. We examined whether PYK2 deficiency participated in the R6/2 mice phenotype by expressing PYK2 in their dorsal striatum using AAV vectors. With an AAV1/Camk2a promoter, we did not observe significant improvement of body weight, clasping, motor activity and coordination (rotarod) alterations observed in R6/2 mice. With an AAV9/SYN1 promoter we found a slightly higher body weight and a trend to better rotarod performance. Both viruses similarly transduced striatal projection neurons and somatostatin-positive interneurons but only AAV9/SYN1 led to PYK2 expression in cholinergic and parvalbumin-positive interneurons. Expression of PYK2 in cholinergic interneurons may contribute to the slight effects observed. We conclude that PYK2 mRNA and protein levels are decreased in the striatum as in hippocampus of HD patients and mouse models. However, in contrast to hippocampus, striatal viral expression of PYK2 has only a minor effect on the R6/2 model striatal phenotype.
Collapse
Affiliation(s)
- Omar Al Massadi
- Inserm UMR-S 1270, Paris 75005, France; Sorbonne Université, Faculty of Sciences and Engineering, Paris 75005, France; Institut du Fer à Moulin, 17 rue du Fer à Moulin, Paris 75005, France.
| | - Mélody Labarchède
- Inserm UMR-S 1270, Paris 75005, France; Sorbonne Université, Faculty of Sciences and Engineering, Paris 75005, France; Institut du Fer à Moulin, 17 rue du Fer à Moulin, Paris 75005, France; Institut du Cerveau, Paris Brain Institute, ICM, Inserm, CNRS, Sorbonne Université, Paris, France
| | - Benoit de Pins
- Inserm UMR-S 1270, Paris 75005, France; Sorbonne Université, Faculty of Sciences and Engineering, Paris 75005, France; Institut du Fer à Moulin, 17 rue du Fer à Moulin, Paris 75005, France
| | - Sophie Longueville
- Inserm UMR-S 1270, Paris 75005, France; Sorbonne Université, Faculty of Sciences and Engineering, Paris 75005, France; Institut du Fer à Moulin, 17 rue du Fer à Moulin, Paris 75005, France
| | - Albert Giralt
- Inserm UMR-S 1270, Paris 75005, France; Sorbonne Université, Faculty of Sciences and Engineering, Paris 75005, France; Institut du Fer à Moulin, 17 rue du Fer à Moulin, Paris 75005, France; Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, 08036 Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; Production and Validation Center of Advanced Therapies (Creatio), University of Barcelona, Barcelona, Spain
| | - Theano Irinopoulou
- Inserm UMR-S 1270, Paris 75005, France; Sorbonne Université, Faculty of Sciences and Engineering, Paris 75005, France; Institut du Fer à Moulin, 17 rue du Fer à Moulin, Paris 75005, France
| | - Mythili Savariradjane
- Inserm UMR-S 1270, Paris 75005, France; Sorbonne Université, Faculty of Sciences and Engineering, Paris 75005, France; Institut du Fer à Moulin, 17 rue du Fer à Moulin, Paris 75005, France
| | - Enejda Subashi
- Sorbonne Université, Faculty of Sciences and Engineering, Paris 75005, France; Neuroscience Paris Seine, Institut de Biologie Paris-Seine, Paris, France; CNRS UMR8246, Paris, France; INSERM U1130, Paris, France
| | - Silvia Ginés
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, 08036 Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; Production and Validation Center of Advanced Therapies (Creatio), University of Barcelona, Barcelona, Spain
| | - Jocelyne Caboche
- Sorbonne Université, Faculty of Sciences and Engineering, Paris 75005, France; Neuroscience Paris Seine, Institut de Biologie Paris-Seine, Paris, France; CNRS UMR8246, Paris, France; INSERM U1130, Paris, France
| | - Louise-Laure Mariani
- Institut du Cerveau, Paris Brain Institute, ICM, Inserm, CNRS, Sorbonne Université, Paris, France
| | - Sandrine Betuing
- Sorbonne Université, Faculty of Sciences and Engineering, Paris 75005, France; Neuroscience Paris Seine, Institut de Biologie Paris-Seine, Paris, France; CNRS UMR8246, Paris, France; INSERM U1130, Paris, France
| | - Jean-Antoine Girault
- Inserm UMR-S 1270, Paris 75005, France; Sorbonne Université, Faculty of Sciences and Engineering, Paris 75005, France; Institut du Fer à Moulin, 17 rue du Fer à Moulin, Paris 75005, France; Institut du Cerveau, Paris Brain Institute, ICM, Inserm, CNRS, Sorbonne Université, Paris, France.
| |
Collapse
|
5
|
Apicella P, Martel AC, Marche K. Striatal function scrutinized through the PAN-TAN-FSI triumvirate. Front Cell Neurosci 2025; 19:1572657. [PMID: 40201383 PMCID: PMC11975669 DOI: 10.3389/fncel.2025.1572657] [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: 02/07/2025] [Accepted: 03/11/2025] [Indexed: 04/10/2025] Open
Abstract
Understanding the information encoded by distinct components of the neuronal circuitry in the striatum represents an avenue for elucidating the role of this subcortical region in adaptive behavior and its dysfunction in pathological conditions. In behaving animals, conventional single neuron recordings generally differentiated between three main electrophysiologically identified neuron subtypes in the striatum, referred to as phasically active neurons (PANs), tonically active neurons (TANs), and fast-spiking interneurons (FSIs), assumed to correspond to GABAergic spiny projection neurons, cholinergic interneurons, and parvalbumin-containing GABAergic interneurons, respectively. Considerable research has been devoted to exploring the behavior-related activities of neurons classified electrophysiologically into PANs, TANs, and FSIs in animals engaged in task performance, mostly monkeys. Although precise neuron identification remains a major challenge, such electrophysiological studies have provided insights into the functional properties of presumed distinct striatal neuronal populations. In this review, we will focus on current ideas about the functions subserved by these neuron subtypes, emphasizing their link to specific aspects of behaviors. We will also underline the issues that are yet to be resolved regarding the classification of striatal neurons into distinct subgroups which emphasize the importance of considering the potential overlap among electrophysiological characteristics and the molecular diversity of neuron types in the striatum.
Collapse
Affiliation(s)
- Paul Apicella
- Institut de Neurosciences de la Timone UMR 7289, Aix Marseille Université, Marseille, France
| | | | | |
Collapse
|
6
|
Hunker AC, Wirthlin ME, Gill G, Johansen NJ, Hooper M, Omstead V, Vargas S, Lerma MN, Taskin N, Weed N, Laird WD, Bishaw YM, Bendrick JL, Gore BB, Ben-Simon Y, Opitz-Araya X, Martinez RA, Way SW, Thyagarajan B, Otto S, Sanchez REA, Alexander JR, Amaya A, Amster A, Arbuckle J, 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, Sigler AR, Rocha DB, Ronellenfitch K, Ruiz A, Sawyer L, et alHunker AC, Wirthlin ME, Gill G, Johansen NJ, Hooper M, Omstead V, Vargas S, Lerma MN, Taskin N, Weed N, Laird WD, Bishaw YM, Bendrick JL, Gore BB, Ben-Simon Y, Opitz-Araya X, Martinez RA, Way SW, Thyagarajan B, Otto S, Sanchez REA, Alexander JR, Amaya A, Amster A, Arbuckle J, 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, Sigler AR, 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, Collman F, 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 2025:2024.09.27.615553. [PMID: 39386678 PMCID: PMC11463465 DOI: 10.1101/2024.09.27.615553] [Show More Authors] [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
| | | | | | | | - Sara Vargas
- Allen Institute for Brain Science, Seattle, WA
| | | | - Naz Taskin
- Allen Institute for Brain Science, Seattle, WA
| | | | | | | | | | | | | | | | | | | | | | - Sven Otto
- 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
|
7
|
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 2025; 97:637-650. [PMID: 39181385 PMCID: PMC11839403 DOI: 10.1016/j.biopsych.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 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
|
8
|
Bompierre S, Byelyayeva Y, Mota E, Lefevre M, Pumo A, Kehler J, Castro LRV, Vincent P. Cross-pathway integration of cAMP signals through cGMP and calcium-regulated phosphodiesterases in mouse striatal cholinergic interneurons. Br J Pharmacol 2025; 182:1236-1253. [PMID: 39604216 DOI: 10.1111/bph.17400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 09/04/2024] [Accepted: 10/21/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND AND PURPOSE Acetylcholine plays a key role in striatal function. Firing properties of striatal cholinergic interneurons depend on intracellular cAMP through the regulation of Ih currents. Yet, the dynamics of cyclic nucleotide signalling in these neurons have remained elusive. EXPERIMENTAL APPROACH We used highly selective FRET biosensors and pharmacological compounds to analyse the functional contribution of phosphodiesterases in striatal cholinergic interneurons in mouse brain slices. KEY RESULTS PDE1A, PDE3A and PDE4 appear as the main controllers of cAMP levels in striatal cholinergic interneurons. The calcium signal elicited through NMDA or metabotropic glutamate receptors activates PDE1A, which degrades both cAMP and cGMP. Interestingly, the nitric oxide/cGMP pathway amplifies cAMP signalling via PDE3A inhibition-a mechanism hitherto unexplored in a neuronal context. CONCLUSIONS AND IMPLICATIONS The expression pattern of specific PDE enzymes in striatal cholinergic interneurons, by integrating diverse intracellular pathways, can adjust cAMP responses bidirectionally. These properties eventually allow striatal cholinergic interneurons to dynamically regulate their overall activity and modulate acetylcholine release. Remarkably, this effect is the opposite of the cGMP-induced inhibition of cAMP signals involving PDE2A in striatal medium-sized spiny neurons, which provides important insights for the understanding of signal integration in the striatum.
