1
|
Sheng M, Lu D, Sheng K, Ding JB. Activity-Dependent Remodeling of Corticostriatal Axonal Boutons During Motor Learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.10.598366. [PMID: 38915677 PMCID: PMC11195117 DOI: 10.1101/2024.06.10.598366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Motor skill learning induces long-lasting synaptic plasticity at not only the inputs, such as dendritic spines1-4, but also at the outputs to the striatum of motor cortical neurons5,6. However, very little is known about the activity and structural plasticity of corticostriatal axons during learning in the adult brain. Here, we used longitudinal in vivo two-photon imaging to monitor the activity and structure of thousands of corticostriatal axonal boutons in the dorsolateral striatum in awake mice. We found that learning a new motor skill induces dynamic regulation of axonal boutons. The activities of motor corticostriatal axonal boutons exhibited selectivity for rewarded movements (RM) and un-rewarded movements (UM). Strikingly, boutons on the same axonal branches showed diverse responses during behavior. Motor learning significantly increased the fraction of RM boutons and reduced the heterogeneity of bouton activities. Moreover, motor learning-induced profound structural dynamism in boutons. By combining structural and functional imaging, we identified that newly formed axonal boutons are more likely to exhibit selectivity for RM and are stabilized during motor learning, while UM boutons are selectively eliminated. Our results highlight a novel form of plasticity at corticostriatal axons induced by motor learning, indicating that motor corticostriatal axonal boutons undergo dynamic reorganization that facilitates the acquisition and execution of motor skills.
Collapse
Affiliation(s)
- Mengjun Sheng
- Department of Neurosurgery, Stanford University School of Medicine
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- These authors contributed equally
| | - Di Lu
- Department of Neurosurgery, Stanford University School of Medicine
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- These authors contributed equally
| | - Kaiwen Sheng
- Department of Neurosurgery, Stanford University School of Medicine
- Stanford Bioengineering PhD program, Stanford University
| | - Jun B Ding
- Department of Neurosurgery, Stanford University School of Medicine
- 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
| |
Collapse
|
2
|
Cording KR, Tu EM, Wang H, Agopyan-Miu AHCW, Bateup HS. Cntnap2 loss drives striatal neuron hyperexcitability and behavioral inflexibility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.09.593387. [PMID: 38766169 PMCID: PMC11100810 DOI: 10.1101/2024.05.09.593387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by two major diagnostic criteria - persistent deficits in social communication and interaction, and the presence of restricted, repetitive patterns of behavior (RRBs). Evidence from both human and animal model studies of ASD suggest that alteration of striatal circuits, which mediate motor learning, action selection, and habit formation, may contribute to the manifestation of RRBs. CNTNAP2 is a syndromic ASD risk gene, and loss of function of Cntnap2 in mice is associated with RRBs. How loss of Cntnap2 impacts striatal neuron function is largely unknown. In this study, we utilized Cntnap2-/- mice to test whether altered striatal neuron activity contributes to aberrant motor behaviors relevant to ASD. We find that Cntnap2-/- mice exhibit increased cortical drive of striatal projection neurons (SPNs), with the most pronounced effects in direct pathway SPNs. This enhanced drive is likely due to increased intrinsic excitability of SPNs, which make them more responsive to cortical inputs. We also find that Cntnap2-/- mice exhibit spontaneous repetitive behaviors, increased motor routine learning, and cognitive inflexibility. Increased corticostriatal drive, in particular of the direct pathway, may contribute to the acquisition of repetitive, inflexible behaviors in Cntnap2 mice.
Collapse
Affiliation(s)
- Katherine R. Cording
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA USA
| | - Emilie M. Tu
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA USA
| | - Hongli Wang
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA USA
| | | | - Helen S. Bateup
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA USA
| |
Collapse
|
3
|
Do QB, Noor H, Marquez-Gomez R, Cramb KML, Ng B, Abbey A, Ibarra-Aizpurua N, Caiazza MC, Sharifi P, Lang C, Beccano-Kelly D, Baleriola J, Bengoa-Vergniory N, Wade-Martins R. Early deficits in an in vitro striatal microcircuit model carrying the Parkinson's GBA-N370S mutation. NPJ Parkinsons Dis 2024; 10:82. [PMID: 38609392 PMCID: PMC11014935 DOI: 10.1038/s41531-024-00694-2] [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: 03/11/2023] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
Understanding medium spiny neuron (MSN) physiology is essential to understand motor impairments in Parkinson's disease (PD) given the architecture of the basal ganglia. Here, we developed a custom three-chambered microfluidic platform and established a cortico-striato-nigral microcircuit partially recapitulating the striatal presynaptic landscape in vitro using induced pluripotent stem cell (iPSC)-derived neurons. We found that, cortical glutamatergic projections facilitated MSN synaptic activity, and dopaminergic transmission enhanced maturation of MSNs in vitro. Replacement of wild-type iPSC-derived dopamine neurons (iPSC-DaNs) in the striatal microcircuit with those carrying the PD-related GBA-N370S mutation led to a depolarisation of resting membrane potential and an increase in rheobase in iPSC-MSNs, as well as a reduction in both voltage-gated sodium and potassium currents. Such deficits were resolved in late microcircuit cultures, and could be reversed in younger cultures with antagonism of protein kinase A activity in iPSC-MSNs. Taken together, our results highlight the unique utility of modelling striatal neurons in a modular physiological circuit to reveal mechanistic insights into GBA1 mutations in PD.
Collapse
Affiliation(s)
- Quyen B Do
- Oxford Parkinson's Disease Centre and Department of Physiology, Anatomy and Genetics, University of Oxford, South Park Road, Oxford, OX1 3QU, UK
- Kavli Institute for Neuroscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Park Road, Oxford, OX1 3QU, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Humaira Noor
- Oxford Parkinson's Disease Centre and Department of Physiology, Anatomy and Genetics, University of Oxford, South Park Road, Oxford, OX1 3QU, UK
- Kavli Institute for Neuroscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Park Road, Oxford, OX1 3QU, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Nuffield Department of Medicine (NDM), University of Oxford, Henry Wellcome Building for Molecular Physiology, Old Road, Oxford, OX3 7BN, UK
| | - Ricardo Marquez-Gomez
- Oxford Parkinson's Disease Centre and Department of Physiology, Anatomy and Genetics, University of Oxford, South Park Road, Oxford, OX1 3QU, UK
- Kavli Institute for Neuroscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Park Road, Oxford, OX1 3QU, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Kaitlyn M L Cramb
- Oxford Parkinson's Disease Centre and Department of Physiology, Anatomy and Genetics, University of Oxford, South Park Road, Oxford, OX1 3QU, UK
- Kavli Institute for Neuroscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Park Road, Oxford, OX1 3QU, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Bryan Ng
- Oxford Parkinson's Disease Centre and Department of Physiology, Anatomy and Genetics, University of Oxford, South Park Road, Oxford, OX1 3QU, UK
- Kavli Institute for Neuroscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Park Road, Oxford, OX1 3QU, UK
| | - Ajantha Abbey
- Oxford Parkinson's Disease Centre and Department of Physiology, Anatomy and Genetics, University of Oxford, South Park Road, Oxford, OX1 3QU, UK
- Kavli Institute for Neuroscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Park Road, Oxford, OX1 3QU, UK
| | - Naroa Ibarra-Aizpurua
- Oxford Parkinson's Disease Centre and Department of Physiology, Anatomy and Genetics, University of Oxford, South Park Road, Oxford, OX1 3QU, UK
- Kavli Institute for Neuroscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Park Road, Oxford, OX1 3QU, UK
| | - Maria Claudia Caiazza
- Oxford Parkinson's Disease Centre and Department of Physiology, Anatomy and Genetics, University of Oxford, South Park Road, Oxford, OX1 3QU, UK
- Kavli Institute for Neuroscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Park Road, Oxford, OX1 3QU, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Parnaz Sharifi
- Oxford Parkinson's Disease Centre and Department of Physiology, Anatomy and Genetics, University of Oxford, South Park Road, Oxford, OX1 3QU, UK
- Kavli Institute for Neuroscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Park Road, Oxford, OX1 3QU, UK
| | - Charmaine Lang
- Oxford Parkinson's Disease Centre and Department of Physiology, Anatomy and Genetics, University of Oxford, South Park Road, Oxford, OX1 3QU, UK
- Kavli Institute for Neuroscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Park Road, Oxford, OX1 3QU, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Dayne Beccano-Kelly
- Oxford Parkinson's Disease Centre and Department of Physiology, Anatomy and Genetics, University of Oxford, South Park Road, Oxford, OX1 3QU, UK
| | - Jimena Baleriola
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- Ikerbasque-Basque Foundation for Science, Bilbao, Spain
| | - Nora Bengoa-Vergniory
- Oxford Parkinson's Disease Centre and Department of Physiology, Anatomy and Genetics, University of Oxford, South Park Road, Oxford, OX1 3QU, UK.
- Kavli Institute for Neuroscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Park Road, Oxford, OX1 3QU, UK.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA.
- Achucarro Basque Center for Neuroscience, Leioa, Spain.
- Ikerbasque-Basque Foundation for Science, Bilbao, Spain.
- University of the Basque Country (UPV/EHU), Department of Neuroscience, Leioa, Spain.
| | - Richard Wade-Martins
- Oxford Parkinson's Disease Centre and Department of Physiology, Anatomy and Genetics, University of Oxford, South Park Road, Oxford, OX1 3QU, UK.
- Kavli Institute for Neuroscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Park Road, Oxford, OX1 3QU, UK.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA.
| |
Collapse
|
4
|
Favila N, Gurney K, Overton PG. Role of the basal ganglia in innate and learned behavioural sequences. Rev Neurosci 2024; 35:35-55. [PMID: 37437141 DOI: 10.1515/revneuro-2023-0038] [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: 03/31/2023] [Accepted: 06/24/2023] [Indexed: 07/14/2023]
Abstract
Integrating individual actions into coherent, organised behavioural units, a process called chunking, is a fundamental, evolutionarily conserved process that renders actions automatic. In vertebrates, evidence points to the basal ganglia - a complex network believed to be involved in action selection - as a key component of action sequence encoding, although the underlying mechanisms are only just beginning to be understood. Central pattern generators control many innate automatic behavioural sequences that form some of the most basic behaviours in an animal's repertoire, and in vertebrates, brainstem and spinal pattern generators are under the control of higher order structures such as the basal ganglia. Evidence suggests that the basal ganglia play a crucial role in the concatenation of simpler behaviours into more complex chunks, in the context of innate behavioural sequences such as chain grooming in rats, as well as sequences in which innate capabilities and learning interact such as birdsong, and sequences that are learned from scratch, such as lever press sequences in operant behaviour. It has been proposed that the role of the striatum, the largest input structure of the basal ganglia, might lie in selecting and allowing the relevant central pattern generators to gain access to the motor system in the correct order, while inhibiting other behaviours. As behaviours become more complex and flexible, the pattern generators seem to become more dependent on descending signals. Indeed, during learning, the striatum itself may adopt the functional characteristics of a higher order pattern generator, facilitated at the microcircuit level by striatal neuropeptides.
Collapse
Affiliation(s)
- Natalia Favila
- German Center for Neurodegenerative Diseases, 53127 Bonn, Germany
| | - Kevin Gurney
- Department of Psychology, The University of Sheffield, Sheffield S1 2LT, UK
| | - Paul G Overton
- Department of Psychology, The University of Sheffield, Sheffield S1 2LT, UK
| |
Collapse
|
5
|
Fang LZ, Creed MC. Updating the striatal-pallidal wiring diagram. Nat Neurosci 2024; 27:15-27. [PMID: 38057614 DOI: 10.1038/s41593-023-01518-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 11/06/2023] [Indexed: 12/08/2023]
Abstract
The striatal and pallidal complexes are basal ganglia structures that orchestrate learning and execution of flexible behavior. Models of how the basal ganglia subserve these functions have evolved considerably, and the advent of optogenetic and molecular tools has shed light on the heterogeneity of subcircuits within these pathways. However, a synthesis of how molecularly diverse neurons integrate into existing models of basal ganglia function is lacking. Here, we provide an overview of the neurochemical and molecular diversity of striatal and pallidal neurons and synthesize recent circuit connectivity studies in rodents that takes this diversity into account. We also highlight anatomical organizational principles that distinguish the dorsal and ventral basal ganglia pathways in rodents. Future work integrating the molecular and anatomical properties of striatal and pallidal subpopulations may resolve controversies regarding basal ganglia network function.
Collapse
Affiliation(s)
- Lisa Z Fang
- Washington University Pain Center, Department of Anesthesiology, St. Louis, MO, USA
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, Newfoundland and Labrador, Canada
| | - Meaghan C Creed
- Washington University Pain Center, Department of Anesthesiology, St. Louis, MO, USA.
- Departments of Psychiatry, Neuroscience and Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
6
|
Frost-Nylén J, Thompson WS, Robertson B, Grillner S. The Basal Ganglia Downstream Control of Action - An Evolutionarily Conserved Strategy. Curr Neuropharmacol 2024; 22:1419-1430. [PMID: 37563813 PMCID: PMC11097981 DOI: 10.2174/1570159x21666230810141746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/30/2023] [Accepted: 02/05/2023] [Indexed: 08/12/2023] Open
Abstract
The motor areas of the cortex and the basal ganglia both contribute to determining which motor actions will be recruited at any moment in time, and their functions are intertwined. Here, we review the basal ganglia mechanisms underlying the selection of behavior of the downstream control of motor centers in the midbrain and brainstem and show that the basic organization of the forebrain motor system is evolutionarily conserved throughout vertebrate phylogeny. The output level of the basal ganglia (e.g. substantia nigra pars reticulata) has GABAergic neurons that are spontaneously active at rest and inhibit a number of specific motor centers, each of which can be relieved from inhibition if the inhibitory output neurons themselves become inhibited. The motor areas of the cortex act partially via the dorsolateral striatum (putamen), which has specific modules for the forelimb, hindlimb, trunk, etc. Each module operates in turn through the two types of striatal projection neurons that control the output modules of the basal ganglia and thereby the downstream motor centers. The mechanisms for lateral inhibition in the striatum are reviewed as well as other striatal mechanisms contributing to action selection. The motor cortex also exerts a direct excitatory action on specific motor centers. An overview is given of the basal ganglia control exerted on the different midbrain/brainstem motor centers, and the efference copy information fed back via the thalamus to the striatum and cortex, which is of importance for the planning of future movements.