Collapse
Affiliation(s)
- Ségolène Bompierre
- CNRS, Biological Adaptation and Ageing, Sorbonne Université, Paris, France
| | | | - Elia Mota
- CNRS, Biological Adaptation and Ageing, Sorbonne Université, Paris, France
| | - Marion Lefevre
- CNRS, Biological Adaptation and Ageing, Sorbonne Université, Paris, France
| | - Anna Pumo
- CNRS, Biological Adaptation and Ageing, Sorbonne Université, Paris, France
| | | | - Liliana R V Castro
- CNRS, Biological Adaptation and Ageing, Sorbonne Université, Paris, France
| | - Pierre Vincent
- CNRS, Biological Adaptation and Ageing, Sorbonne Université, Paris, France
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
| |
Collapse
|
9
|
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 2025:2024.06.07.597996. [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 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
|
10
|
Pérez IR, Avalos-Fuentes JA, Paz-Bermúdez F, Cortes H, Medina GT, Jijón-Lorenzo R, Florán B. D1 Receptor Functional Asymmetry at Striatonigral Neurons: A Neurochemical and Behavioral Study in Male Wistar Rats. J Neurosci Res 2025; 103:e70014. [PMID: 39737598 DOI: 10.1002/jnr.70014] [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: 09/11/2024] [Revised: 11/27/2024] [Accepted: 12/16/2024] [Indexed: 01/01/2025]
Abstract
Lateralization of motor behavior, a common phenomenon in humans and several species, is modulated by the basal ganglia, a site pointed out for the interhemispheric differences related to lateralization. Our study aims to shed light on the potential role of the striatonigral D1 receptor in functional asymmetry in normal conditions through neurochemical and behavioral means. We found that D1 receptor activation and D1/D3 receptor coactivation in striatonigral neurons leads to more cAMP production by adenylyl cyclase in the striatum and GABA release in their terminals in the right hemisphere compared to the left. These differences are linked to a higher receptor sensitivity and potentially a better coupling of Golf proteins. When we assessed motor behavior through intranigral injection of the D1 receptor agonist SKF 38393 in the left or right substantia nigra, we found higher contralateral circling when injected on the right side. Thus, differences in motor activity correlate with neurochemical data, indicating that D1 receptor signaling plays a significant role in motor asymmetry.
Collapse
Affiliation(s)
- Ihosvany Rodríguez Pérez
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de Mexico, Mexico
| | - José Arturo Avalos-Fuentes
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de Mexico, Mexico
| | - Francisco Paz-Bermúdez
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de Mexico, Mexico
| | - Hernan Cortes
- Instituto Nacional de Rehabilitación "Luís Guillermo Ibarra", Ciudad de Mexico, Mexico
| | - Gisela Tovar Medina
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de Mexico, Mexico
| | - Rafael Jijón-Lorenzo
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de Mexico, Mexico
| | - Benjamín Florán
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de Mexico, Mexico
| |
Collapse
|
11
|
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 PMCID: PMC11653103 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
|
12
|
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
|
13
|
Barbano MF, Qi J, Chen E, Mohammad U, Espinoza O, Candido M, Wang H, Liu B, Hahn S, Vautier F, Morales M. VTA glutamatergic projections to the nucleus accumbens suppress psychostimulant-seeking behavior. Neuropsychopharmacology 2024; 49:1905-1915. [PMID: 38926603 PMCID: PMC11473768 DOI: 10.1038/s41386-024-01905-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024]
Abstract
Converging evidence indicates that both dopamine and glutamate neurotransmission within the nucleus accumbens (NAc) play a role in psychostimulant self-administration and relapse in rodent models. Increased NAc dopamine release from ventral tegmental area (VTA) inputs is critical to psychostimulant self-administration and NAc glutamate release from prelimbic prefrontal cortex (PFC) inputs synapsing on medium spiny neurons (MSNs) is critical to reinstatement of psychostimulant-seeking after extinction. The regulation of the activity of MSNs by VTA dopamine inputs has been extensively studied, and recent findings have demonstrated that VTA glutamate neurons target the NAc medial shell. Here, we determined whether the mesoaccumbal glutamatergic pathway plays a role in psychostimulant conditioned place preference and self-administration in mice. We used optogenetics to induce NAc release of glutamate from VTA inputs during the acquisition, expression, and reinstatement phases of cocaine- or methamphetamine-induced conditioned place preference (CPP), and during priming-induced reinstatement of cocaine-seeking behavior. We found that NAc medial shell release of glutamate resulting from the activation of VTA glutamatergic fibers did not affect the acquisition of cocaine-induced CPP, but it blocked the expression, stress- and priming-induced reinstatement of cocaine- and methamphetamine CPP, as well as it blocked the priming-induced reinstatement of cocaine-seeking behavior after extinction. These findings indicate that in contrast to the well-recognized mesoaccumbal dopamine system that is critical to psychostimulant reward and relapse, there is a parallel mesoaccumbal glutamatergic system that suppresses reward and psychostimulant-seeking behavior.
Collapse
Affiliation(s)
- M Flavia Barbano
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Jia Qi
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Emma Chen
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Uzma Mohammad
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Orlando Espinoza
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Marcos Candido
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Huiling Wang
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Bing Liu
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Suyun Hahn
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - François Vautier
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Marisela Morales
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA.
| |
Collapse
|
14
|
Green N, Gao H, Chu X, Yuan Q, McGuire P, Lai D, Jiang G, Xuei X, Reiter JL, Stevens J, Sutherland GT, Goate AM, Pang ZP, Slesinger PA, Hart RP, Tischfield JA, Agrawal A, Wang Y, Duren Z, Edenberg HJ, Liu Y. Integrated Single-Cell Multiomic Profiling of Caudate Nucleus Suggests Key Mechanisms in Alcohol Use Disorder. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.02.606355. [PMID: 39149227 PMCID: PMC11326171 DOI: 10.1101/2024.08.02.606355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Alcohol use disorder (AUD) induces complex transcriptional and regulatory changes across multiple brain regions including the caudate nucleus, which remains understudied. Using paired single-nucleus RNA-seq and ATAC-seq on caudate samples from 143 human postmortem brains, including 74 with AUD, we identified 17 distinct cell types. We found that a significant portion of the alcohol-induced changes in gene expression occurred through altered chromatin accessibility. Notably, we identified novel transcriptional and chromatin accessibility differences in medium spiny neurons, impacting pathways such as RNA metabolism and immune response. A small cluster of D1/D2 hybrid neurons showed distinct differences, suggesting a unique role in AUD. Microglia exhibited distinct activation states deviating from classical M1/M2 designations, and astrocytes entered a reactive state partially regulated by JUND , affecting glutamatergic synapse pathways. Oligodendrocyte dysregulation, driven in part by OLIG2 , was linked to demyelination and increased TGF-β1 signaling from microglia and astrocytes. We also observed increased microglia-astrocyte communication via the IL-1β pathway. Leveraging our multiomic data, we performed cell type-specific expression quantitative trait loci analysis, integrating that with public genome-wide association studies to identify AUD risk genes such as ADAL and PPP2R3C , providing a direct link between genetic variants, chromatin accessibility, and gene expression in AUD. These findings not only provide new insights into the genetic and cellular mechanisms in the caudate related to AUD but also demonstrate the broader utility of large-scale multiomic studies in uncovering complex gene regulation across diverse cell types, which has implications beyond the substance use field.
Collapse
|
15
|
Roth RH, Ding JB. Cortico-basal ganglia plasticity in motor learning. Neuron 2024; 112:2486-2502. [PMID: 39002543 PMCID: PMC11309896 DOI: 10.1016/j.neuron.2024.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/29/2024] [Accepted: 06/17/2024] [Indexed: 07/15/2024]
Abstract
One key function of the brain is to control our body's movements, allowing us to interact with the world around us. Yet, many motor behaviors are not innate but require learning through repeated practice. Among the brain's motor regions, the cortico-basal ganglia circuit is particularly crucial for acquiring and executing motor skills, and neuronal activity in these regions is directly linked to movement parameters. Cell-type-specific adaptations of activity patterns and synaptic connectivity support the learning of new motor skills. Functionally, neuronal activity sequences become structured and associated with learned movements. On the synaptic level, specific connections become potentiated during learning through mechanisms such as long-term synaptic plasticity and dendritic spine dynamics, which are thought to mediate functional circuit plasticity. These synaptic and circuit adaptations within the cortico-basal ganglia circuitry are thus critical for motor skill acquisition, and disruptions in this plasticity can contribute to movement disorders.
Collapse
Affiliation(s)
- Richard H Roth
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA.
| | - Jun B Ding
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA; Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA; The Phil & Penny Knight Initiative for Brain Resilience at the Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
16
|
Spiliotis K, Köhling R, Just W, Starke J. Data-driven and equation-free methods for neurological disorders: analysis and control of the striatum network. FRONTIERS IN NETWORK PHYSIOLOGY 2024; 4:1399347. [PMID: 39171120 PMCID: PMC11335688 DOI: 10.3389/fnetp.2024.1399347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 07/16/2024] [Indexed: 08/23/2024]
Abstract
The striatum as part of the basal ganglia is central to both motor, and cognitive functions. Here, we propose a large-scale biophysical network for this part of the brain, using modified Hodgkin-Huxley dynamics to model neurons, and a connectivity informed by a detailed human atlas. The model shows different spatio-temporal activity patterns corresponding to lower (presumably normal) and increased cortico-striatal activation (as found in, e.g., obsessive-compulsive disorder), depending on the intensity of the cortical inputs. By applying equation-free methods, we are able to perform a macroscopic network analysis directly from microscale simulations. We identify the mean synaptic activity as the macroscopic variable of the system, which shows similarity with local field potentials. The equation-free approach results in a numerical bifurcation and stability analysis of the macroscopic dynamics of the striatal network. The different macroscopic states can be assigned to normal/healthy and pathological conditions, as known from neurological disorders. Finally, guided by the equation-free bifurcation analysis, we propose a therapeutic close loop control scheme for the striatal network.
Collapse
Affiliation(s)
- Konstantinos Spiliotis
- Institute of Mathematics, University of Rostock, Rostock, Germany
- Laboratory of Mathematics and Informatics (ISCE), Department of Civil Engineering, Democritus University of Thrace, Xanthi, Greece
| | - Rüdiger Köhling
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, Rostock, Germany
| | - Wolfram Just
- Institute of Mathematics, University of Rostock, Rostock, Germany
| | - Jens Starke
- Institute of Mathematics, University of Rostock, Rostock, Germany
| |
Collapse
|
17
|
Zhao Z, Stern SA. Homeostatic feeding in hedonic centres. Nat Metab 2024; 6:1433-1434. [PMID: 39147932 DOI: 10.1038/s42255-024-01089-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Affiliation(s)
- Zhe Zhao
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Sarah A Stern
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA.
| |
Collapse
|
18
|
Marinescu AM, Labouesse MA. The nucleus accumbens shell: a neural hub at the interface of homeostatic and hedonic feeding. Front Neurosci 2024; 18:1437210. [PMID: 39139500 PMCID: PMC11319282 DOI: 10.3389/fnins.2024.1437210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/16/2024] [Indexed: 08/15/2024] Open
Abstract
Feeding behavior is a complex physiological process regulated by the interplay between homeostatic and hedonic feeding circuits. Among the neural structures involved, the nucleus accumbens (NAc) has emerged as a pivotal region at the interface of these two circuits. The NAc comprises distinct subregions and in this review, we focus mainly on the NAc shell (NAcSh). Homeostatic feeding circuits, primarily found in the hypothalamus, ensure the organism's balance in energy and nutrient requirements. These circuits monitor peripheral signals, such as insulin, leptin, and ghrelin, and modulate satiety and hunger states. The NAcSh receives input from these homeostatic circuits, integrating information regarding the organism's metabolic needs. Conversely, so-called hedonic feeding circuits involve all other non-hunger and -satiety processes, i.e., the sensory information, associative learning, reward, motivation and pleasure associated with food consumption. The NAcSh is interconnected with hedonics-related structures like the ventral tegmental area and prefrontal cortex and plays a key role in encoding hedonic information related to palatable food seeking or consumption. In sum, the NAcSh acts as a crucial hub in feeding behavior, integrating signals from both homeostatic and hedonic circuits, to facilitate behavioral output via its downstream projections. Moreover, the NAcSh's involvement extends beyond simple integration, as it directly impacts actions related to food consumption. In this review, we first focus on delineating the inputs targeting the NAcSh; we then present NAcSh output projections to downstream structures. Finally we discuss how the NAcSh regulates feeding behavior and can be seen as a neural hub integrating homeostatic and hedonic feeding signals, via a functionally diverse set of projection neuron subpopulations.
Collapse
Affiliation(s)
- Alina-Măriuca Marinescu
- Brain, Wire and Behavior Group, Translational Nutritional Biology Laboratory, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Marie A. Labouesse
- Brain, Wire and Behavior Group, Translational Nutritional Biology Laboratory, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
19
|
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
|
20
|
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
|
21
|
Montaño-Rodriguez AR, Schorling T, Andressoo JO. Striatal GDNF Neurons Chemoattract RET-Positive Dopamine Axons at Seven Times Farther Distance Than Medium Spiny Neurons. Cells 2024; 13:1059. [PMID: 38920687 PMCID: PMC11202212 DOI: 10.3390/cells13121059] [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: 05/16/2024] [Revised: 06/08/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) is among the strongest dopamine neuron function- and survival-promoting factors known. Due to this reason, it has clinical relevance in dopamine disorders such as Parkinson's disease and schizophrenia. In the striatum, GDNF is exclusively expressed in interneurons, which make up only about 0.6% of striatal cells. Despite clinical significance, histological analysis of striatal GDNF system arborization and relevance to incoming dopamine axons, which bear its receptor RET, has remained enigmatic. This is mainly due to the lack of antibodies able to visualize GDNF- and RET-positive cellular processes; here, we overcome this problem by using knock-in marker alleles. We find that GDNF neurons chemoattract RET+ axons at least seven times farther in distance than medium spiny neurons (MSNs), which make up 95% of striatal neurons. Furthermore, we provide evidence that tyrosine hydroxylase, the rate-limiting enzyme in dopamine synthesis, is enriched towards GDNF neurons in the dopamine axons. Finally, we find that GDNF neuron arborizations occupy approximately only twelve times less striatal volume than 135 times more abundant MSNs. Collectively, our results improve our understanding of how endogenous GDNF affects striatal dopamine system function.
Collapse
Affiliation(s)
- Ana Rosa Montaño-Rodriguez
- Department of Pharmacology, Faculty of Medicine, Helsinki Institute of Life Science, University of Helsinki, 00290 Helsinki, Finland; (A.R.M.-R.); (T.S.)
| | - Tabea Schorling
- Department of Pharmacology, Faculty of Medicine, Helsinki Institute of Life Science, University of Helsinki, 00290 Helsinki, Finland; (A.R.M.-R.); (T.S.)
| | - Jaan-Olle Andressoo
- Department of Pharmacology, Faculty of Medicine, Helsinki Institute of Life Science, University of Helsinki, 00290 Helsinki, Finland; (A.R.M.-R.); (T.S.)