Collapse
Affiliation(s)
| | | | - Brita Robertson
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Sten Grillner
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
7
|
Tiroshi L, Atamna Y, Gilin N, Berkowitz N, Goldberg JA. Striatal Neurons Are Recruited Dynamically into Collective Representations of Self-Initiated and Learned Actions in Freely Moving Mice. eNeuro 2024; 11:ENEURO.0315-23.2023. [PMID: 38164559 PMCID: PMC11057506 DOI: 10.1523/eneuro.0315-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 11/05/2023] [Accepted: 11/17/2023] [Indexed: 01/03/2024] Open
Abstract
Striatal spiny projection neurons are hyperpolarized-at-rest (HaR) and driven to action potential threshold by a small number of powerful inputs-an input-output configuration that is detrimental to response reliability. Because the striatum is important for habitual behaviors and goal-directed learning, we conducted a microendoscopic imaging in freely moving mice that express a genetically encoded Ca2+ indicator sparsely in striatal HaR neurons to evaluate their response reliability during self-initiated movements and operant conditioning. The sparse expression was critical for longitudinal studies of response reliability, and for studying correlations among HaR neurons while minimizing spurious correlations arising from contamination by the background signal. We found that HaR neurons are recruited dynamically into action representation, with distinct neuronal subsets being engaged in a moment-by-moment fashion. While individual neurons respond with little reliability, the population response remained stable across days. Moreover, we found evidence for the temporal coupling between neuronal subsets during conditioned (but not innate) behaviors.
Collapse
Affiliation(s)
- Lior Tiroshi
- Department of Medical Neurobiology, Institute of Medical Research Israel - Canada, The Faculty of Medicine, The Hebrew University of Jerusalem, 9112102, Jerusalem, Israel
| | - Yara Atamna
- Department of Medical Neurobiology, Institute of Medical Research Israel - Canada, The Faculty of Medicine, The Hebrew University of Jerusalem, 9112102, Jerusalem, Israel
| | - Naomi Gilin
- Department of Medical Neurobiology, Institute of Medical Research Israel - Canada, The Faculty of Medicine, The Hebrew University of Jerusalem, 9112102, Jerusalem, Israel
| | - Noa Berkowitz
- Department of Medical Neurobiology, Institute of Medical Research Israel - Canada, The Faculty of Medicine, The Hebrew University of Jerusalem, 9112102, Jerusalem, Israel
| | - Joshua A Goldberg
- Department of Medical Neurobiology, Institute of Medical Research Israel - Canada, The Faculty of Medicine, The Hebrew University of Jerusalem, 9112102, Jerusalem, Israel
| |
Collapse
|
8
|
Gómez-Ocádiz R, Silberberg G. Corticostriatal pathways for bilateral sensorimotor functions. Curr Opin Neurobiol 2023; 83:102781. [PMID: 37696188 DOI: 10.1016/j.conb.2023.102781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 08/15/2023] [Accepted: 08/16/2023] [Indexed: 09/13/2023]
Abstract
Corticostriatal pathways are essential for a multitude of motor, sensory, cognitive, and affective functions. They are mediated by cortical pyramidal neurons, roughly divided into two projection classes: the pyramidal tract (PT) and the intratelencephalic tract (IT). These pathways have been the focus of numerous studies in recent years, revealing their distinct structural and functional properties. Notably, their synaptic connectivity within ipsi- and contralateral cortical and striatal microcircuits is characterized by a high degree of target selectivity, providing a means to regulate the local neuromodulatory landscape in the striatum. Here, we discuss recent findings regarding the functional organization of the PT and IT corticostriatal pathways and its implications for bilateral sensorimotor functions.
Collapse
Affiliation(s)
- Ruy Gómez-Ocádiz
- Department of Neuroscience, Karolinska Institutet, Stockholm 17177, Sweden. https://twitter.com/@RuyGomezOcadiz
| | - Gilad Silberberg
- Department of Neuroscience, Karolinska Institutet, Stockholm 17177, Sweden.
| |
Collapse
|
9
|
Cording KR, Bateup HS. Altered motor learning and coordination in mouse models of autism spectrum disorder. Front Cell Neurosci 2023; 17:1270489. [PMID: 38026686 PMCID: PMC10663323 DOI: 10.3389/fncel.2023.1270489] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 09/25/2023] [Indexed: 12/01/2023] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder with increasing prevalence. Over 1,000 risk genes have now been implicated in ASD, suggesting diverse etiology. However, the diagnostic criteria for the disorder still comprise two major behavioral domains - deficits in social communication and interaction, and the presence of restricted and repetitive patterns of behavior (RRBs). The RRBs associated with ASD include both stereotyped repetitive movements and other motor manifestations including changes in gait, balance, coordination, and motor skill learning. In recent years, the striatum, the primary input center of the basal ganglia, has been implicated in these ASD-associated motor behaviors, due to the striatum's role in action selection, motor learning, and habit formation. Numerous mouse models with mutations in ASD risk genes have been developed and shown to have alterations in ASD-relevant behaviors. One commonly used assay, the accelerating rotarod, allows for assessment of both basic motor coordination and motor skill learning. In this corticostriatal-dependent task, mice walk on a rotating rod that gradually increases in speed. In the extended version of this task, mice engage striatal-dependent learning mechanisms to optimize their motor routine and stay on the rod for longer periods. This review summarizes the findings of studies examining rotarod performance across a range of ASD mouse models, and the resulting implications for the involvement of striatal circuits in ASD-related motor behaviors. While performance in this task is not uniform across mouse models, there is a cohort of models that show increased rotarod performance. A growing number of studies suggest that this increased propensity to learn a fixed motor routine may reflect a common enhancement of corticostriatal drive across a subset of mice with mutations in ASD-risk genes.
Collapse
Affiliation(s)
- Katherine R. Cording
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, United States
| | - Helen S. Bateup
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, United States
- Molecular and Cell Biology Department, University of California, Berkeley, Berkeley, CA, United States
- Chan Zuckerberg Biohub, San Francisco, CA, United States
| |
Collapse
|
10
|
Klavinskis-Whiting S, Bitzenhofer S, Hanganu-Opatz I, Ellender T. Generation and propagation of bursts of activity in the developing basal ganglia. Cereb Cortex 2023; 33:10595-10613. [PMID: 37615347 PMCID: PMC10560579 DOI: 10.1093/cercor/bhad307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/25/2023] Open
Abstract
The neonatal brain is characterized by intermittent bursts of oscillatory activity interspersed by relative silence. Although well-characterized for many cortical areas, to what extent these propagate and interact with subcortical brain areas is largely unknown. Here, early network activity was recorded from the developing basal ganglia, including motor/somatosensory cortex, dorsal striatum, and intralaminar thalamus, during the first postnatal weeks in mice. An unsupervised detection and classification method revealed two main classes of bursting activity, namely spindle bursts and nested gamma spindle bursts, characterized by oscillatory activity at ~ 10 and ~ 30 Hz frequencies, respectively. These were reliably identified across all three brain regions and exhibited region-specific differences in their structural, spectral, and developmental characteristics. Bursts of the same type often co-occurred in different brain regions and coherence and cross-correlation analyses reveal dynamic developmental changes in their interactions. The strongest interactions were seen for cortex and striatum, from the first postnatal week onwards, and cortex appeared to drive burst events in subcortical regions. Together, these results provide the first detailed description of early network activity within the developing basal ganglia and suggest that cortex is one of the main drivers of activity in downstream nuclei during this postnatal period.
Collapse
Affiliation(s)
| | - Sebastian Bitzenhofer
- Department of Biomedical Sciences, Institute of Developmental Neurophysiology, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Ileana Hanganu-Opatz
- Department of Biomedical Sciences, Institute of Developmental Neurophysiology, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Tommas Ellender
- Department of Pharmacology, University of Oxford, Mansfield Rd, Oxford, OX13QT, United Kingdom
- Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| |
Collapse
|
11
|
Zhong M, Wang Y, Lin G, Liao FF, Zhou FM. Dopamine-independent development and maintenance of mouse striatal medium spiny neuron dendritic spines. Neurobiol Dis 2023; 181:106096. [PMID: 37001611 PMCID: PMC10864017 DOI: 10.1016/j.nbd.2023.106096] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 03/18/2023] [Accepted: 03/20/2023] [Indexed: 03/31/2023] Open
Abstract
Striatal medium spiny neurons (MSNs) and striatal dopamine (DA) innervation are profoundly important for brain function such as motor control and cognition. A widely accepted theory posits that striatal DA loss causes (or leads to) MSN dendritic atrophy. However, examination of the literature indicates that the data from Parkinson's disease (PD) patients and animal PD models were contradictory among studies and hard to interpret. Here we have re-examined the potential effects of DA activity on MSN morphology or lack thereof. We found that in 15-day, 4- and 12-month old Pitx3 null mutant mice that have severe DA denervation in the dorsal striatum while having substantial residual DA innervation in the ventral striatum, MSN dendrites and spine numbers were similar in dorsal and ventral striatum, and also similar to those in normal mice. In 15-day, 4- and 12-month old tyrosine hydroxylase knockout mice that cannot synthesize L-dopa and thus have no endogenous DA in the entire brain, MSN dendrites and spine numbers were also indistinguishable from age-matched wild-type (WT) mice. Furthermore, in adult WT mice, unilateral 6-OHDA lesion at 12 months of age caused an almost complete striatal DA denervation in the lesioned side, but MSN dendrites and spine numbers were similar in the lesioned and control sides. Taken together, our data indicate that in mice, the development and maintenance of MSN dendrites and spines are DA-independent such that DA depletion does not trigger MSN dendritic atrophy; our data also suggest that the reported MSN dendritic atrophy in PD may be a component of neurodegeneration in PD rather than a consequence of DA denervation.
Collapse
Affiliation(s)
- Manli Zhong
- College of Life and Health Sciences, Northeastern University, No.195, Chuangxin Road, Hunnan District, Shenyang 110169, China; Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, University of Tennessee, Memphis, TN 38103, USA.
| | - Yuhan Wang
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, University of Tennessee, Memphis, TN 38103, USA
| | - Geng Lin
- Teaching Center for Basic Medical Experiments, China Medical University, Shenyang 110122, China; Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, University of Tennessee, Memphis, TN 38103, USA
| | - Francesca-Fang Liao
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, University of Tennessee, Memphis, TN 38103, USA
| | - Fu-Ming Zhou
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, University of Tennessee, Memphis, TN 38103, USA.
| |
Collapse
|
12
|
Arasaratnam CJ, Song JJ, Yoshida T, Curtis MA, Graybiel AM, Faull RLM, Waldvogel HJ. DARPP-32 cells and neuropil define striosomal system and isolated matrix cells in human striatum. J Comp Neurol 2023; 531:888-920. [PMID: 37002560 PMCID: PMC10392785 DOI: 10.1002/cne.25473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 01/30/2023] [Accepted: 02/28/2023] [Indexed: 04/04/2023]
Abstract
The dorsal striatum forms a central node of the basal ganglia interconnecting the neocortex and thalamus with circuits modulating mood and movement. Striatal projection neurons (SPNs) include relatively intermixed populations expressing D1-type or D2-type dopamine receptors (dSPNs and iSPNs) that give rise to the direct (D1) and indirect (D2) output systems of the basal ganglia. Overlaid on this organization is a compartmental organization, in which a labyrinthine system of striosomes made up of sequestered SPNs is embedded within the larger striatal matrix. Striosomal SPNs also include D1-SPNs and D2-SPNs, but they can be distinguished from matrix SPNs by many neurochemical markers. In the rodent striatum the key signaling molecule, DARPP-32, is a exception to these compartmental expression patterns, thought to befit its functions through opposite actions in both D1- and D2-expressing SPNs. We demonstrate here, however, that in the dorsal human striatum, DARPP-32 is concentrated in the neuropil and SPNs of striosomes, especially in the caudate nucleus and dorsomedial putamen, relative to the matrix neuropil in these regions. The generally DARPP-32-poor matrix contains scattered DARPP-32-positive cells. DARPP-32 cell bodies in both compartments proved negative for conventional intraneuronal markers. These findings raise the potential for specialized DARPP-32 expression in the human striosomal system and in a set of DARPP-32-positive neurons in the matrix. If DARPP-32 immunohistochemical positivity predicts differential functional DARPP-32 activity, then the distributions demonstrated here could render striosomes and dispersed matrix cells susceptible to differential signaling through cAMP and other signaling systems in health and disease. DARPP-32 is highly concentrated in cells and neuropil of striosomes in post-mortem human brain tissue, particularly in the dorsal caudate nucleus. Scattered DARPP-32-positive cells are found in the human striatal matrix. Calbindin and DARPP-32 do not colocalize within every spiny projection neuron in the dorsal human caudate nucleus.