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, 17177 Stockholm, Sweden
| |
Collapse
|
22
|
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
|
23
|
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
|
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
|
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
|
26
|
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: 6] [Impact Index Per Article: 6.0] [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
|
27
|
DiFiglia M, Leavitt BR, Macdonald D, Thompson LM. Towards Standardizing Nomenclature in Huntington's Disease Research. J Huntingtons Dis 2024; 13:119-131. [PMID: 38968054 PMCID: PMC11307060 DOI: 10.3233/jhd-240044] [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] [Accepted: 06/10/2024] [Indexed: 07/07/2024]
Abstract
The field of Huntington's disease research covers many different scientific disciplines, from molecular biology all the way through to clinical practice, and as our understanding of the disease has progressed over the decades, a great deal of different terminology has accrued. The field is also renowned for its collaborative spirit and use of standardized reagents, assays, datasets, models, and clinical measures, so the use of standardized terms is especially important. We have set out to determine, through a consensus exercise involving basic and clinical scientists working in the field, the most appropriate language to use across disciplines. Nominally, this article will serve as the style guide for the Journal of Huntington's Disease (JHD), the only journal devoted exclusively to HD, and we lay out the preferred and standardized terminology and nomenclature for use in JHD publications. However, we hope that this article will also serve as a useful resource to the HD research community at large and that these recommended naming conventions will be adopted widely.
Collapse
Affiliation(s)
- Marian DiFiglia
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Harvard Medical School, Charlestown, MA, USA
| | - Blair R. Leavitt
- Center for Molecular Medicine and Therapeutics and Departments of Medical Genetics and Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Douglas Macdonald
- CHDI Management, Inc., The Company that Manages the Scientific Activities of CHDI Foundation, Inc, Los Angeles, CA, USA
| | - Leslie M. Thompson
- Departments of Psychiatry and Human Behavior and Neurobiology and Behavior, University of California, Irvine, CA, USA
| |
Collapse
|
28
|
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
|
29
|
Krienen FM, Levandowski KM, Zaniewski H, del Rosario RC, Schroeder ME, Goldman M, Wienisch M, Lutservitz A, Beja-Glasser VF, Chen C, Zhang Q, Chan KY, Li KX, Sharma J, McCormack D, Shin TW, Harrahill A, Nyase E, Mudhar G, Mauermann A, Wysoker A, Nemesh J, Kashin S, Vergara J, Chelini G, Dimidschstein J, Berretta S, Deverman BE, Boyden E, McCarroll SA, Feng G. A marmoset brain cell census reveals regional specialization of cellular identities. SCIENCE ADVANCES 2023; 9:eadk3986. [PMID: 37824615 PMCID: PMC10569717 DOI: 10.1126/sciadv.adk3986] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 09/26/2023] [Indexed: 10/14/2023]
Abstract
The mammalian brain is composed of many brain structures, each with its own ontogenetic and developmental history. We used single-nucleus RNA sequencing to sample over 2.4 million brain cells across 18 locations in the common marmoset, a New World monkey primed for genetic engineering, and examined gene expression patterns of cell types within and across brain structures. The adult transcriptomic identity of most neuronal types is shaped more by developmental origin than by neurotransmitter signaling repertoire. Quantitative mapping of GABAergic types with single-molecule FISH (smFISH) reveals that interneurons in the striatum and neocortex follow distinct spatial principles, and that lateral prefrontal and other higher-order cortical association areas are distinguished by high proportions of VIP+ neurons. We use cell type-specific enhancers to drive AAV-GFP and reconstruct the morphologies of molecularly resolved interneuron types in neocortex and striatum. Our analyses highlight how lineage, local context, and functional class contribute to the transcriptional identity and biodistribution of primate brain cell types.
Collapse
Affiliation(s)
- Fenna M. Krienen
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kirsten M. Levandowski
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Heather Zaniewski
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ricardo C.H. del Rosario
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Margaret E. Schroeder
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Melissa Goldman
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Martin Wienisch
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Alyssa Lutservitz
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Victoria F. Beja-Glasser
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Cindy Chen
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Qiangge Zhang
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ken Y. Chan
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Katelyn X. Li
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jitendra Sharma
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Dana McCormack
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Tay Won Shin
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Howard Hughes Medical Institute, Cambridge, MA 02139, USA
| | - Andrew Harrahill
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Eric Nyase
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Gagandeep Mudhar
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| | - Abigail Mauermann
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Howard Hughes Medical Institute, Cambridge, MA 02139, USA
| | - Alec Wysoker
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - James Nemesh
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Seva Kashin
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Josselyn Vergara
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Gabriele Chelini
- Center for Mind/Brain Sciences, University of Trento, Piazza della Manifattura n.1, Rovereto (TN) 38068, Italy
| | - Jordane Dimidschstein
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Sabina Berretta
- Basic Neuroscience Division, McLean Hospital, Belmont, MA 02478, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA
| | - Benjamin E. Deverman
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ed Boyden
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Howard Hughes Medical Institute, Cambridge, MA 02139, USA
| | - Steven A. McCarroll
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Guoping Feng
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| |
Collapse
|
30
|
Marshall P. Finding an Optimal Level of GDNF Overexpression: Insights from Dopamine Cycling. Cell Mol Neurobiol 2023; 43:3179-3189. [PMID: 37410316 PMCID: PMC10477250 DOI: 10.1007/s10571-023-01375-z] [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: 03/14/2023] [Accepted: 06/12/2023] [Indexed: 07/07/2023]
Abstract
The application of glial cell line-derive neurotrophic factor (GDNF) to cell cultures and animal models has demonstrated positive effects upon dopaminergic neuronal survival and development, function, restoration, and protection. On this basis, recombinant GDNF protein has been trialled in the treatment of late-stage human Parkinson's disease patients with only limited success that is likely due to a lack of viable receptor targets in an advanced state of neurodegeneration. The latest research points to more refined approaches of modulating GDNF signalling and an optimal quantity and spatial regulation of GDNF can be extrapolated using regulation of dopamine as a proxy measure. The basic research literature on dopaminergic effects of GDNF in animal models is reviewed, concluding that a twofold increase in natively expressing cells increases dopamine turnover and maximises neuroprotective and beneficial motor effects whilst minimising hyperdopaminergia and other side-effects. Methodological considerations for measurement of dopamine levels and neuroanatomical distinctions are made between populations of dopamine neurons and their respective effects upon movement and behaviour that will inform future research into this still-relevant growth factor.
Collapse
Affiliation(s)
- Pepin Marshall
- Neuroscience Center, University of Helsinki, 00014, Helsinki, Finland.
- Institute of Pharmacology, Toxicology and Pharmacy, Ludwig-Maximilians-University, Munich, Germany.
| |
Collapse
|
31
|
Alegre-Zurano L, García-Baos A, Castro-Zavala A, Medrano M, Gallego-Landin I, Valverde O. The FAAH inhibitor URB597 reduces cocaine intake during conditioned punishment and mitigates cocaine seeking during withdrawal. Biomed Pharmacother 2023; 165:115194. [PMID: 37499453 DOI: 10.1016/j.biopha.2023.115194] [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: 05/26/2023] [Revised: 06/30/2023] [Accepted: 07/18/2023] [Indexed: 07/29/2023] Open
Abstract
The endocannabinoid system is prominently implicated in the control of cocaine reinforcement due to its relevant role in synaptic plasticity and neurotransmitter modulation in the mesocorticolimbic system. The inhibition of fatty acid amide hydrolase (FAAH), and the resulting increase in anandamide and other N-acylethanolamines, represents a promising strategy for reducing drug seeking. In the present study, we aimed to assess the effects of the FAAH inhibitor URB597 (1 mg/kg) on crucial features of cocaine addictive-like behaviour in mice. Therefore, we tested the effects of URB597 on acquisition of cocaine (0.6 mg/kg/inf) self-administration, compulsive-like cocaine intake and cue-induced drug-seeking behaviour during withdrawal. URB597 reduced cocaine intake under conditioned punishment while having no impact on acquisition. This result was associated to increased cannabinoid receptor 1 gene expression in the ventral striatum and medium spiny neurons activation in the nucleus accumbens shell. Moreover, URB597 mitigated cue-induced drug-seeking behaviour during prolonged abstinence and prevented the withdrawal-induced increase in FAAH gene expression in the ventral striatum. In this case, URB597 decreased activation of medium spiny neurons in the nucleus accumbens core. Our findings evidence the prominent role of endocannabinoids in the development of cocaine addictive-like behaviours and support the potential of FAAH inhibition as a therapeutical target for the treatment of cocaine addiction.