Collapse
Affiliation(s)
- Christine J Arasaratnam
- Department of Anatomy and Medical Imaging, Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Jennifer J Song
- Department of Anatomy and Medical Imaging, Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Tomoko Yoshida
- Department of Brain and Cognitive Sciences, McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Maurice A Curtis
- Department of Anatomy and Medical Imaging, Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Ann M Graybiel
- Department of Brain and Cognitive Sciences, McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Richard L M Faull
- Department of Anatomy and Medical Imaging, Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Henry J Waldvogel
- Department of Anatomy and Medical Imaging, Centre for Brain Research, University of Auckland, Auckland, New Zealand
| |
Collapse
|
13
|
Domain and cell type-specific immunolocalisation of voltage-gated potassium channels in the mouse striatum. J Chem Neuroanat 2023; 128:102233. [PMID: 36640913 DOI: 10.1016/j.jchemneu.2023.102233] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/09/2023] [Accepted: 01/09/2023] [Indexed: 01/13/2023]
Abstract
Diverse classes of voltage-gated potassium channels (Kv) are integral to the variety of electrical activity patterns that distinguish different classes of neurons in the brain. A feature of their heterogenous expression patterns is the highly precise manner in which specific cell types target their location within functionally specialised sub-cellular domains. Although Kv expression profiles in cortical brain regions are widely reported, their immunolocalisation in sub-cortical areas such as the striatum, and in associated diseases such as Parkinson's disease (PD), remain less well described. Therefore, the broad aims of this study were to provide a high resolution immunolocalisation analysis of various Kv subtypes within the mouse striatum and assess their potential plasticity in a model of PD. Immunohistochemistry and confocal microscopy revealed that immunoreactivity for Kv1.1, 1.2 and 1.4 overlapped to varying degrees with excitatory and inhibitory axonal marker proteins suggesting these Kv subtypes are targeted to axons innervating striatal medium spiny neurons (MSNs). Immunoreactivity for Kv1.3 strongly overlapped with signal for mitochondrial marker proteins in MSN somata and dendrites. Kv1.5 immunoreactivity was expressed in parvalbumin-immunopositive neurons whereas Kv1.6 was located in cells immunopositive for microglia. Signal for Kv2.1 was concentrated on the somatic and proximal dendritic plasma membrane of MSNs, whilst immunoreactivity for Kv4.2 was targeted to their distal dendritic regions. Finally, striatal Kv2.1 expression, at both the mRNA and protein levels, was decreased in alpha-synuclein overexpressing mice, yet increased in alpha-synuclein knockout mice, compared to wild-type counterparts. The data indicate a variety of Kv expression patterns that are distinctive to the striatum and susceptible to pathology that mirrors PD. Furthermore, these findings advance our understanding of the molecular diversity of various striatal cell types, and potentially have implications for the homeostatic changes of MSN excitability during associated medical conditions such as PD.
Collapse
|
14
|
Sapp E, Boudi A, Reid SJ, Trombetta BA, Kivisäkk P, Taghian T, Arnold SE, Howland D, Gray-Edwards H, Kegel-Gleason KB, DiFiglia M. Levels of Synaptic Proteins in Brain and Neurofilament Light Chain in Cerebrospinal Fluid and Plasma of OVT73 Huntington's Disease Sheep Support a Prodromal Disease State. J Huntingtons Dis 2023; 12:201-213. [PMID: 37661892 DOI: 10.3233/jhd-230590] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
BACKGROUND Synaptic changes occur early in patients with Huntington's disease (HD) and in mouse models of HD. An analysis of synaptic changes in HD transgenic sheep (OVT73) is fitting since they have been shown to have some phenotypes. They also have larger brains, longer lifespan, and greater motor and cognitive capacities more aligned with humans, and can provide abundant biofluids for in vivo monitoring of therapeutic interventions. OBJECTIVE The objective of this study was to determine if there were differences between 5- and 10-year-old OVT73 and wild-type (WT) sheep in levels of synaptic proteins in brain and in neurofilament light chain (NfL) in cerebrospinal fluid (CSF) and plasma. METHODS Mutant huntingtin (mHTT) and other proteins were measured by western blot assay in synaptosomes prepared from caudate, motor, and piriform cortex in 5-year-old and caudate, putamen, motor; and piriform cortex in 10-year-old WT and OVT73 sheep. Levels of NfL, a biomarker for neuronal damage increased in many neurological disorders including HD, were examined in CSF and plasma samples from 10-year-old WT and OVT73 sheep using the Simoa NfL Advantage kit. RESULTS Western blot analysis showed mHTT protein expression in synaptosomes from OVT73 sheep was 23% of endogenous sheep HTT levels at both ages. Significant changes were detected in brain levels of PDE10A, SCN4B, DARPP32, calmodulin, SNAP25, PSD95, VGLUT 1, VAMP1, and Na+/K+-ATPase, which depended on age and brain region. There was no difference in NfL levels in CSF and plasma in OVT73 sheep compared to age-matched WT sheep. CONCLUSIONS These results show that synaptic changes occur in brain of 5- and 10-year-old OVT73 sheep, but levels of NfL in biofluids are unaffected. Altogether, the data support a prodromal disease state in OVT73 sheep that involves the caudate, putamen and cortex.
Collapse
Affiliation(s)
- Ellen Sapp
- Department of Neurology, Mass General Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Adel Boudi
- Department of Neurology, Mass General Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Suzanne J Reid
- Centre for Brain Research, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Bianca A Trombetta
- Department of Neurology, Alzheimer's Clinical and Translational Research Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Pia Kivisäkk
- Department of Neurology, Alzheimer's Clinical and Translational Research Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Toloo Taghian
- Department of Radiology and Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Steven E Arnold
- Department of Neurology, Alzheimer's Clinical and Translational Research Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Heather Gray-Edwards
- Department of Radiology and Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Kimberly B Kegel-Gleason
- Department of Neurology, Mass General Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Marian DiFiglia
- Department of Neurology, Mass General Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
15
|
Heck N, Santos MD. Dendritic Spines in Learning and Memory: From First Discoveries to Current Insights. ADVANCES IN NEUROBIOLOGY 2023; 34:311-348. [PMID: 37962799 DOI: 10.1007/978-3-031-36159-3_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
The central nervous system is composed of neural ensembles, and their activity patterns are neural correlates of cognitive functions. Those ensembles are networks of neurons connected to each other by synapses. Most neurons integrate synaptic signal through a remarkable subcellular structure called spine. Dendritic spines are protrusions whose diverse shapes make them appear as a specific neuronal compartment, and they have been the focus of studies for more than a century. Soon after their first description by Ramón y Cajal, it has been hypothesized that spine morphological changes could modify neuronal connectivity and sustain cognitive abilities. Later studies demonstrated that changes in spine density and morphology occurred in experience-dependent plasticity during development, and in clinical cases of mental retardation. This gave ground for the assumption that dendritic spines are the particular locus of cerebral plasticity. With the discovery of synaptic long-term potentiation, a research program emerged with the aim to establish whether dendritic spine plasticity could explain learning and memory. The development of live imaging methods revealed on the one hand that dendritic spine remodeling is compatible with learning process and, on the other hand, that their long-term stability is compatible with lifelong memories. Furthermore, the study of the mechanisms of spine growth and maintenance shed new light on the rules of plasticity. In behavioral paradigms of memory, spine formation or elimination and morphological changes were found to correlate with learning. In a last critical step, recent experiments have provided evidence that dendritic spines play a causal role in learning and memory.
Collapse
Affiliation(s)
- Nicolas Heck
- Laboratory Neurosciences Paris Seine, Sorbonne Université, Paris, France.
| | - Marc Dos Santos
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
16
|
Peng JY, Shen KL, Fan XJ, Qi ZX, Huang HW, Jiang JL, Lu JH, Wang XQ, Fang XX, Yuan WR, Deng QX, Chen S, Chen L, Zhuang QX. Receptor and Ionic Mechanism of Histamine on Mouse Dorsolateral Striatal Neurons. Mol Neurobiol 2023; 60:183-202. [PMID: 36245064 DOI: 10.1007/s12035-022-03076-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 10/08/2022] [Indexed: 12/30/2022]
Abstract
The dorsolateral striatum (DLS) is the critical neural substrate that plays a role in motor control and motor learning. Our past study revealed a direct histaminergic projection from the tuberomammillary nucleus (TMN) of the hypothalamus to the rat striatum. However, the afferent of histaminergic fibers in the mouse DLS, the effect of histamine on DLS neurons, and the underlying receptor and ionic mechanisms remain unclear. Here, we demonstrated a direct histaminergic innervation from the TMN in the mouse DLS, and histamine excited both the direct-pathway spiny projection neurons (d-SPNs) and the indirect-pathway spiny projection neurons (i-SPNs) of DLS via activation of postsynaptic H1R and H2R, albeit activation of presynaptic H3R suppressed neuronal activity by inhibiting glutamatergic synaptic transmission on d-SPNs and i-SPNs in DLS. Moreover, sodium-calcium exchanger 3 (NCX3), potassium-leak channels linked to H1R, and hyperpolarization-activated cyclic nucleotide-gated channel 2 (HCN2) coupled to H2R co-mediated the excitatory effect induced by histamine on d-SPNs and i-SPNs in DLS. These results demonstrated the pre- and postsynaptic receptors and their downstream multiple ionic mechanisms underlying the inhibitory and excitatory effects of histamine on d-SPNs and i-SPNs in DLS, suggesting a potential modulatory effect of the central histaminergic system on the DLS as well as its related motor control and motor learning.
Collapse
Affiliation(s)
- Jian-Ya Peng
- Department of Physiology, School of Medicine, and Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Kang-Li Shen
- Department of Physiology, School of Medicine, and Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Xiu-Juan Fan
- Department of Physiology, School of Medicine, and Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Zeng-Xin Qi
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200030, China.,National Center for Neurological Disorders, Shanghai, 200030, China.,Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, 200030, China
| | - Hui-Wei Huang
- Department of Physiology, School of Medicine, and Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Jian-Lan Jiang
- Department of Physiology, School of Medicine, and Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Jian-Hua Lu
- Department of Physiology, School of Medicine, and Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Xiao-Qin Wang
- Department of Physiology, School of Medicine, and Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Xiao-Xia Fang
- Department of Physiology, School of Medicine, and Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Wang-Rui Yuan
- Department of Physiology, School of Medicine, and Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Qiao-Xuan Deng
- Department of Physiology, School of Medicine, and Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Shu Chen
- Department of Physiology, School of Medicine, and Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Liang Chen
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200030, China. .,National Center for Neurological Disorders, Shanghai, 200030, China. .,Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, 200030, China. .,State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Basic Medical Sciences and Institutes of Brain Science, Fudan University, Shanghai, 200030, China.
| | - Qian-Xing Zhuang
- Department of Physiology, School of Medicine, and Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
17
|
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: 8] [Impact Index Per Article: 4.0] [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
|
18
|
Vertes RP, Linley SB, Rojas AKP. Structural and functional organization of the midline and intralaminar nuclei of the thalamus. Front Behav Neurosci 2022; 16:964644. [PMID: 36082310 PMCID: PMC9445584 DOI: 10.3389/fnbeh.2022.964644] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 07/07/2022] [Indexed: 12/03/2022] Open
Abstract
The midline and intralaminar nuclei of the thalamus form a major part of the "limbic thalamus;" that is, thalamic structures anatomically and functionally linked with the limbic forebrain. The midline nuclei consist of the paraventricular (PV) and paratenial nuclei, dorsally and the rhomboid and nucleus reuniens (RE), ventrally. The rostral intralaminar nuclei (ILt) consist of the central medial (CM), paracentral (PC) and central lateral (CL) nuclei. We presently concentrate on RE, PV, CM and CL nuclei of the thalamus. The nucleus reuniens receives a diverse array of input from limbic-related sites, and predominantly projects to the hippocampus and to "limbic" cortices. The RE participates in various cognitive functions including spatial working memory, executive functions (attention, behavioral flexibility) and affect/fear behavior. The PV receives significant limbic-related afferents, particularly the hypothalamus, and mainly distributes to "affective" structures of the forebrain including the bed nucleus of stria terminalis, nucleus accumbens and the amygdala. Accordingly, PV serves a critical role in "motivated behaviors" such as arousal, feeding/consummatory behavior and drug addiction. The rostral ILt receives both limbic and sensorimotor-related input and distributes widely over limbic and motor regions of the frontal cortex-and throughout the dorsal striatum. The intralaminar thalamus is critical for maintaining consciousness and directly participates in various sensorimotor functions (visuospatial or reaction time tasks) and cognitive tasks involving striatal-cortical interactions. As discussed herein, while each of the midline and intralaminar nuclei are anatomically and functionally distinct, they collectively serve a vital role in several affective, cognitive and executive behaviors - as major components of a brainstem-diencephalic-thalamocortical circuitry.
Collapse
Affiliation(s)
- Robert P. Vertes
- Center for Complex Systems and Brain Sciences, Florida Atlantic University, Boca Raton, FL, United States
- Department of Psychology, Florida Atlantic University, Boca Raton, FL, United States
| | - Stephanie B. Linley
- Center for Complex Systems and Brain Sciences, Florida Atlantic University, Boca Raton, FL, United States
- Department of Psychology, Florida Atlantic University, Boca Raton, FL, United States
- Department of Psychological Science, University of North Georgia, Dahlonega, GA, United States
| | - Amanda K. P. Rojas
- Center for Complex Systems and Brain Sciences, Florida Atlantic University, Boca Raton, FL, United States
| |
Collapse
|
19
|
Li H, Datunashvili M, Reyes RC, Voglmaier SM. Inositol hexakisphosphate kinases differentially regulate trafficking of vesicular glutamate transporters 1 and 2. Front Cell Neurosci 2022; 16:926794. [PMID: 35936490 PMCID: PMC9355605 DOI: 10.3389/fncel.2022.926794] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 06/27/2022] [Indexed: 11/16/2022] Open
Abstract
Inositol pyrophosphates have been implicated in cellular signaling and membrane trafficking, including synaptic vesicle (SV) recycling. Inositol hexakisphosphate kinases (IP6Ks) and their product, diphosphoinositol pentakisphosphate (PP-IP5 or IP7), directly and indirectly regulate proteins important in vesicle recycling by the activity-dependent bulk endocytosis pathway (ADBE). In the present study, we show that two isoforms, IP6K1 and IP6K3, are expressed in axons. The role of the kinases in SV recycling are investigated using pharmacologic inhibition, shRNA knockdown, and IP6K1 and IP6K3 knockout mice. Live-cell imaging experiments use optical reporters of SV recycling based on vesicular glutamate transporter isoforms, VGLUT1- and VGLUT2-pHluorins (pH), which recycle differently. VGLUT1-pH recycles by classical AP-2 dependent endocytosis under moderate stimulation conditions, while VGLUT2-pH recycles using AP-1 and AP-3 adaptor proteins as well. Using a short stimulus to release the readily releasable pool (RRP), we show that IP6K1 KO increases exocytosis of both VGLUT1-and VGLUT2-pH, while IP6K3 KO decreases the amount of both transporters in the RRP. In electrophysiological experiments we measure glutamate signaling with short stimuli and under the intense stimulation conditions that trigger bulk endocytosis. IP6K1 KO increases synaptic facilitation and IP6K3 KO decreases facilitation compared to wild type in CA1 hippocampal Schaffer collateral synapses. After intense stimulation, the rate of endocytosis of VGLUT2-pH, but not VGLUT1-pH, is increased by knockout, knockdown, and pharmacologic inhibition of IP6Ks. Thus IP6Ks differentially affect the endocytosis of two SV protein cargos that use different endocytic pathways. However, while IP6K1 KO and IP6K3 KO exert similar effects on endocytosis after stimulation, the isoforms exert different effects on exocytosis earlier in the stimulus and on the early phase of glutamate release. Taken together, the data indicate a role for IP6Ks both in exocytosis early in the stimulation period and in endocytosis, particularly under conditions that may utilize AP-1/3 adaptors.
Collapse
|
20
|
Oprisan SA, Novo D, Buhusi M, Buhusi CV. Resource Allocation in the Noise-Free Striatal Beat Frequency Model of Interval Timing. TIMING & TIME PERCEPTION 2022; 11:103-123. [PMID: 37065683 PMCID: PMC10103836 DOI: 10.1163/22134468-bja10056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Abstract
The Striatal Beat Frequency (SBF) model of interval timing uses many neural oscillators, presumably located in the frontal cortex (FC), to produce beats at a specific criterion time Tc. The coincidence detection produces the beats in the basal ganglia spiny neurons by comparing the current state of the FC neural oscillators against the long-term memory values stored at reinforcement time Tc. The neurobiologically realistic SBF model has been previously used for producing precise and scalar timing in the presence of noise. Here we simplified the SBF model to gain insight into the resource allocation problem in interval timing networks. Specifically, we used a noise-free SBF model to explore the lower limits of the number of neural oscillators required for producing accurate timing. Using abstract sine-wave neural oscillators in the SBF-sin model, we found that the lower limit of the number of oscillators needed is proportional to the criterion time Tc and the frequency span (fmax − fmin) of the FC neural oscillators. Using biophysically realistic Morris–Lecar model neurons in the SBF-ML model, the lower bound increased by one to two orders of magnitude compared to the SBF-sin model.
Collapse
Affiliation(s)
- Sorinel A. Oprisan
- Department of Physics and Astronomy, College of Charleston, Charleston, SC 29424, USA
| | - Dereck Novo
- Department of Physics and Astronomy, College of Charleston, Charleston, SC 29424, USA
| | - Mona Buhusi
- Department of Psychology, Utah State University, Logan, UT 84322, USA
| | - Catalin V. Buhusi
- Department of Psychology, Utah State University, Logan, UT 84322, USA
| |
Collapse
|
21
|
Mair RG, Francoeur MJ, Krell EM, Gibson BM. Where Actions Meet Outcomes: Medial Prefrontal Cortex, Central Thalamus, and the Basal Ganglia. Front Behav Neurosci 2022; 16:928610. [PMID: 35864847 PMCID: PMC9294389 DOI: 10.3389/fnbeh.2022.928610] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/14/2022] [Indexed: 11/17/2022] Open
Abstract
Medial prefrontal cortex (mPFC) interacts with distributed networks that give rise to goal-directed behavior through afferent and efferent connections with multiple thalamic nuclei and recurrent basal ganglia-thalamocortical circuits. Recent studies have revealed individual roles for different thalamic nuclei: mediodorsal (MD) regulation of signaling properties in mPFC neurons, intralaminar control of cortico-basal ganglia networks, ventral medial facilitation of integrative motor function, and hippocampal functions supported by ventral midline and anterior nuclei. Large scale mapping studies have identified functionally distinct cortico-basal ganglia-thalamocortical subnetworks that provide a structural basis for understanding information processing and functional heterogeneity within the basal ganglia. Behavioral analyses comparing functional deficits produced by lesions or inactivation of specific thalamic nuclei or subregions of mPFC or the basal ganglia have elucidated the interdependent roles of these areas in adaptive goal-directed behavior. Electrophysiological recordings of mPFC neurons in rats performing delayed non-matching-to position (DNMTP) and other complex decision making tasks have revealed populations of neurons with activity related to actions and outcomes that underlie these behaviors. These include responses related to motor preparation, instrumental actions, movement, anticipation and delivery of action outcomes, memory delay, and spatial context. Comparison of results for mPFC, MD, and ventral pallidum (VP) suggest critical roles for mPFC in prospective processes that precede actions, MD for reinforcing task-relevant responses in mPFC, and VP for providing feedback about action outcomes. Synthesis of electrophysiological and behavioral results indicates that different networks connecting mPFC with thalamus and the basal ganglia are organized to support distinct functions that allow organisms to act efficiently to obtain intended outcomes.
Collapse
Affiliation(s)
- Robert G. Mair
- Department of Psychology, The University of New Hampshire, Durham, NH, United States
| | - Miranda J. Francoeur
- Neural Engineering and Translation Labs, University of California, San Diego, San Diego, CA, United States
| | - Erin M. Krell
- Department of Psychology, The University of New Hampshire, Durham, NH, United States
| | - Brett M. Gibson
- Department of Psychology, The University of New Hampshire, Durham, NH, United States
| |
Collapse
|
22
|
Paget-Blanc V, Pfeffer ME, Pronot M, Lapios P, Angelo MF, Walle R, Cordelières FP, Levet F, Claverol S, Lacomme S, Petrel M, Martin C, Pitard V, De Smedt Peyrusse V, Biederer T, Perrais D, Trifilieff P, Herzog E. A synaptomic analysis reveals dopamine hub synapses in the mouse striatum. Nat Commun 2022; 13:3102. [PMID: 35660742 PMCID: PMC9166739 DOI: 10.1038/s41467-022-30776-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 05/18/2022] [Indexed: 11/13/2022] Open
Abstract
Dopamine transmission is involved in reward processing and motor control, and its impairment plays a central role in numerous neurological disorders. Despite its strong pathophysiological relevance, the molecular and structural organization of the dopaminergic synapse remains to be established. Here, we used targeted labelling and fluorescence activated sorting to purify striatal dopaminergic synaptosomes. We provide the proteome of dopaminergic synapses with 57 proteins specifically enriched. Beyond canonical markers of dopamine neurotransmission such as dopamine biosynthetic enzymes and cognate receptors, we validated 6 proteins not previously described as enriched. Moreover, our data reveal the adhesion of dopaminergic synapses to glutamatergic, GABAergic or cholinergic synapses in structures we named “dopamine hub synapses”. At glutamatergic synapses, pre- and postsynaptic markers are significantly increased upon association with dopamine synapses. Dopamine hub synapses may thus support local dopaminergic signalling, complementing volume transmission thought to be the major mechanism by which monoamines modulate network activity. The neurotransmitter dopamine is an important regulator of brain function. Here the authors describe “dopamine hub synapses”, where dopamine transmission may act in synergy with other neurotransmitters.
Collapse
Affiliation(s)
- Vincent Paget-Blanc
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000, Bordeaux, France
| | - Marlene E Pfeffer
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000, Bordeaux, France
| | - Marie Pronot
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000, Bordeaux, France
| | - Paul Lapios
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000, Bordeaux, France
| | - Maria-Florencia Angelo
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000, Bordeaux, France
| | - Roman Walle
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - Fabrice P Cordelières
- Univ. Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UAR 3420, US 4, F-33000, Bordeaux, France
| | - Florian Levet
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000, Bordeaux, France.,Univ. Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UAR 3420, US 4, F-33000, Bordeaux, France
| | | | - Sabrina Lacomme
- Univ. Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UAR 3420, US 4, F-33000, Bordeaux, France
| | - Mélina Petrel
- Univ. Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UAR 3420, US 4, F-33000, Bordeaux, France
| | - Christelle Martin
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000, Bordeaux, France
| | - Vincent Pitard
- UB'FACSility CNRS UMS 3427, INSERM US 005, Univ. Bordeaux, F-33000, Bordeaux, France
| | | | - Thomas Biederer
- Department of Neurology, Yale School of Medicine, New Haven, CT, 06511, USA
| | - David Perrais
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000, Bordeaux, France
| | - Pierre Trifilieff
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - Etienne Herzog
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000, Bordeaux, France.
| |
Collapse
|
23
|
Allichon MC, Ortiz V, Pousinha P, Andrianarivelo A, Petitbon A, Heck N, Trifilieff P, Barik J, Vanhoutte P. Cell-Type-Specific Adaptions in Striatal Medium-Sized Spiny Neurons and Their Roles in Behavioral Responses to Drugs of Abuse. Front Synaptic Neurosci 2022; 13:799274. [PMID: 34970134 PMCID: PMC8712310 DOI: 10.3389/fnsyn.2021.799274] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 11/26/2021] [Indexed: 12/21/2022] Open
Abstract
Drug addiction is defined as a compulsive pattern of drug-seeking- and taking- behavior, with recurrent episodes of abstinence and relapse, and a loss of control despite negative consequences. Addictive drugs promote reinforcement by increasing dopamine in the mesocorticolimbic system, which alters excitatory glutamate transmission within the reward circuitry, thereby hijacking reward processing. Within the reward circuitry, the striatum is a key target structure of drugs of abuse since it is at the crossroad of converging glutamate inputs from limbic, thalamic and cortical regions, encoding components of drug-associated stimuli and environment, and dopamine that mediates reward prediction error and incentive values. These signals are integrated by medium-sized spiny neurons (MSN), which receive glutamate and dopamine axons converging onto their dendritic spines. MSN primarily form two mostly distinct populations based on the expression of either DA-D1 (D1R) or DA-D2 (D2R) receptors. While a classical view is that the two MSN populations act in parallel, playing antagonistic functional roles, the picture seems much more complex. Herein, we review recent studies, based on the use of cell-type-specific manipulations, demonstrating that dopamine differentially modulates dendritic spine density and synapse formation, as well as glutamate transmission, at specific inputs projecting onto D1R-MSN and D2R-MSN to shape persistent pathological behavioral in response to drugs of abuse. We also discuss the identification of distinct molecular events underlying the detrimental interplay between dopamine and glutamate signaling in D1R-MSN and D2R-MSN and highlight the relevance of such cell-type-specific molecular studies for the development of innovative strategies with potential therapeutic value for addiction. Because drug addiction is highly prevalent in patients with other psychiatric disorders when compared to the general population, we last discuss the hypothesis that shared cellular and molecular adaptations within common circuits could explain the co-occurrence of addiction and depression. We will therefore conclude this review by examining how the nucleus accumbens (NAc) could constitute a key interface between addiction and depression.
Collapse
Affiliation(s)
- Marie-Charlotte Allichon
- CNRS, UMR 8246, Neuroscience Paris Seine, Paris, France.,INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | - Vanesa Ortiz
- Université Côte d'Azur, Nice, France.,Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR 7275, Valbonne, France
| | - Paula Pousinha
- Université Côte d'Azur, Nice, France.,Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR 7275, Valbonne, France
| | - Andry Andrianarivelo
- CNRS, UMR 8246, Neuroscience Paris Seine, Paris, France.,INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | - Anna Petitbon
- Université Bordeaux, INRAE, Bordeaux INP, NutriNeuro, Bordeaux, France
| | - Nicolas Heck
- CNRS, UMR 8246, Neuroscience Paris Seine, Paris, France.,INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | - Pierre Trifilieff
- Université Bordeaux, INRAE, Bordeaux INP, NutriNeuro, Bordeaux, France
| | - Jacques Barik
- Université Côte d'Azur, Nice, France.,Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR 7275, Valbonne, France
| | - Peter Vanhoutte
- CNRS, UMR 8246, Neuroscience Paris Seine, Paris, France.,INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| |
Collapse
|
24
|
Barry J, Bui MTN, Levine MS, Cepeda C. Synaptic pathology in Huntington's disease: Beyond the corticostriatal pathway. Neurobiol Dis 2022; 162:105574. [PMID: 34848336 PMCID: PMC9328779 DOI: 10.1016/j.nbd.2021.105574] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/22/2021] [Accepted: 11/26/2021] [Indexed: 12/12/2022] Open
Abstract
Huntington's disease (HD) is a heritable, fatal neurodegenerative disorder caused by a mutation in the Huntingtin gene. It is characterized by chorea, as well as cognitive and psychiatric symptoms. Histopathologically, there is a massive loss of striatal projection neurons and less but significant loss in other areas throughout the cortico-basal ganglia-thalamocortical (CBGTC) loop. The mutant huntingtin protein has been implicated in numerous functions, including an important role in synaptic transmission. Most studies on anatomical and physiological alterations in HD have focused on striatum and cerebral cortex. However, based on recent CBGTC projectome evidence, the need to study other pathways has become increasingly clear. In this review, we examine the current status of our knowledge of morphological and electrophysiological alterations of those pathways in animal models of HD. Based on recent studies, there is accumulating evidence that synaptic disconnection, particularly along excitatory pathways, is pervasive and almost universal in HD, thus supporting a critical role of the huntingtin protein in synaptic transmission.