Collapse
Affiliation(s)
- Laia Alegre-Zurano
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Alba García-Baos
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain; Neuroscience Research Program, IMIM-Hospital Del Mar Research Institute, Barcelona, Spain
| | - Adriana Castro-Zavala
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Mireia Medrano
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Ines Gallego-Landin
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Olga Valverde
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain; Neuroscience Research Program, IMIM-Hospital Del Mar Research Institute, Barcelona, Spain.
| |
Collapse
|
32
|
Gallegos DA, Minto M, Liu F, Hazlett MF, Aryana Yousefzadeh S, Bartelt LC, West AE. Cell-type specific transcriptional adaptations of nucleus accumbens interneurons to amphetamine. Mol Psychiatry 2023; 28:3414-3428. [PMID: 35173267 PMCID: PMC9378812 DOI: 10.1038/s41380-022-01466-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 01/13/2022] [Accepted: 01/26/2022] [Indexed: 11/09/2022]
Abstract
Parvalbumin-expressing (PV+) interneurons of the nucleus accumbens (NAc) play an essential role in the addictive-like behaviors induced by psychostimulant exposure. To identify molecular mechanisms of PV+ neuron plasticity, we isolated interneuron nuclei from the NAc of male and female mice following acute or repeated exposure to amphetamine (AMPH) and sequenced for cell type-specific RNA expression and chromatin accessibility. AMPH regulated the transcription of hundreds of genes in PV+ interneurons, and this program was largely distinct from that regulated in other NAc GABAergic neurons. Chromatin accessibility at enhancers predicted cell-type specific gene regulation, identifying transcriptional mechanisms of differential AMPH responses. Finally, we assessed expression of PV-enriched, AMPH-regulated genes in an Mecp2 mutant mouse strain that shows heightened behavioral sensitivity to psychostimulants to explore the functional importance of this transcriptional program. Together these data provide novel insight into the cell-type specific programs of transcriptional plasticity in NAc neurons that underlie addictive-like behaviors.
Collapse
Affiliation(s)
- David A Gallegos
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA
| | - Melyssa Minto
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA
| | - Fang Liu
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA
| | - Mariah F Hazlett
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA
| | | | - Luke C Bartelt
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA
| | - Anne E West
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
33
|
Petroccione MA, D'Brant LY, Affinnih N, Wehrle PH, Todd GC, Zahid S, Chesbro HE, Tschang IL, Scimemi A. Neuronal glutamate transporters control reciprocal inhibition and gain modulation in D1 medium spiny neurons. eLife 2023; 12:e81830. [PMID: 37435808 PMCID: PMC10411972 DOI: 10.7554/elife.81830] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 07/09/2023] [Indexed: 07/13/2023] Open
Abstract
Understanding the function of glutamate transporters has broad implications for explaining how neurons integrate information and relay it through complex neuronal circuits. Most of what is currently known about glutamate transporters, specifically their ability to maintain glutamate homeostasis and limit glutamate diffusion away from the synaptic cleft, is based on studies of glial glutamate transporters. By contrast, little is known about the functional implications of neuronal glutamate transporters. The neuronal glutamate transporter EAAC1 is widely expressed throughout the brain, particularly in the striatum, the primary input nucleus of the basal ganglia, a region implicated with movement execution and reward. Here, we show that EAAC1 limits synaptic excitation onto a population of striatal medium spiny neurons identified for their expression of D1 dopamine receptors (D1-MSNs). In these cells, EAAC1 also contributes to strengthen lateral inhibition from other D1-MSNs. Together, these effects contribute to reduce the gain of the input-output relationship and increase the offset at increasing levels of synaptic inhibition in D1-MSNs. By reducing the sensitivity and dynamic range of action potential firing in D1-MSNs, EAAC1 limits the propensity of mice to rapidly switch between behaviors associated with different reward probabilities. Together, these findings shed light on some important molecular and cellular mechanisms implicated with behavior flexibility in mice.
Collapse
Affiliation(s)
| | | | | | | | | | - Shergil Zahid
- SUNY Albany, Department of BiologyAlbanyUnited States
| | | | - Ian L Tschang
- SUNY Albany, Department of BiologyAlbanyUnited States
| | | |
Collapse
|
34
|
Patton MS, Sheats SH, Siclair AN, Mathur BN. Alcohol potentiates multiple GABAergic inputs to dorsal striatum fast-spiking interneurons. Neuropharmacology 2023; 232:109527. [PMID: 37011784 PMCID: PMC10122715 DOI: 10.1016/j.neuropharm.2023.109527] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 03/16/2023] [Accepted: 03/28/2023] [Indexed: 04/04/2023]
Abstract
Parvalbumin-expressing dorsal striatal fast-spiking interneurons, comprising ∼1% of the total dorsal striatal neuronal population, are necessary for the expression of compulsive-like ethanol consumption mice. Fast-spiking interneurons are driven to fire by glutamatergic inputs derived primarily from the cortex. However, these neurons also receive substantial GABAergic input from two sources: the globus pallidus and the reticular nucleus of the thalamus. How ethanol modulates inhibitory input onto fast-spiking neurons is unclear and, more broadly, alcohol effects on GABAergic synaptic transmission onto GABAergic interneurons are understudied. Examining this, we found that acute bath application of ethanol (50 mM) potentiated GABAergic transmission from both the globus pallidus and the reticular nucleus of the thalamus onto fast-spiking interneurons in mouse of both sexes. This ethanol-induced potentiation required postsynaptic calcium and was not accompanied by a sustained change in presynaptic GABA release probability. Examining whether this ethanol effect persisted following chronic intermittent ethanol exposure, we found attenuated acute-ethanol potentiation of GABAergic transmission from both the globus pallidus and the reticular nucleus of the thalamus onto striatal fast-spiking interneurons. These data underscore the impact of ethanol on GABAergic signaling in the dorsal striatum and support the notion that ethanol may disinhibit the dorsolateral striatum.
Collapse
Affiliation(s)
- Michael S Patton
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Samuel H Sheats
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Allison N Siclair
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Brian N Mathur
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
35
|
Becchi S, Chieng B, Bradfield LA, Capellán R, Leung BK, Balleine BW. Cognitive effects of thalamostriatal degeneration are ameliorated by normalizing striatal cholinergic activity. SCIENCE ADVANCES 2023; 9:eade8247. [PMID: 37352346 PMCID: PMC10289650 DOI: 10.1126/sciadv.ade8247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 05/19/2023] [Indexed: 06/25/2023]
Abstract
The loss of neurons in parafascicular thalamus (Pf) and their inputs to dorsomedial striatum (DMS) in Lewy body disease (LBD) and Parkinson's disease dementia (PDD) have been linked to the effects of neuroinflammation. We found that, in rats, these inputs were necessary for both the function of striatal cholinergic interneurons (CINs) and the flexible encoding of the action-outcome (AO) associations necessary for goal-directed action, producing a burst-pause pattern of CIN firing but only during the remapping elicited by a shift in AO contingency. Neuroinflammation in the Pf abolished these changes in CIN activity and goal-directed control after the shift in contingency. However, both effects were rescued by either the peripheral or the intra-DMS administration of selegiline, a monoamine oxidase B inhibitor that we found also enhances adenosine triphosphatase activity in CINs. These findings suggest a potential treatment for the cognitive deficits associated with neuroinflammation affecting the function of the Pf and related structures.
Collapse
Affiliation(s)
- Serena Becchi
- School of Psychology, Faculty of Science, University of New South Wales, Sydney, Australia
| | - Billy Chieng
- School of Psychology, Faculty of Science, University of New South Wales, Sydney, Australia
| | - Laura A. Bradfield
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, Australia
| | - Roberto Capellán
- School of Psychology, Department of Psychobiology, National University for Distance Learning, Madrid, Spain
| | - Beatrice K. Leung
- School of Psychology, Faculty of Science, University of New South Wales, Sydney, Australia
| | - Bernard W. Balleine
- School of Psychology, Faculty of Science, University of New South Wales, Sydney, Australia
| |
Collapse
|
36
|
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: 0.5] [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
|
37
|
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
|
38
|
Araújo de Góis Morais PL, de Souza Cavalcante J, Engelberth RC, Guzen FP, Junior ESN, Paiva Cavalcanti JRL. Morphology and morphometry of interneuron subpopulations of the marmoset monkey (Callithrix jacchus) striatum. Neurosci Res 2023:S0168-0102(23)00036-6. [PMID: 36804600 DOI: 10.1016/j.neures.2023.02.002] [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: 10/11/2022] [Revised: 01/04/2023] [Accepted: 02/07/2023] [Indexed: 02/18/2023]
Abstract
The mammalian striatum has long been considered a homogeneous entity. However, neuroanatomical and histochemical studies reveal that the striatum is much more heterogeneous than previously suspected. The caudate (Cd) and putamen (Pu) are composed of two chemical compartments: the matrix and the striosomes. Striatal interneurons have been classified into a variety of morphological and neurochemical subtypes. In this study, we compared the distribution of multiple neurochemical markers in the striatum of marmosets and described the morphology of different types of striatum interneurons. The immunoreactivities of choline-acetyl transferase (ChAT), neuropeptide Y (NPY), nitric oxide synthase (NOS), calretinin (CR), parvalbumin (PV) were analyzed along the entire rostrocaudal extent of the marmoset striatum. Calbindin immunohistochemistry is useful in identifying medium spiny neurons (MSNs), with efficient soma staining. Based on the size of the CB-positive cells, considered medium-sized, as expected, cholinergic cells are larger in area and diameter than the other subpopulations investigated, followed by NOS, NPY, PV and CR. In adjacent CB and PV-stained sections, the matrix and striosomes were clearly distinguished. The matrix is strongly reactive to CB and PV neuropils, while the striosomes exhibit low reactivity, especially in the dorsal Cd. Therefore, we provide a detailed description morphology and distribution of striatal interneuron populations in a model as a valuable tool for studying neurodegenerative pathogenesis, progression and treatment strategies.