Collapse
Affiliation(s)
- Joshua Barry
- IDDRC, Jane and Terry Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Minh T N Bui
- IDDRC, Jane and Terry Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Michael S Levine
- IDDRC, Jane and Terry Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Carlos Cepeda
- IDDRC, Jane and Terry Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
25
|
Liu J, Dai R, Damiescu R, Efferth T, Lee DYW. Role of Levo-tetrahydropalmatine and its metabolites for management of chronic pain and opioid use disorders. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 90:153594. [PMID: 34144869 DOI: 10.1016/j.phymed.2021.153594] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/18/2021] [Accepted: 05/07/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Opioids have been prescribed to reduce suffering from pain and to enhance quality of life. Due to the addictive potential and the lack of other effective alternatives to treat severe acute and chronic pains, opioids remain a serious public health issue. While, opioids directly influence the drug-seeking behavior, tolerance and withdrawal processes, through neuroadaptation, the brain's endogenous opioid system also adapts in the presence of chronic pain and could contribute to the difficulty of treatment. Despite the seemingly obvious interaction between the presence of pain and opioid-abuse, little is known about the underlying mechanisms in the brain. PURPOSE To review the current understanding of the interaction mechanisms of neurotransmitter circuitries in pain modulation and reward in the brain and the effects of L-tetrahydropalmatine (L-THP) and its metabolites in pain management and opioid use disorder and gain a better insight on the pharmacological profile and in vivo effects of L-THP and its metabolites. METHOD A detailed literature search on available (preclinical and clinical) studies about the effects of L-THP and its metabolites against drug addiction and chronic pain has been performed. The data was collected using various search engines such as PubMed, ScienceDirect, Google scholar and articles in English up to December 2020 were included in this review. RESULTS L-THP and its metabolites demonstrated analgesic and anti-addiction effects. Due to their dual pharmacological properties (D1 partial agonist and D2 antagonist) these compounds could be used as molecular tools to provide a better understanding of the interactions between pain and addiction. CONCLUSION The available data confirms the potential of L-THP and its metabolites to treat both chronic pain and drug addiction. However, further clinical trials are needed to establish safety and efficacy.
Collapse
Affiliation(s)
- Jing Liu
- Bio-Organic and Natural Products Laboratory, McLean Hospital, Harvard Medical School, 115 Mill Street, Belmont, MA 02478, USA
| | - Ronghua Dai
- Bio-Organic and Natural Products Laboratory, McLean Hospital, Harvard Medical School, 115 Mill Street, Belmont, MA 02478, USA; School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Roxana Damiescu
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, 55128 Mainz, Germany
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, 55128 Mainz, Germany
| | - David Y W Lee
- Bio-Organic and Natural Products Laboratory, McLean Hospital, Harvard Medical School, 115 Mill Street, Belmont, MA 02478, USA.
| |
Collapse
|
26
|
Benthall KN, Cording KR, Agopyan-Miu AHCW, Wong CD, Chen EY, Bateup HS. Loss of Tsc1 from striatal direct pathway neurons impairs endocannabinoid-LTD and enhances motor routine learning. Cell Rep 2021; 36:109511. [PMID: 34380034 PMCID: PMC8404511 DOI: 10.1016/j.celrep.2021.109511] [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] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 05/28/2021] [Accepted: 07/21/2021] [Indexed: 02/08/2023] Open
Abstract
Tuberous sclerosis complex (TSC) is a neurodevelopmental disorder that often presents with psychiatric conditions, including autism spectrum disorder (ASD). ASD is characterized by restricted, repetitive, and inflexible behaviors, which may result from abnormal activity in striatal circuits that mediate motor learning and action selection. To test whether altered striatal activity contributes to aberrant motor behaviors in the context of TSC, we conditionally deleted Tsc1 from direct or indirect pathway striatal projection neurons (dSPNs or iSPNs, respectively). We find that dSPN-specific loss of Tsc1 impairs endocannabinoid-mediated long-term depression (eCB-LTD) at cortico-dSPN synapses and strongly enhances corticostriatal synaptic drive, which is not observed in iSPNs. dSPN-Tsc1 KO, but not iSPN-Tsc1 KO, mice show enhanced motor learning, a phenotype observed in several mouse models of ASD. These findings demonstrate that dSPNs are particularly sensitive to Tsc1 loss and suggest that enhanced corticostriatal activation may contribute to altered motor behaviors in TSC.
Collapse
Affiliation(s)
- Katelyn N Benthall
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Katherine R Cording
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | | | - Corinna D Wong
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Emily Y Chen
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Helen S Bateup
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA.
| |
Collapse
|
27
|
De Groote A, de Kerchove d'Exaerde A. Thalamo-Nucleus Accumbens Projections in Motivated Behaviors and Addiction. Front Syst Neurosci 2021; 15:711350. [PMID: 34335197 PMCID: PMC8322971 DOI: 10.3389/fnsys.2021.711350] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 06/22/2021] [Indexed: 12/28/2022] Open
Abstract
The ventral striatum, also called nucleus accumbens (NAc), has long been known to integrate information from cortical, thalamic, midbrain and limbic nuclei to mediate goal-directed behaviors. Until recently thalamic afferents have been overlooked when studying the functions and connectivity of the NAc. However, findings from recent studies have shed light on the importance and roles of precise Thalamus to NAc connections in motivated behaviors and in addiction. In this review, we summarize studies using techniques such as chemo- and optogenetics, electrophysiology and in vivo calcium imaging to elucidate the complex functioning of the thalamo-NAc afferents, with a particular highlight on the projections from the Paraventricular Thalamus (PVT) to the NAc. We will focus on the recent advances in the understanding of the roles of these neuronal connections in motivated behaviors, with a special emphasis on their implications in addiction, from cue-reward association to the mechanisms driving relapse.
Collapse
Affiliation(s)
- Aurélie De Groote
- Laboratory of Neurophysiology, ULB Neuroscience Institute, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Alban de Kerchove d'Exaerde
- Laboratory of Neurophysiology, ULB Neuroscience Institute, Université Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
28
|
Zheng X, Sun L, Liu B, Huang Z, Zhu Y, Chen T, Jia L, Li Y, Lei W. Morphological Study of the Cortical and Thalamic Glutamatergic Synaptic Inputs of Striatal Parvalbumin Interneurons in Rats. Neurochem Res 2021; 46:1659-1673. [PMID: 33770320 DOI: 10.1007/s11064-021-03302-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 02/14/2021] [Accepted: 03/17/2021] [Indexed: 10/21/2022]
Abstract
Parvalbumin-immunoreactive (Parv+) interneurons is an important component of striatal GABAergic microcircuits, which receive excitatory inputs from the cortex and thalamus, and then target striatal projection neurons. The present study aimed to examine ultrastructural synaptic connection features of Parv+ neruons with cortical and thalamic input, and striatal projection neurons by using immuno-electron microscopy (immuno-EM) and immunofluorescence techniques. Our results showed that both Parv+ somas and dendrites received numerous asymmetric synaptic inputs, and Parv+ terminals formed symmetric synapses with Parv- somas, dendrites and spine bases. Most interestingly, spine bases targeted by Parv+ terminals simultaneously received excitatory inputs at their heads. Electrical stimulation of the motor cortex (M1) induced higher proportion of striatal Parv+ neurons express c-Jun than stimulation of the parafascicular nucleus (PFN), and indicated that cortical- and thalamic-inputs differentially modulate Parv+ neurons. Consistent with that, both Parv + soma and dendrites received more VGlut1+ than VGlut2+ terminals. However, the proportion of VGlut1+ terminal targeting onto Parv+ proximal and distal dendrites was not different, but VGlut2+ terminals tended to target Parv+ somas and proximal dendrites than distal dendrites. These functional and morphological results suggested excitatory cortical and thalamic glutamatergic inputs differently modulate Parv+ interneurons, which provided inhibition inputs onto striatal projection neurons. To maintain the balance between the cortex and thalamus onto Parv+ interneurons may be an important therapeutic target for neurological disorders.
Collapse
Affiliation(s)
- Xuefeng Zheng
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, China
| | - Liping Sun
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Bingbing Liu
- Department of Anesthesiology, Guangdong Second Provincial General Hospital, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Ziyun Huang
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Yaofeng Zhu
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Institute of Medicine, College of Medicine, Jishou University, Jishou, China
| | - Tao Chen
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Linju Jia
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Yanmei Li
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Wanlong Lei
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
29
|
Verduzco-Mendoza A, Carrillo-Mora P, Avila-Luna A, Gálvez-Rosas A, Olmos-Hernández A, Mota-Rojas D, Bueno-Nava A. Role of the Dopaminergic System in the Striatum and Its Association With Functional Recovery or Rehabilitation After Brain Injury. Front Neurosci 2021; 15:693404. [PMID: 34248494 PMCID: PMC8264205 DOI: 10.3389/fnins.2021.693404] [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: 04/10/2021] [Accepted: 06/03/2021] [Indexed: 01/06/2023] Open
Abstract
Disabilities are estimated to occur in approximately 2% of survivors of traumatic brain injury (TBI) worldwide, and disability may persist even decades after brain injury. Facilitation or modulation of functional recovery is an important goal of rehabilitation in all patients who survive severe TBI. However, this recovery tends to vary among patients because it is affected by the biological and physical characteristics of the patients; the types, doses, and application regimens of the drugs used; and clinical indications. In clinical practice, diverse dopaminergic drugs with various dosing and application procedures are used for TBI. Previous studies have shown that dopamine (DA) neurotransmission is disrupted following moderate to severe TBI and have reported beneficial effects of drugs that affect the dopaminergic system. However, the mechanisms of action of dopaminergic drugs have not been completely clarified, partly because dopaminergic receptor activation can lead to restoration of the pathway of the corticobasal ganglia after injury in brain structures with high densities of these receptors. This review aims to provide an overview of the functionality of the dopaminergic system in the striatum and its roles in functional recovery or rehabilitation after TBI.
Collapse
Affiliation(s)
- Antonio Verduzco-Mendoza
- Ph.D. Program in Biological and Health Sciences, Universidad Autónoma Metropolitana, Mexico City, Mexico
- Division of Biotechnology-Bioterio and Experimental Surgery, Instituto Nacional de Rehabilitación-Luis Guillermo Ibarra Ibarra, Mexico City, Mexico
| | - Paul Carrillo-Mora
- Division of Neurosciences, Instituto Nacional de Rehabilitación-Luis Guillermo Ibarra Ibarra, Mexico City, Mexico
| | - Alberto Avila-Luna
- Division of Neurosciences, Instituto Nacional de Rehabilitación-Luis Guillermo Ibarra Ibarra, Mexico City, Mexico
| | - Arturo Gálvez-Rosas
- Division of Neurosciences, Instituto Nacional de Rehabilitación-Luis Guillermo Ibarra Ibarra, Mexico City, Mexico
| | - Adriana Olmos-Hernández
- Division of Biotechnology-Bioterio and Experimental Surgery, Instituto Nacional de Rehabilitación-Luis Guillermo Ibarra Ibarra, Mexico City, Mexico
| | - Daniel Mota-Rojas
- Neurophysiology, Behavior and Animal Welfare Assessment, DPAA, Universidad Autónoma Metropolitana, Mexico City, Mexico
| | - Antonio Bueno-Nava
- Division of Neurosciences, Instituto Nacional de Rehabilitación-Luis Guillermo Ibarra Ibarra, Mexico City, Mexico
| |
Collapse
|
30
|
Villalba RM, Behnke JA, Pare JF, Smith Y. Comparative Ultrastructural Analysis of Thalamocortical Innervation of the Primary Motor Cortex and Supplementary Motor Area in Control and MPTP-Treated Parkinsonian Monkeys. Cereb Cortex 2021; 31:3408-3425. [PMID: 33676368 PMCID: PMC8599722 DOI: 10.1093/cercor/bhab020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/29/2020] [Accepted: 01/19/2021] [Indexed: 12/15/2022] Open
Abstract
The synaptic organization of thalamic inputs to motor cortices remains poorly understood in primates. Thus, we compared the regional and synaptic connections of vGluT2-positive thalamocortical glutamatergic terminals in the supplementary motor area (SMA) and the primary motor cortex (M1) between control and MPTP-treated parkinsonian monkeys. In controls, vGluT2-containing fibers and terminal-like profiles invaded layer II-III and Vb of M1 and SMA. A significant reduction of vGluT2 labeling was found in layer Vb, but not in layer II-III, of parkinsonian animals, suggesting a potential thalamic denervation of deep cortical layers in parkinsonism. There was a significant difference in the pattern of synaptic connectivity in layers II-III, but not in layer Vb, between M1 and SMA of control monkeys. However, this difference was abolished in parkinsonian animals. No major difference was found in the proportion of perforated versus macular post-synaptic densities at thalamocortical synapses between control and parkinsonian monkeys in both cortical regions, except for a slight increase in the prevalence of perforated axo-dendritic synapses in the SMA of parkinsonian monkeys. Our findings suggest that disruption of the thalamic innervation of M1 and SMA may underlie pathophysiological changes of the motor thalamocortical loop in the state of parkinsonism.
Collapse
Affiliation(s)
- Rosa M Villalba
- Division of Neuropharmacology and Neurological Diseases, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- UDALL Center for Excellence for Parkinson’s Disease, Emory University, Atlanta, GA 30329, USA
| | - Joseph A Behnke
- Division of Neuropharmacology and Neurological Diseases, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- UDALL Center for Excellence for Parkinson’s Disease, Emory University, Atlanta, GA 30329, USA
| | - Jean-Francois Pare
- Division of Neuropharmacology and Neurological Diseases, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- UDALL Center for Excellence for Parkinson’s Disease, Emory University, Atlanta, GA 30329, USA
| | - Yoland Smith
- Division of Neuropharmacology and Neurological Diseases, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- UDALL Center for Excellence for Parkinson’s Disease, Emory University, Atlanta, GA 30329, USA
- Department of Neurology, School of Medicine, Emory University, Atlanta, GA 30329, USA
| |
Collapse
|
31
|
Davatolhagh MF, Fuccillo MV. Neurexin1⍺ differentially regulates synaptic efficacy within striatal circuits. Cell Rep 2021; 34:108773. [PMID: 33626349 PMCID: PMC8071350 DOI: 10.1016/j.celrep.2021.108773] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 11/18/2020] [Accepted: 01/29/2021] [Indexed: 02/08/2023] Open
Abstract
Mutations in genes essential for synaptic function, such as the presynaptic adhesion molecule Neurexin1α (Nrxn1α), are strongly implicated in neuropsychiatric pathophysiology. As the input nucleus of the basal ganglia, the striatum integrates diverse excitatory projections governing cognitive and motor control, and its impairment may represent a recurrent pathway to disease. Here, we test the functional relevance of Nrxn1α in striatal circuits by employing optogenetic-mediated afferent recruitment of dorsal prefrontal cortical (dPFC) and parafascicular thalamic connections onto dorsomedial striatal (DMS) spiny projection neurons (SPNs). For dPFC-DMS circuits, we find decreased synaptic strength specifically onto indirect pathway SPNs in both Nrxn1α+/- and Nrxn1α-/- mice, driven by reductions in neurotransmitter release. In contrast, thalamic excitatory inputs to DMS exhibit relatively normal excitatory synaptic strength despite changes in synaptic N-methyl-D-aspartate receptor (NMDAR) content. These findings suggest that dysregulation of Nrxn1α modulates striatal function in an input- and target-specific manner.
Collapse
Affiliation(s)
- M Felicia Davatolhagh
- Neuroscience Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marc V Fuccillo
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
32
|
Alegre-Cortés J, Sáez M, Montanari R, Reig R. Medium spiny neurons activity reveals the discrete segregation of mouse dorsal striatum. eLife 2021; 10:e60580. [PMID: 33599609 PMCID: PMC7924950 DOI: 10.7554/elife.60580] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 02/15/2021] [Indexed: 01/08/2023] Open
Abstract
Behavioral studies differentiate the rodent dorsal striatum (DS) into lateral and medial regions; however, anatomical evidence suggests that it is a unified structure. To understand striatal dynamics and basal ganglia functions, it is essential to clarify the circuitry that supports this behavioral-based segregation. Here, we show that the mouse DS is made of two non-overlapping functional circuits divided by a boundary. Combining in vivo optopatch-clamp and extracellular recordings of spontaneous and evoked sensory activity, we demonstrate different coupling of lateral and medial striatum to the cortex together with an independent integration of the spontaneous activity, due to particular corticostriatal connectivity and local attributes of each region. Additionally, we show differences in slow and fast oscillations and in the electrophysiological properties between striatonigral and striatopallidal neurons. In summary, these results demonstrate that the rodent DS is segregated in two neuronal circuits, in homology with the caudate and putamen nuclei of primates.
Collapse
Affiliation(s)
| | - María Sáez
- Instituto de Neurociencias CSIC-UMHSan Juan de AlicanteSpain
| | | | - Ramon Reig
- Instituto de Neurociencias CSIC-UMHSan Juan de AlicanteSpain
| |
Collapse
|
33
|
Kuhlmann N, Wagner Valladolid M, Quesada-Ramírez L, Farrer MJ, Milnerwood AJ. Chronic and Acute Manipulation of Cortical Glutamate Transmission Induces Structural and Synaptic Changes in Co-cultured Striatal Neurons. Front Cell Neurosci 2021; 15:569031. [PMID: 33679324 PMCID: PMC7930618 DOI: 10.3389/fncel.2021.569031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 01/26/2021] [Indexed: 12/13/2022] Open
Abstract
In contrast to the prenatal topographic development of sensory cortices, striatal circuit organization is slow and requires the functional maturation of cortical and thalamic excitatory inputs throughout the first postnatal month. While mechanisms regulating synapse development and plasticity are quite well described at excitatory synapses of glutamatergic neurons in the neocortex, comparatively little is known of how this translates to glutamate synapses onto GABAergic neurons in the striatum. Here we investigate excitatory striatal synapse plasticity in an in vitro system, where glutamate can be studied in isolation from dopamine and other neuromodulators. We examined pre-and post-synaptic structural and functional plasticity in GABAergic striatal spiny projection neurons (SPNs), co-cultured with glutamatergic cortical neurons. After synapse formation, medium-term (24 h) TTX silencing increased the density of filopodia, and modestly decreased dendritic spine density, when assayed at 21 days in vitro (DIV). Spine reductions appeared to require residual spontaneous activation of ionotropic glutamate receptors. Conversely, chronic (14 days) TTX silencing markedly reduced spine density without any observed increase in filopodia density. Time-dependent, biphasic changes to the presynaptic marker Synapsin-1 were also observed, independent of residual spontaneous activity. Acute silencing (3 h) did not affect presynaptic markers or postsynaptic structures. To induce rapid, activity-dependent plasticity in striatal neurons, a chemical NMDA receptor-dependent “long-term potentiation (LTP)” paradigm was employed. Within 30 min, this increased spine and GluA1 cluster densities, and the percentage of spines containing GluA1 clusters, without altering the presynaptic signal. The results demonstrate that the growth and pruning of dendritic protrusions is an active process, requiring glutamate receptor activity in striatal projection neurons. Furthermore, NMDA receptor activation is sufficient to drive glutamatergic structural plasticity in SPNs, in the absence of dopamine or other neuromodulators.
Collapse
Affiliation(s)
- Naila Kuhlmann
- Centre for Applied Neurogenetics (CAN), University of British Columbia, Vancouver, BC, Canada.,Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | | | - Lucía Quesada-Ramírez
- Centre for Applied Neurogenetics (CAN), University of British Columbia, Vancouver, BC, Canada
| | - Matthew J Farrer
- Centre for Applied Neurogenetics (CAN), University of British Columbia, Vancouver, BC, Canada.,McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Austen J Milnerwood
- Centre for Applied Neurogenetics (CAN), University of British Columbia, Vancouver, BC, Canada.,Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| |
Collapse
|
34
|
Aft T, Oprisan SA, Buhusi CV. Is the scalar property of interval timing preserved after hippocampus lesions? J Theor Biol 2021; 516:110605. [PMID: 33508325 PMCID: PMC7980776 DOI: 10.1016/j.jtbi.2021.110605] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 01/19/2021] [Accepted: 01/21/2021] [Indexed: 11/28/2022]
Abstract
Time perception is fundamental for decision-making, adaptation, and survival. In the peak-interval (PI) paradigm, one of the critical features of time perception is its scale invariance, i.e., the error in time estimation increases linearly with the to-be-timed interval. Brain lesions can profoundly alter time perception, but do they also change its scalar property? In particular, hippocampus (HPC) lesions affect the memory of the reinforced durations. Experiments found that ventral hippocampus (vHPC) lesions shift the perceived durations to longer values while dorsal hippocampus (dHPC) lesions produce opposite effects. Here we used our implementation of the Striatal Beat Frequency (SBFML) model with biophysically realistic Morris-Lecar (ML) model neurons and a topological map of HPC memory to predict analytically and verify numerically the effect of HPC lesions on scalar property. We found that scalar property still holds after both vHPC and dHPC lesions in our SBFML-HPC network simulation. Our numerical results show that PI durations are shifted in the correct direction and match the experimental results. In our simulations, the relative peak shift of the behavioral response curve is controlled by two factors: (1) the lesion size, and (2) the cellular-level memory variance of the temporal durations stored in the HPC. The coefficient of variance (CV) of the behavioral response curve remained constant over the tested durations of PI procedure, which suggests that scalar property is not affected by HPC lesions.
Collapse
Affiliation(s)
- Tristan Aft
- Department of Physics and Astronomy, College of Charleston, United States
| | - Sorinel A Oprisan
- Department of Physics and Astronomy, College of Charleston, United States
| | | |
Collapse
|
35
|
Whole-Brain Mapping of Direct Inputs to Dopamine D1 and D2 Receptor-Expressing Medium Spiny Neurons in the Posterior Dorsomedial Striatum. eNeuro 2021; 8:ENEURO.0348-20.2020. [PMID: 33380525 PMCID: PMC7877463 DOI: 10.1523/eneuro.0348-20.2020] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 12/13/2020] [Accepted: 12/19/2020] [Indexed: 12/12/2022] Open
Abstract
The posterior dorsomedial striatum (pDMS) is mainly composed of medium spiny neurons (MSNs) expressing either dopamine D1 receptors (D1Rs) or D2Rs. Activation of these two MSN types produces opposing effects on addictive behaviors. However, it remains unclear whether pDMS D1-MSNs or D2-MSNs receive afferent inputs from different brain regions or whether the extrastriatal afferents express distinct dopamine receptors. To assess whether these afferents also contained D1Rs or D2Rs, we generated double transgenic mice, in which D1R-expressing and D2R-expressing neurons were fluorescently labeled. We used rabies virus-mediated retrograde tracing in these mice to perform whole-brain mapping of direct inputs to D1-MSNs or D2-MSNs in the pDMS. We found that D1-MSNs preferentially received inputs from the secondary motor, secondary visual, and cingulate cortices, whereas D2-MSNs received inputs from the primary motor and primary sensory cortices, and the thalamus. We also discovered that the bed nucleus of the stria terminalis (BNST) and the central nucleus of the amygdala (CeA) contained abundant D2R-expressing, but few D1R-expressing, neurons in a triple transgenic mouse model. Remarkably, although limited D1R or D2R expression was observed in extrastriatal neurons that projected to D1-MSNs or D2-MSNs, we found that cortical structures preferentially contained D1R-expressing neurons that projected to D1-MSNs or D2-MSNs, while the thalamus, substantia nigra pars compacta (SNc), and BNST had more D2R-expressing cells that projected to D2-MSNs. Taken together, these findings provide a foundation for future understanding of the pDMS circuit and its role in action selection and reward-based behaviors.
Collapse
|
36
|
Johansson Y, Silberberg G. The Functional Organization of Cortical and Thalamic Inputs onto Five Types of Striatal Neurons Is Determined by Source and Target Cell Identities. Cell Rep 2020; 30:1178-1194.e3. [PMID: 31995757 PMCID: PMC6990404 DOI: 10.1016/j.celrep.2019.12.095] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 11/28/2019] [Accepted: 12/27/2019] [Indexed: 11/22/2022] Open
Abstract
To understand striatal function, it is essential to know the functional organization of the numerous inputs targeting the diverse population of striatal neurons. Using optogenetics, we activated terminals from ipsi- or contralateral primary somatosensory cortex (S1) or primary motor cortex (M1), or thalamus while obtaining simultaneous whole-cell recordings from pairs or triplets of striatal medium spiny neurons (MSNs) and adjacent interneurons. Ipsilateral corticostriatal projections provided stronger excitation to fast-spiking interneurons (FSIs) than to MSNs and only sparse and weak excitation to low threshold-spiking interneurons (LTSIs) and cholinergic interneurons (ChINs). Projections from contralateral M1 evoked the strongest responses in LTSIs but none in ChINs, whereas thalamus provided the strongest excitation to ChINs but none to LTSIs. In addition, inputs varied in their glutamate receptor composition and their short-term plasticity. Our data revealed a highly selective organization of excitatory striatal afferents, which is determined by both pre- and postsynaptic neuronal identity. Whole-cell recordings are obtained from neighboring striatal neurons of different types FSIs receive the strongest inputs from S1, M1, and thalamic PF LTSIs are primarily excited by contralateral M1 ChINs are primarily excited by PF and receive no input from contralateral M1 and S1
Collapse
Affiliation(s)
- Yvonne Johansson
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Gilad Silberberg
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
37
|
Johnson KA, Voyvodic L, Loewinger GC, Mateo Y, Lovinger DM. Operant self-stimulation of thalamic terminals in the dorsomedial striatum is constrained by metabotropic glutamate receptor 2. Neuropsychopharmacology 2020; 45:1454-1462. [PMID: 31995814 PMCID: PMC7360544 DOI: 10.1038/s41386-020-0626-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 01/10/2020] [Accepted: 01/16/2020] [Indexed: 12/25/2022]
Abstract
Dorsal striatal manipulations including stimulation of dopamine release and activation of medium spiny neurons (MSNs) are sufficient to drive reinforcement-based learning. Glutamatergic innervation of the striatum by the cortex and thalamus is a critical determinant of MSN activity and local regulation of dopamine release. However, the relationship between striatal glutamatergic afferents and behavioral reinforcement is not well understood. We evaluated the reinforcing properties of optogenetic stimulation of thalamostriatal terminals, which are associated with vesicular glutamate transporter 2 (Vglut2) expression, in the dorsomedial striatum (DMS), a region implicated in goal-directed behaviors. In mice expressing channelrhodopsin-2 (ChR2) under control of the Vglut2 promoter, optical stimulation of the DMS reinforced operant lever-pressing behavior. Mice also acquired operant self-stimulation of thalamostriatal terminals when ChR2 expression was virally targeted to the intralaminar thalamus. Stimulation trains that supported operant responding evoked dopamine release in the DMS and excitatory postsynaptic currents in DMS MSNs. Our previous work demonstrated that the presynaptic G protein-coupled receptor metabotropic glutamate receptor 2 (mGlu2) robustly inhibits glutamate and dopamine release induced by activation of thalamostriatal afferents. Thus, we examined the regulation of thalamostriatal self-stimulation by mGlu2. Administration of an mGlu2/3 agonist or an mGlu2-selective positive allosteric modulator reduced self-stimulation. Conversely, blockade of these receptors increased thalamostriatal self-stimulation, suggesting that endogenous activation of these receptors negatively modulates the reinforcing properties of thalamostriatal activity. These findings demonstrate that stimulation of thalamic terminals in the DMS is sufficient to reinforce a self-initiated action, and that thalamostriatal reinforcement is constrained by mGlu2 activation.