Collapse
|
39
|
Ahmed NY, Knowles R, Liu L, Yan Y, Li X, Schumann U, Wang Y, Sontani Y, Reynolds N, Natoli R, Wen J, Del Pino I, Mi D, Dehorter N. Developmental deficits of MGE-derived interneurons in the Cntnap2 knockout mouse model of autism spectrum disorder. Front Cell Dev Biol 2023; 11:1112062. [PMID: 36819097 PMCID: PMC9930104 DOI: 10.3389/fcell.2023.1112062] [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: 11/30/2022] [Accepted: 01/17/2023] [Indexed: 02/04/2023] Open
Abstract
Interneurons are fundamental cells for maintaining the excitation-inhibition balance in the brain in health and disease. While interneurons have been shown to play a key role in the pathophysiology of autism spectrum disorder (ASD) in adult mice, little is known about how their maturation is altered in the developing striatum in ASD. Here, we aimed to track striatal developing interneurons and elucidate the molecular and physiological alterations in the Cntnap2 knockout mouse model. Using Stereo-seq and single-cell RNA sequencing data, we first characterized the pattern of expression of Cntnap2 in the adult brain and at embryonic stages in the medial ganglionic eminence (MGE), a transitory structure producing most cortical and striatal interneurons. We found that Cntnap2 is enriched in the striatum, compared to the cortex, particularly in the developing striatal cholinergic interneurons. We then revealed enhanced MGE-derived cell proliferation, followed by increased cell loss during the canonical window of developmental cell death in the Cntnap2 knockout mice. We uncovered specific cellular and molecular alterations in the developing Lhx6-expressing cholinergic interneurons of the striatum, which impacts interneuron firing properties during the first postnatal week. Overall, our work unveils some of the mechanisms underlying the shift in the developmental trajectory of striatal interneurons which greatly contribute to the ASD pathogenesis.
Collapse
Affiliation(s)
- Noorya Yasmin Ahmed
- The Australian National University, The John Curtin School of Medical Research, Canberra, ACT, Australia
| | - Rhys Knowles
- The Australian National University, The John Curtin School of Medical Research, Canberra, ACT, Australia
| | - Lixinyu Liu
- The Australian National University, The John Curtin School of Medical Research, Canberra, ACT, Australia
| | - Yiming Yan
- Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing, China
| | - Xiaohan Li
- Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing, China
| | - Ulrike Schumann
- The Australian National University, The John Curtin School of Medical Research, Canberra, ACT, Australia
| | - Yumeng Wang
- The Australian National University, The John Curtin School of Medical Research, Canberra, ACT, Australia
| | - Yovina Sontani
- The Australian National University, The John Curtin School of Medical Research, Canberra, ACT, Australia
| | - Nathan Reynolds
- The Australian National University, The John Curtin School of Medical Research, Canberra, ACT, Australia
| | - Riccardo Natoli
- The Australian National University, The John Curtin School of Medical Research, Canberra, ACT, Australia
| | - Jiayu Wen
- The Australian National University, The John Curtin School of Medical Research, Canberra, ACT, Australia
| | - Isabel Del Pino
- Institute of Neurosciences, Spanish National Research Council (CSIC), Sant Joan d’Alacant, Spain
| | - Da Mi
- Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing, China
| | - Nathalie Dehorter
- The Australian National University, The John Curtin School of Medical Research, Canberra, ACT, Australia,*Correspondence: Nathalie Dehorter,
| |
Collapse
|
40
|
Metzger JM, Wang Y, Neuman SS, Snow KJ, Murray SA, Lutz CM, Bondarenko V, Felton J, Gimse K, Xie R, Li D, Zhao Y, Flowers MT, Simmons HA, Roy S, Saha K, Levine JE, Emborg ME, Gong S. Efficient in vivo neuronal genome editing in the mouse brain using nanocapsules containing CRISPR-Cas9 ribonucleoproteins. Biomaterials 2023; 293:121959. [PMID: 36527789 PMCID: PMC9868115 DOI: 10.1016/j.biomaterials.2022.121959] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 12/05/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022]
Abstract
Genome editing of somatic cells via clustered regularly interspaced short palindromic repeats (CRISPR) offers promise for new therapeutics to treat a variety of genetic disorders, including neurological diseases. However, the dense and complex parenchyma of the brain and the post-mitotic state of neurons make efficient genome editing challenging. In vivo delivery systems for CRISPR-Cas proteins and single guide RNA (sgRNA) include both viral vectors and non-viral strategies, each presenting different advantages and disadvantages for clinical application. We developed non-viral and biodegradable PEGylated nanocapsules (NCs) that deliver preassembled Cas9-sgRNA ribonucleoproteins (RNPs). Here, we show that the RNP NCs led to robust genome editing in neurons following intracerebral injection into the healthy mouse striatum. Genome editing was predominantly observed in medium spiny neurons (>80%), with occasional editing in cholinergic, calretinin, and parvalbumin interneurons. Glial activation was minimal and was localized along the needle tract. Our results demonstrate that the RNP NCs are capable of safe and efficient neuronal genome editing in vivo.
Collapse
Affiliation(s)
- Jeanette M Metzger
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Yuyuan Wang
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53715, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Samuel S Neuman
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Kathy J Snow
- The Jackson Laboratory, Bar Harbor, ME, 04609, USA
| | | | | | - Viktoriya Bondarenko
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Jesi Felton
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Kirstan Gimse
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Ruosen Xie
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53715, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Dongdong Li
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53715, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Yi Zhao
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53715, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Matthew T Flowers
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Heather A Simmons
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Subhojit Roy
- Departments of Pathology and Neuroscience, University of California-San Diego, San Diego, CA, 92093, USA
| | - Krishanu Saha
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Jon E Levine
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, 53715, USA; Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Marina E Emborg
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, 53715, USA; Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, 53715, USA.
| | - Shaoqin Gong
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53715, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA.
| |
Collapse
|
41
|
Martel AC, Galvan A. Connectivity of the corticostriatal and thalamostriatal systems in normal and parkinsonian states: An update. Neurobiol Dis 2022; 174:105878. [PMID: 36183947 PMCID: PMC9976706 DOI: 10.1016/j.nbd.2022.105878] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 09/23/2022] [Accepted: 09/28/2022] [Indexed: 02/06/2023] Open
Abstract
The striatum receives abundant glutamatergic afferents from the cortex and thalamus. These inputs play a major role in the functions of the striatal neurons in normal conditions, and are significantly altered in pathological states, such as Parkinson's disease. This review summarizes the current knowledge of the connectivity of the corticostriatal and thalamostriatal pathways, with emphasis on the most recent advances in the field. We also discuss novel findings regarding structural changes in cortico- and thalamostriatal connections that occur in these connections as a consequence of striatal loss of dopamine in parkinsonism.