Collapse
Affiliation(s)
- Kari A. Johnson
- 0000 0001 2297 5165grid.94365.3dLaboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, US National Institutes of Health, Rockville, MD 20852 USA ,0000 0001 0421 5525grid.265436.0Present Address: Department of Pharmacology, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, C2019, Bethesda, MD 20814 USA
| | - Lucas Voyvodic
- 0000 0001 2297 5165grid.94365.3dLaboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, US National Institutes of Health, Rockville, MD 20852 USA
| | - Gabriel C. Loewinger
- 000000041936754Xgrid.38142.3cDepartment of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115 USA
| | - Yolanda Mateo
- 0000 0001 2297 5165grid.94365.3dLaboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, US National Institutes of Health, Rockville, MD 20852 USA
| | - David M. Lovinger
- 0000 0001 2297 5165grid.94365.3dLaboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, US National Institutes of Health, Rockville, MD 20852 USA
| |
Collapse
|
38
|
Salery M, Trifilieff P, Caboche J, Vanhoutte P. From Signaling Molecules to Circuits and Behaviors: Cell-Type-Specific Adaptations to Psychostimulant Exposure in the Striatum. Biol Psychiatry 2020; 87:944-953. [PMID: 31928716 DOI: 10.1016/j.biopsych.2019.11.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 10/30/2019] [Accepted: 11/01/2019] [Indexed: 12/17/2022]
Abstract
Addiction is characterized by a compulsive pattern of drug seeking and consumption and a high risk of relapse after withdrawal that are thought to result from persistent adaptations within brain reward circuits. Drugs of abuse increase dopamine (DA) concentration in these brain areas, including the striatum, which shapes an abnormal memory trace of drug consumption that virtually highjacks reward processing. Long-term neuronal adaptations of gamma-aminobutyric acidergic striatal projection neurons (SPNs) evoked by drugs of abuse are critical for the development of addiction. These neurons form two mostly segregated populations, depending on the DA receptor they express and their output projections, constituting the so-called direct (D1 receptor) and indirect (D2 receptor) SPN pathways. Both SPN subtypes receive converging glutamate inputs from limbic and cortical regions, encoding contextual and emotional information, together with DA, which mediates reward prediction and incentive values. DA differentially modulates the efficacy of glutamate synapses onto direct and indirect SPN pathways by recruiting distinct striatal signaling pathways, epigenetic and genetic responses likely involved in the transition from casual drug use to addiction. Herein we focus on recent studies that have assessed psychostimulant-induced alterations in a cell-type-specific manner, from remodeling of input projections to the characterization of specific molecular events in each SPN subtype and their impact on long-lasting behavioral adaptations. We discuss recent evidence revealing the complex and concerted action of both SPN populations on drug-induced behavioral responses, as these studies can contribute to the design of future strategies to alleviate specific behavioral components of addiction.
Collapse
Affiliation(s)
- Marine Salery
- Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Pierre Trifilieff
- NutriNeuro, Unité Mixte de Recherche (UMR) 1286, Institut National de la Recherche Agronomique, Bordeaux Institut Polytechnique, University of Bordeaux, Bordeaux, France
| | - Jocelyne Caboche
- Neuroscience Paris Seine, Institut de Biologie Paris-Seine, Sorbonne Université, Faculty of Sciences, Paris, France; Centre National de la Recherche Scientifique, UMR8246, Paris, France; Institut National de la Santé et de la Recherche Médicale, U1130, Paris France.
| | - Peter Vanhoutte
- Neuroscience Paris Seine, Institut de Biologie Paris-Seine, Sorbonne Université, Faculty of Sciences, Paris, France; Centre National de la Recherche Scientifique, UMR8246, Paris, France; Institut National de la Santé et de la Recherche Médicale, U1130, Paris France
| |
Collapse
|
39
|
Mendes A, Vignoud G, Perez S, Perrin E, Touboul J, Venance L. Concurrent Thalamostriatal and Corticostriatal Spike-Timing-Dependent Plasticity and Heterosynaptic Interactions Shape Striatal Plasticity Map. Cereb Cortex 2020; 30:4381-4401. [DOI: 10.1093/cercor/bhaa024] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Abstract
The striatum integrates inputs from the cortex and thalamus, which display concomitant or sequential activity. The striatum assists in forming memory, with acquisition of the behavioral repertoire being associated with corticostriatal (CS) plasticity. The literature has mainly focused on that CS plasticity, and little remains known about thalamostriatal (TS) plasticity rules or CS and TS plasticity interactions. We undertook here the study of these plasticity rules. We found bidirectional Hebbian and anti-Hebbian spike-timing-dependent plasticity (STDP) at the thalamic and cortical inputs, respectively, which were driving concurrent changes at the striatal synapses. Moreover, TS- and CS-STDP induced heterosynaptic plasticity. We developed a calcium-based mathematical model of the coupled TS and CS plasticity, and simulations predict complex changes in the CS and TS plasticity maps depending on the precise cortex–thalamus–striatum engram. These predictions were experimentally validated using triplet-based STDP stimulations, which revealed the significant remodeling of the CS-STDP map upon TS activity, which is notably the induction of the LTD areas in the CS-STDP for specific timing regimes. TS-STDP exerts a greater influence on CS plasticity than CS-STDP on TS plasticity. These findings highlight the major impact of precise timing in cortical and thalamic activity for the memory engram of striatal synapses.
Collapse
Affiliation(s)
- Alexandre Mendes
- Dynamics and Pathophysiology of Neuronal Networks Team, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS UMR7241, INSERM U1050, PSL Research University, Paris, 75005, France
| | - Gaetan Vignoud
- Dynamics and Pathophysiology of Neuronal Networks Team, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS UMR7241, INSERM U1050, PSL Research University, Paris, 75005, France
- Department of Mathematics, Volen National Center for Complex Systems, Brandeis University, Waltham, MA 2454-9110, USA
| | - Sylvie Perez
- Dynamics and Pathophysiology of Neuronal Networks Team, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS UMR7241, INSERM U1050, PSL Research University, Paris, 75005, France
| | - Elodie Perrin
- Dynamics and Pathophysiology of Neuronal Networks Team, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS UMR7241, INSERM U1050, PSL Research University, Paris, 75005, France
| | - Jonathan Touboul
- Department of Mathematics, Volen National Center for Complex Systems, Brandeis University, Waltham, MA 2454-9110, USA
| | - Laurent Venance
- Dynamics and Pathophysiology of Neuronal Networks Team, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS UMR7241, INSERM U1050, PSL Research University, Paris, 75005, France
| |
Collapse
|
40
|
Durante V, de Iure A, Loffredo V, Vaikath N, De Risi M, Paciotti S, Quiroga-Varela A, Chiasserini D, Mellone M, Mazzocchetti P, Calabrese V, Campanelli F, Mechelli A, Di Filippo M, Ghiglieri V, Picconi B, El-Agnaf OM, De Leonibus E, Gardoni F, Tozzi A, Calabresi P. Alpha-synuclein targets GluN2A NMDA receptor subunit causing striatal synaptic dysfunction and visuospatial memory alteration. Brain 2020; 142:1365-1385. [PMID: 30927362 DOI: 10.1093/brain/awz065] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 01/07/2019] [Accepted: 01/25/2019] [Indexed: 01/22/2023] Open
Abstract
Parkinson's disease is a progressive neurodegenerative disorder characterized by altered striatal dopaminergic signalling that leads to motor and cognitive deficits. Parkinson's disease is also characterized by abnormal presence of soluble toxic forms of α-synuclein that, when clustered into Lewy bodies, represents one of the pathological hallmarks of the disease. However, α-synuclein oligomers might also directly affect synaptic transmission and plasticity in Parkinson's disease models. Accordingly, by combining electrophysiological, optogenetic, immunofluorescence, molecular and behavioural analyses, here we report that α-synuclein reduces N-methyl-d-aspartate (NMDA) receptor-mediated synaptic currents and impairs corticostriatal long-term potentiation of striatal spiny projection neurons, of both direct (D1-positive) and indirect (putative D2-positive) pathways. Intrastriatal injections of α-synuclein produce deficits in visuospatial learning associated with reduced function of GluN2A NMDA receptor subunit indicating that this protein selectively targets this subunit both in vitro and ex vivo. Interestingly, this effect is observed in spiny projection neurons activated by optical stimulation of either cortical or thalamic glutamatergic afferents. We also found that treatment of striatal slices with antibodies targeting α-synuclein prevents the α-synuclein-induced loss of long-term potentiation and the reduced synaptic localization of GluN2A NMDA receptor subunit suggesting that this strategy might counteract synaptic dysfunction occurring in Parkinson's disease.
Collapse
Affiliation(s)
- Valentina Durante
- Neurological Clinic, Department of Medicine, Hospital Santa Maria della Misericordia, University of Perugia, Perugia, Italy
| | - Antonio de Iure
- Neurological Clinic, Department of Medicine, Hospital Santa Maria della Misericordia, University of Perugia, Perugia, Italy.,Laboratory of Experimental Neurophysiology, IRCCS San Raffaele Pisana, Rome, Italy
| | - Vittorio Loffredo
- Institute of Cellular Biology and Neurobiology, National Research Council, Monterotondo (Rome), Italy.,PhD Program in Behavioral Neuroscience, Sapienza University of Rome, Italy
| | - Nishant Vaikath
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Maria De Risi
- Telethon Institute of Genetics and Medicine, Telethon Foundation, Pozzuoli (NA), Italy
| | - Silvia Paciotti
- Department of Experimental Medicine, Section of Physiology and Biochemistry, University of Perugia, Perugia, Italy
| | - Ana Quiroga-Varela
- Neurological Clinic, Department of Medicine, Hospital Santa Maria della Misericordia, University of Perugia, Perugia, Italy
| | - Davide Chiasserini
- Neurological Clinic, Department of Medicine, Hospital Santa Maria della Misericordia, University of Perugia, Perugia, Italy
| | - Manuela Mellone
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Italy
| | - Petra Mazzocchetti
- Neurological Clinic, Department of Medicine, Hospital Santa Maria della Misericordia, University of Perugia, Perugia, Italy
| | - Valeria Calabrese
- Neurological Clinic, Department of Medicine, Hospital Santa Maria della Misericordia, University of Perugia, Perugia, Italy.,Laboratory of Experimental Neurophysiology, IRCCS San Raffaele Pisana, Rome, Italy
| | - Federica Campanelli
- Neurological Clinic, Department of Medicine, Hospital Santa Maria della Misericordia, University of Perugia, Perugia, Italy.,Laboratory of Neurophysiology, Santa Lucia Foundation, IRCCS, Rome, Italy
| | - Alessandro Mechelli
- Neurological Clinic, Department of Medicine, Hospital Santa Maria della Misericordia, University of Perugia, Perugia, Italy
| | - Massimiliano Di Filippo
- Neurological Clinic, Department of Medicine, Hospital Santa Maria della Misericordia, University of Perugia, Perugia, Italy
| | - Veronica Ghiglieri
- Laboratory of Neurophysiology, Santa Lucia Foundation, IRCCS, Rome, Italy.,Department of Philosophy, Human, Social and Educational Sciences, University of Perugia, Perugia, Italy
| | - Barbara Picconi
- Laboratory of Experimental Neurophysiology, IRCCS San Raffaele Pisana, Rome, Italy.,University of San Raffaele, Rome, Italy
| | - Omar M El-Agnaf
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Elvira De Leonibus
- Institute of Cellular Biology and Neurobiology, National Research Council, Monterotondo (Rome), Italy.,Telethon Institute of Genetics and Medicine, Telethon Foundation, Pozzuoli (NA), Italy
| | - Fabrizio Gardoni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Italy
| | - Alessandro Tozzi
- Department of Experimental Medicine, Section of Physiology and Biochemistry, University of Perugia, Perugia, Italy.,Laboratory of Neurophysiology, Santa Lucia Foundation, IRCCS, Rome, Italy
| | - Paolo Calabresi
- Neurological Clinic, Department of Medicine, Hospital Santa Maria della Misericordia, University of Perugia, Perugia, Italy.,Laboratory of Neurophysiology, Santa Lucia Foundation, IRCCS, Rome, Italy
| |
Collapse
|
41
|
The prefrontal cortex and the caudate nucleus respond conjointly to methylphenidate (Ritalin). Concomitant behavioral and neuronal recording study. Brain Res Bull 2020; 157:77-89. [PMID: 31987926 DOI: 10.1016/j.brainresbull.2019.10.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/15/2019] [Accepted: 10/23/2019] [Indexed: 01/07/2023]
Abstract
Methylphenidate (MPD) is commonly used to treat attention-deficit hyperactivity disorder (ADHD). Recently, it is being abused for cognitive enhancement and recreation leading to concerns regarding its addictive potential. The prefrontal cortex (PFC) and caudate nucleus (CN) are two of the brain structures involved in the motive/reward circuit most affected by MPD and are also thought to be responsible for ADHD phenomena. This study is unique in that it investigated acute and chronic, dose-response MPD exposure on animals' behavior activity concomitantly with PFC and CN neuronal circuitry in freely behaving adult animals without the interference of anesthesia. Further, it compared acute and chronic MPD action on over 1,000 subcortical and cortical neurons simultaneously, allowing for a more accurate interpretation of drug action on corticostriatal neuronal circuitry. For this experiment, four groups of animals were used: saline (control), 0.6, 2.5, and 10.0 mg/kg MPD following acute and repetitive exposure. The data shows that the same MPD dose elicits behavioral sensitization in some animals and tolerance in others and that the PFC and CN neuronal activity correlates with the animals' behavioral responses to MPD. The expression of sensitization and tolerance are experimental biomarkers indicating that a drug has addictive potential. In general, a greater percentage of CN units responded to both acute and chronic MPD exposure as compared to PFC units. Dose response differences between the PFC and the CN units were observed. The dichotomy that some PFC and CN units responded to the same MPD dose by excitation and other units by attenuation in neuronal firing rate is discussed. In conclusion, to understand the mechanism of action of the drug, it is essential to study, simultaneously, on more than one brain site, the electrophysiological and behavioral effects of acute and chronic drug exposure, as sensitization and tolerance are experimental biomarkers indicating that a drug has addictive potential. The behavioral and neuronal data obtained from this study indicates that chronic MPD exposure results in behavioral and biochemical changes consistent with a substance abuse disorder.