Collapse
Affiliation(s)
- Anne-Caroline Martel
- Emory National Primate Research Center, Emory University, Atlanta, GA, USA; Udall Center of Excellence for Parkinson's Disease Research, Emory University, Atlanta, GA, USA
| | - Adriana Galvan
- Emory National Primate Research Center, Emory University, Atlanta, GA, USA; Udall Center of Excellence for Parkinson's Disease Research, Emory University, Atlanta, GA, USA; Department of Neurology, School of Medicine, Emory University, Atlanta, GA, USA.
| |
Collapse
|
42
|
Joshi A, Schott M, la Fleur SE, Barrot M. Role of the striatal dopamine, GABA and opioid systems in mediating feeding and fat intake. Neurosci Biobehav Rev 2022; 139:104726. [PMID: 35691472 DOI: 10.1016/j.neubiorev.2022.104726] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 12/08/2021] [Accepted: 06/05/2022] [Indexed: 10/18/2022]
Abstract
Food intake, which is a highly reinforcing behavior, provides nutrients required for survival in all animals. However, when fat and sugar consumption goes beyond the daily needs, it can favor obesity. The prevalence and severity of this health problem has been increasing with time. Besides covering nutrient and energy needs, food and in particular its highly palatable components, such as fats, also induce feelings of joy and pleasure. Experimental evidence supports a role of the striatal complex and of the mesolimbic dopamine system in both feeding and food-related reward processing, with the nucleus accumbens as a key target for reward or reinforcing-associated signaling during food intake behavior. In this review, we provide insights concerning the impact of feeding, including fat intake, on different types of receptors and neurotransmitters present in the striatal complex. Reciprocally, we also cover the evidence for a modulation of palatable food intake by different neurochemical systems in the striatal complex and in particular the nucleus accumbens, with a focus on dopamine, GABA and the opioid system.
Collapse
Affiliation(s)
- Anil Joshi
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France; Amsterdam UMC, University of Amsterdam, Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam Gastroenterology & Metabolism, Amsterdam, the Netherlands; Amsterdam UMC, University of Amsterdam, Department of Endocrinology & Metabolism, Amsterdam Neuroscience, Amsterdam, the Netherlands; Metabolism and Reward Group, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW), Amsterdam, the Netherlands
| | - Marion Schott
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - Susanne Eva la Fleur
- Amsterdam UMC, University of Amsterdam, Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam Gastroenterology & Metabolism, Amsterdam, the Netherlands; Amsterdam UMC, University of Amsterdam, Department of Endocrinology & Metabolism, Amsterdam Neuroscience, Amsterdam, the Netherlands; Metabolism and Reward Group, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW), Amsterdam, the Netherlands.
| | - Michel Barrot
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France.
| |
Collapse
|
43
|
Tokarska A, Silberberg G. GABAergic interneurons expressing the α2 nicotinic receptor subunit are functionally integrated in the striatal microcircuit. Cell Rep 2022; 39:110842. [PMID: 35613598 DOI: 10.1016/j.celrep.2022.110842] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 03/08/2022] [Accepted: 04/28/2022] [Indexed: 11/29/2022] Open
Abstract
The interactions between the striatal cholinergic and GABAergic systems are crucial in shaping reward-related behavior and reinforcement learning; however, the synaptic pathways mediating them are largely unknown. Here, we use Chrna2-Cre mice to characterize striatal interneurons (INs) expressing the nicotinic α2 receptor subunit. Using triple patch-clamp recordings combined with optogenetic stimulations, we characterize the electrophysiological, morphological, and synaptic properties of striatal Chrna2-INs. Striatal Chrna2-INs have diverse electrophysiological properties, distinct from their counterparts in other brain regions, including the hippocampus and neocortex. Unlike in other regions, most striatal Chrna2-INs are fast-spiking INs expressing parvalbumin. Striatal Chrna2-INs are intricately integrated in the striatal microcircuit, forming inhibitory synaptic connections with striatal projection neurons and INs, including other Chrna2-INs. They receive excitatory inputs from primary motor cortex mediated by both AMPA and NMDA receptors. A subpopulation of Chrna2-INs responds to nicotinic input, suggesting reciprocal interactions between this GABAergic interneuron population and striatal cholinergic synapses.
Collapse
Affiliation(s)
- Anna Tokarska
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Gilad Silberberg
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden.
| |
Collapse
|
44
|
Matityahu L, Malgady JM, Schirelman M, Johansson Y, Wilking J, Silberberg G, Goldberg JA, Plotkin JL. A tonic nicotinic brake controls spike timing in striatal spiny projection neurons. eLife 2022; 11:75829. [PMID: 35579422 PMCID: PMC9142149 DOI: 10.7554/elife.75829] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 05/15/2022] [Indexed: 11/13/2022] Open
Abstract
Striatal spiny projection neurons (SPNs) transform convergent excitatory corticostriatal inputs into an inhibitory signal that shapes basal ganglia output. This process is fine-tuned by striatal GABAergic interneurons (GINs), which receive overlapping cortical inputs and mediate rapid corticostriatal feedforward inhibition of SPNs. Adding another level of control, cholinergic interneurons (CINs), which are also vigorously activated by corticostriatal excitation, can disynaptically inhibit SPNs by activating α4β2 nicotinic acetylcholine receptors (nAChRs) on various GINs. Measurements of this disynaptic inhibitory pathway, however, indicate that it is too slow to compete with direct GIN-mediated feedforward inhibition. Moreover, functional nAChRs are also present on populations of GINs that respond only weakly to phasic activation of CINs, such as parvalbumin-positive fast-spiking interneurons (PV-FSIs), making the overall role of nAChRs in shaping striatal synaptic integration unclear. Using acute striatal slices from mice we show that upon synchronous optogenetic activation of corticostriatal projections blockade of α4β2 nAChRs shortened SPN spike latencies and increased postsynaptic depolarizations. The nAChR-dependent inhibition was mediated by downstream GABA release, and data suggest that the GABA source was not limited to GINs that respond strongly to phasic CIN activation. In particular, the observed decrease in spike latency caused by nAChR blockade was associated with a diminished frequency of spontaneous inhibitory postsynaptic currents in SPNs, a parallel hyperpolarization of PV-FSIs, and was occluded by pharmacologically preventing cortical activation of PV-FSIs. Taken together, we describe a role for tonic (as opposed to phasic) activation of nAChRs in striatal function. We conclude that tonic activation of nAChRs by CINs maintains a GABAergic brake on cortically-driven striatal output by ‘priming’ feedforward inhibition, a process that may shape SPN spike timing, striatal processing, and synaptic plasticity.
Collapse
Affiliation(s)
- Lior Matityahu
- Department of Medical Neurobiology, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Jeffrey M Malgady
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, United States
| | - Meital Schirelman
- Department of Medical Neurobiology, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yvonne Johansson
- Sainsbury Wellcome Centre for Neural Circuits and Behaviour, University College London, London, United Kingdom
| | - Jennifer Wilking
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, United States
| | - Gilad Silberberg
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Joshua A Goldberg
- Department of Medical Neurobiology, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Joshua L Plotkin
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, United States
| |
Collapse
|
45
|
Del Rey NLG, Trigo-Damas I, Obeso JA, Cavada C, Blesa J. Neuron types in the primate striatum: stereological analysis of projection neurons and interneurons in control and parkinsonian monkeys. Neuropathol Appl Neurobiol 2022; 48:e12812. [PMID: 35274336 DOI: 10.1111/nan.12812] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 02/08/2022] [Accepted: 02/12/2022] [Indexed: 11/26/2022]
Abstract
AIMS The striatum is mainly composed of projection neurons. It also contains interneurons, which modulate and control striatal output. The aim of the present study was to assess the percentages of projection neurons and interneuron populations in the striatum of control monkeys and of parkinsonian monkeys. METHODS Unbiased stereology was used to estimate the volume density of every neuron population in the caudate, putamen and ventral striatum of control monkeys and of monkeys treated with MPTP, which results in striatal dopamine depletion. The various neuron population phenotypes were identified by immunohistochemistry. All analyses were performed within the same subjects using similar processing and analysis parameters, thus allowing for reliable data comparisons. RESULTS In control monkeys, the projection neurons, which express the Dopamine-and-cAMP-Regulated-Phosphoprotein, 32-KDa (DARPP-32), were the most abundant: ~86% of the total neurons counted. The interneurons accounted for the remaining 14%. Among the interneurons, those expressing Calretinin were the most abundant (Cr+: ~57%; ~8% of the total striatal neurons counted), followed those expressing Parvalbumin (Pv+: ~18 %; 2.6%), Dinucleotide Phosphate-Diaphorase (NADPH+: ~13 %; 1.8%), Choline Acetyltransferase (ChAT+: ~11%; 1.5%) and Tyrosine Hydroxylase (TH+: ~0.5%; 0.1%). No significant changes in volume densities occurred in any population following dopamine depletion, except for the TH+ interneurons, which increased in parkinsonian non-symptomatic monkeys and even more in symptomatic monkeys. CONCLUSIONS These data are relevant for translational studies targeting specific neuron populations of the striatum. The fact that dopaminergic denervation does not cause neuron loss in any population has potential pathophysiological implications.
Collapse
Affiliation(s)
- Natalia López-González Del Rey
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain.,CIBERNED (Center for Networked Biomedical Research on Neurodegenerative Diseases), Instituto Carlos III, Madrid, Spain.,PhD Program in Neuroscience Autónoma de Madrid University-Cajal Institute, Madrid, Spain
| | - Inés Trigo-Damas
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain.,CIBERNED (Center for Networked Biomedical Research on Neurodegenerative Diseases), Instituto Carlos III, Madrid, Spain
| | - J A Obeso
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain.,CIBERNED (Center for Networked Biomedical Research on Neurodegenerative Diseases), Instituto Carlos III, Madrid, Spain
| | - Carmen Cavada
- PhD Program in Neuroscience Autónoma de Madrid University-Cajal Institute, Madrid, Spain.,Department of Anatomy, Histology and Neuroscience, School of Medicine, Autónoma de Madrid University, Madrid, Spain
| | - Javier Blesa
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain.,CIBERNED (Center for Networked Biomedical Research on Neurodegenerative Diseases), Instituto Carlos III, Madrid, Spain
| |
Collapse
|
46
|
Xu L, Zhang C, Zhong M, Che F, Guan C, Zheng X, Liu S. Role of histidine decarboxylase gene in the pathogenesis of Tourette syndrome. Brain Behav 2022; 12:e2511. [PMID: 35114079 PMCID: PMC8933785 DOI: 10.1002/brb3.2511] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 11/18/2021] [Accepted: 01/06/2022] [Indexed: 12/15/2022] Open
Abstract
Tourette syndrome (TS) is caused by complex genetic and environmental factors and is characterized by tics. Histidine decarboxylase (HDC) mutation is a rare genetic cause with high penetrance in patients with TS. HDC-knockout (KO) mice have similar behavioral and neurochemical abnormalities as patients with TS. Therefore, HDC-KO mice are considered a valuable TS pathophysiological model as it reveals the underlying pathological mechanisms that cannot be obtained from patients with TS, thus advancing the development of treatment strategies for TS and other tic disorders. This review summarizes some of the recent research hotspots and progress in HDC-KO mice, aiming to deepen our understanding of brain mechanisms relevant to TS. Furthermore, we encapsulate the possible brain nerve cell changes in HDC-KO mice and their potential roles in TS to provide multiple directions for the future research on tics.
Collapse
Affiliation(s)
- Lulu Xu
- Department of Geriatric Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Cheng Zhang
- Department of Neurology, The Eleventh Clinical Medical College of Qingdao University, Linyi People's Hospital, Linyi, Shandong, China
| | - Meixiang Zhong
- Department of Geriatric Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Fengyuan Che
- Department of Neurology, The Eleventh Clinical Medical College of Qingdao University, Linyi People's Hospital, Linyi, Shandong, China
| | - Chengcheng Guan
- Department of Medical Cenetics, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xueping Zheng
- Department of Geriatric Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Shiguo Liu
- Department of Medical Cenetics, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
47
|
Scarduzio M, Hess EJ, Standaert DG, Eskow Jaunarajs KL. Striatal synaptic dysfunction in dystonia and levodopa-induced dyskinesia. Neurobiol Dis 2022; 166:105650. [DOI: 10.1016/j.nbd.2022.105650] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 12/16/2022] Open
|
48
|
Shipman ML, Corbit LH. Diet-induced deficits in goal-directed control are rescued by agonism of group II metabotropic glutamate receptors in the dorsomedial striatum. Transl Psychiatry 2022; 12:42. [PMID: 35091538 PMCID: PMC8799694 DOI: 10.1038/s41398-022-01807-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 01/06/2022] [Accepted: 01/12/2022] [Indexed: 11/09/2022] Open
Abstract
Many overweight or obese people struggle to sustain the behavioural changes necessary to achieve and maintain weight loss. In rodents, obesogenic diet can disrupt goal-directed control of responding for food reinforcers, which may indicate that diet can disrupt brain regions associated with behavioural control. We investigated a potential glutamatergic mechanism to return goal-directed control to rats who had been given an obesogenic diet prior to operant training. We found that an obesogenic diet reduced goal-directed control and that systemic injection of LY379268, a Group II metabotropic glutamate receptor (mGluR2/3) agonist, returned goal-directed responding in these rats. Further, we found that direct infusion of LY379268 into the dorsomedial striatum, a region associated with goal-directed control, also restored goal-directed responding in the obesogenic-diet group. This indicates that one mechanism through which obesogenic diet disrupts goal-directed control is glutamatergic, and infusion of a mGluR2/3 agonist into the DMS is sufficient to ameliorate deficits in goal-directed control.
Collapse
Affiliation(s)
- Megan L Shipman
- University of Toronto Department of Psychology, 100 St. George Street, Toronto, ON, M5S 3G3, Canada
| | - Laura H Corbit
- University of Toronto Department of Psychology, 100 St. George Street, Toronto, ON, M5S 3G3, Canada.
| |
Collapse
|
49
|
Dopamine depletion selectively disrupts interactions between striatal neuron subtypes and LFP oscillations. Cell Rep 2022; 38:110265. [PMID: 35045299 PMCID: PMC8820590 DOI: 10.1016/j.celrep.2021.110265] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 09/20/2021] [Accepted: 12/22/2021] [Indexed: 12/25/2022] Open
Abstract
Dopamine degeneration in Parkinson’s disease (PD) dysregulates the striatal neural network and causes motor deficits. However, it is unclear how altered striatal circuits relate to dopamine-acetylcholine chemical imbalance and abnormal local field potential (LFP) oscillations observed in PD. We perform a multimodal analysis of the dorsal striatum using cell-type-specific calcium imaging and LFP recording. We reveal that dopamine depletion selectively enhances LFP beta oscillations during impaired locomotion, supporting beta oscillations as a biomarker for PD. We further demonstrate that dynamic cholinergic interneuron activity during locomotion remains unaltered, even though cholinergic tone is implicated in PD. Instead, dysfunctional striatal output arises from elevated coordination within striatal output neurons, which is accompanied by reduced locomotor encoding of parvalbumin interneurons and transient pathological LFP high-gamma oscillations. These results identify a pathological striatal circuit state following dopamine depletion where distinct striatal neuron subtypes are selectively coordinated with LFP oscillations during locomotion. Zemel et al. demonstrate that dopamine loss disrupts striatal neural network and enhances local field potential beta oscillations during impaired locomotion. Specifically, striatal projecting neuron activation is abnormally coordinated and accompanied by pathological high-gamma oscillations. While parvalbumin interneurons reduce locomotor encoding, cholinergic interneurons strengthen their interactions with projecting neurons.
Collapse
|
50
|
LRRK2 at Striatal Synapses: Cell-Type Specificity and Mechanistic Insights. Cells 2022; 11:cells11010169. [PMID: 35011731 PMCID: PMC8750662 DOI: 10.3390/cells11010169] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/10/2021] [Accepted: 12/21/2021] [Indexed: 12/13/2022] Open
Abstract
Mutations in leucine-rich repeat kinase 2 (LRRK2) cause Parkinson’s disease with a similar clinical presentation and progression to idiopathic Parkinson’s disease, and common variation is linked to disease risk. Recapitulation of the genotype in rodent models causes abnormal dopamine release and increases the susceptibility of dopaminergic neurons to insults, making LRRK2 a valuable model for understanding the pathobiology of Parkinson’s disease. It is also a promising druggable target with targeted therapies currently in development. LRRK2 mRNA and protein expression in the brain is highly variable across regions and cellular identities. A growing body of work has demonstrated that pathogenic LRRK2 mutations disrupt striatal synapses before the onset of overt neurodegeneration. Several substrates and interactors of LRRK2 have been identified to potentially mediate these pre-neurodegenerative changes in a cell-type-specific manner. This review discusses the effects of pathogenic LRRK2 mutations in striatal neurons, including cell-type-specific and pathway-specific alterations. It also highlights several LRRK2 effectors that could mediate the alterations to striatal function, including Rabs and protein kinase A. The lessons learned from improving our understanding of the pathogenic effects of LRRK2 mutations in striatal neurons will be applicable to both dissecting the cell-type specificity of LRRK2 function in the transcriptionally diverse subtypes of dopaminergic neurons and also increasing our understanding of basal ganglia development and biology. Finally, it will inform the development of therapeutics for Parkinson’s disease.
Collapse
|