Collapse
|
42
|
Duhne M, Lara‐González E, Laville A, Padilla‐Orozco M, Ávila‐Cascajares F, Arias‐García M, Galarraga E, Bargas J. Activation of parvalbumin‐expressing neurons reconfigures neuronal ensembles in murine striatal microcircuits. Eur J Neurosci 2020; 53:2149-2164. [DOI: 10.1111/ejn.14670] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 12/23/2019] [Accepted: 01/02/2020] [Indexed: 12/30/2022]
Affiliation(s)
- Mariana Duhne
- División Neurociencias Instituto de Fisiología Celular Universidad Nacional Autónoma de México México City Mexico
| | - Esther Lara‐González
- División Neurociencias Instituto de Fisiología Celular Universidad Nacional Autónoma de México México City Mexico
- Facultad de Ciencias Químicas Benemérita Universidad Autónoma de Puebla Puebla Mexico
| | - Antonio Laville
- División Neurociencias Instituto de Fisiología Celular Universidad Nacional Autónoma de México México City Mexico
| | - Montserrat Padilla‐Orozco
- División Neurociencias Instituto de Fisiología Celular Universidad Nacional Autónoma de México México City Mexico
| | - Fatima Ávila‐Cascajares
- División Neurociencias Instituto de Fisiología Celular Universidad Nacional Autónoma de México México City Mexico
| | - Mario Arias‐García
- División Neurociencias Instituto de Fisiología Celular Universidad Nacional Autónoma de México México City Mexico
| | - Elvira Galarraga
- División Neurociencias Instituto de Fisiología Celular Universidad Nacional Autónoma de México México City Mexico
| | - José Bargas
- División Neurociencias Instituto de Fisiología Celular Universidad Nacional Autónoma de México México City Mexico
| |
Collapse
|
43
|
Lieberman OJ, Frier MD, McGuirt AF, Griffey CJ, Rafikian E, Yang M, Yamamoto A, Borgkvist A, Santini E, Sulzer D. Cell-type-specific regulation of neuronal intrinsic excitability by macroautophagy. eLife 2020; 9:e50843. [PMID: 31913125 PMCID: PMC6984822 DOI: 10.7554/elife.50843] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 01/07/2020] [Indexed: 12/28/2022] Open
Abstract
The basal ganglia are a group of subcortical nuclei that contribute to action selection and reinforcement learning. The principal neurons of the striatum, spiny projection neurons of the direct (dSPN) and indirect (iSPN) pathways, maintain low intrinsic excitability, requiring convergent excitatory inputs to fire. Here, we examined the role of autophagy in mouse SPN physiology and animal behavior by generating conditional knockouts of Atg7 in either dSPNs or iSPNs. Loss of autophagy in either SPN population led to changes in motor learning but distinct effects on cellular physiology. dSPNs, but not iSPNs, required autophagy for normal dendritic structure and synaptic input. In contrast, iSPNs, but not dSPNs, were intrinsically hyperexcitable due to reduced function of the inwardly rectifying potassium channel, Kir2. These findings define a novel mechanism by which autophagy regulates neuronal activity: control of intrinsic excitability via the regulation of potassium channel function.
Collapse
Affiliation(s)
- Ori J Lieberman
- Department of PsychiatryColumbia University Vagelos College of Physicians and SurgeonsNew YorkUnited States
| | - Micah D Frier
- Department of PsychiatryColumbia University Vagelos College of Physicians and SurgeonsNew YorkUnited States
| | - Avery F McGuirt
- Department of PsychiatryColumbia University Vagelos College of Physicians and SurgeonsNew YorkUnited States
| | - Christopher J Griffey
- Department of NeurologyColumbia University Vagelos College of Physicians and SurgeonsNew YorkUnited States
| | - Elizabeth Rafikian
- Mouse NeuroBehavior Core, Institute for Genomic MedicineColumbia University Vagelos College of Physicians and SurgeonsNew YorkUnited States
| | - Mu Yang
- Mouse NeuroBehavior Core, Institute for Genomic MedicineColumbia University Vagelos College of Physicians and SurgeonsNew YorkUnited States
| | - Ai Yamamoto
- Department of NeurologyColumbia University Vagelos College of Physicians and SurgeonsNew YorkUnited States
| | | | | | - David Sulzer
- Department of PsychiatryColumbia University Vagelos College of Physicians and SurgeonsNew YorkUnited States
- Department of NeurologyColumbia University Vagelos College of Physicians and SurgeonsNew YorkUnited States
- Department of PharmacologyColumbia University Vagelos College of Physicians and SurgeonsNew YorkUnited States
- Division of Molecular TherapeuticsNew York State Psychiatric InstituteNew YorkUnited States
| |
Collapse
|
44
|
Filipović M, Ketzef M, Reig R, Aertsen A, Silberberg G, Kumar A. Direct pathway neurons in mouse dorsolateral striatum in vivo receive stronger synaptic input than indirect pathway neurons. J Neurophysiol 2019; 122:2294-2303. [DOI: 10.1152/jn.00481.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Striatal projection neurons, the medium spiny neurons (MSNs), play a crucial role in various motor and cognitive functions. MSNs express either D1- or D2-type dopamine receptors and initiate the direct-pathway (dMSNs) or indirect pathways (iMSNs) of the basal ganglia, respectively. dMSNs have been shown to receive more inhibition than iMSNs from intrastriatal sources. Based on these findings, computational modeling of the striatal network has predicted that under healthy conditions dMSNs should receive more total input than iMSNs. To test this prediction, we analyzed in vivo whole cell recordings from dMSNs and iMSNs in healthy and dopamine-depleted (6OHDA) anaesthetized mice. By comparing their membrane potential fluctuations, we found that dMSNs exhibited considerably larger membrane potential fluctuations over a wide frequency range. Furthermore, by comparing the spike-triggered average membrane potentials, we found that dMSNs depolarized toward the spike threshold significantly faster than iMSNs did. Together, these findings (in particular the STA analysis) corroborate the theoretical prediction that direct-pathway MSNs receive stronger total input than indirect-pathway neurons. Finally, we found that dopamine-depleted mice exhibited no difference between the membrane potential fluctuations of dMSNs and iMSNs. These data provide new insights into the question of how the lack of dopamine may lead to behavioral deficits associated with Parkinson’s disease. NEW & NOTEWORTHY The direct and indirect pathways of the basal ganglia originate from the D1- and D2-type dopamine receptor expressing medium spiny neurons (dMSNs and iMSNs). Theoretical results have predicted that dMSNs should receive stronger synaptic input than iMSNs. Using in vivo intracellular membrane potential data, we provide evidence that dMSNs indeed receive stronger input than iMSNs, as has been predicted by the computational model.
Collapse
Affiliation(s)
- Marko Filipović
- Division of Computational Science and Technology, School of Electrical Engineering and Computer Science, KTH Royal Institute of Technology, Stockholm, Sweden
- Bernstein Center Freiburg and Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Maya Ketzef
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Ramon Reig
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernandez, San Juan de Alicante, Spain
| | - Ad Aertsen
- Bernstein Center Freiburg and Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Gilad Silberberg
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Arvind Kumar
- Division of Computational Science and Technology, School of Electrical Engineering and Computer Science, KTH Royal Institute of Technology, Stockholm, Sweden
- Bernstein Center Freiburg and Faculty of Biology, University of Freiburg, Freiburg, Germany
| |
Collapse
|
45
|
Kosillo P, Doig NM, Ahmed KM, Agopyan-Miu AHCW, Wong CD, Conyers L, Threlfell S, Magill PJ, Bateup HS. Tsc1-mTORC1 signaling controls striatal dopamine release and cognitive flexibility. Nat Commun 2019; 10:5426. [PMID: 31780742 PMCID: PMC6882901 DOI: 10.1038/s41467-019-13396-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 11/07/2019] [Indexed: 11/13/2022] Open
Abstract
Tuberous Sclerosis Complex (TSC) is a neurodevelopmental disorder caused by mutations in TSC1 or TSC2, which encode proteins that negatively regulate mTOR complex 1 (mTORC1). TSC is associated with significant cognitive, psychiatric, and behavioral problems, collectively termed TSC-Associated Neuropsychiatric Disorders (TAND), and the cell types responsible for these manifestations are largely unknown. Here we use cell type-specific Tsc1 deletion to test whether dopamine neurons, which modulate cognitive, motivational, and affective behaviors, are involved in TAND. We show that loss of Tsc1 and constitutive activation of mTORC1 in dopamine neurons causes somatodendritic hypertrophy, reduces intrinsic excitability, alters axon terminal structure, and impairs striatal dopamine release. These perturbations lead to a selective deficit in cognitive flexibility, preventable by genetic reduction of the mTOR-binding protein Raptor. Our results establish a critical role for Tsc1-mTORC1 signaling in setting the functional properties of dopamine neurons, and indicate that dopaminergic dysfunction may contribute to cognitive inflexibility in TSC.
Collapse
Affiliation(s)
- Polina Kosillo
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Natalie M Doig
- Medical Research Council Brain Network Dynamics Unit, University of Oxford, Oxford, OX1 3TH, UK
| | - Kamran M Ahmed
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, 94720, USA
| | | | - Corinna D Wong
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Lisa Conyers
- Medical Research Council Brain Network Dynamics Unit, University of Oxford, Oxford, OX1 3TH, UK
| | - Sarah Threlfell
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, OX1 3QX, UK
| | - Peter J Magill
- Medical Research Council Brain Network Dynamics Unit, University of Oxford, Oxford, OX1 3TH, UK
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, OX1 3QX, UK
| | - Helen S Bateup
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, 94720, USA.
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, 94720, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, 94158, USA.
| |
Collapse
|
46
|
Krajeski RN, Macey-Dare A, van Heusden F, Ebrahimjee F, Ellender TJ. Dynamic postnatal development of the cellular and circuit properties of striatal D1 and D2 spiny projection neurons. J Physiol 2019; 597:5265-5293. [PMID: 31531863 PMCID: PMC6900874 DOI: 10.1113/jp278416] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 09/04/2019] [Indexed: 12/15/2022] Open
Abstract
KEY POINTS Imbalances in the activity of the D1-expressing direct pathway and D2-expressing indirect pathway striatal projection neurons (SPNs) are thought to contribute to many basal ganglia disorders, including early-onset neurodevelopmental disorders such as obsessive-compulsive disorder, attention deficit hyperactivity disorder and Tourette's syndrome. This study provides the first detailed quantitative investigation of development of D1 and D2 SPNs, including their cellular properties and connectivity within neural circuits, during the first postnatal weeks. This period is highly dynamic with many properties changing, but it is possible to make three main observations: many aspects of D1 and D2 SPNs progressively mature in parallel; there are notable exceptions when they diverge; and many of the defining properties of mature striatal SPNs and circuits are already established by the first and second postnatal weeks, suggesting guidance through intrinsic developmental programmes. These findings provide an experimental framework for future studies of striatal development in both health and disease. ABSTRACT Many basal ganglia neurodevelopmental disorders are thought to result from imbalances in the activity of the D1-expressing direct pathway and D2-expressing indirect pathway striatal projection neurons (SPNs). Insight into these disorders is reliant on our understanding of normal D1 and D2 SPN development. Here we provide the first detailed study and quantification of the striatal cellular and circuit changes occurring for both D1 and D2 SPNs in the first postnatal weeks using in vitro whole-cell patch-clamp electrophysiology. Characterization of their intrinsic electrophysiological and morphological properties, the excitatory long-range inputs coming from cortex and thalamus, as well their local gap junction and inhibitory synaptic connections reveals this period to be highly dynamic with numerous properties changing. However it is possible to make three main observations. Firstly, many aspects of SPNs mature in parallel, including intrinsic membrane properties, increases in dendritic arbours and spine densities, general synaptic inputs and expression of specific glutamate receptors. Secondly, there are notable exceptions, including a transient stronger thalamic innervation of D2 SPNs and stronger cortical NMDA receptor-mediated inputs to D1 SPNs, both in the second postnatal week. Thirdly, many of the defining properties of mature D1 and D2 SPNs and striatal circuits are already established by the first and second postnatal weeks, including different electrophysiological properties as well as biased local inhibitory connections between SPNs, suggesting this is guided through intrinsic developmental programmes. Together these findings provide an experimental framework for future studies of D1 and D2 SPN development in health and disease.
Collapse
Affiliation(s)
- Rohan N Krajeski
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK
| | - Anežka Macey-Dare
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK
| | - Fran van Heusden
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK
| | - Farid Ebrahimjee
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK
| | - Tommas J Ellender
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK
| |
Collapse
|
47
|
Melendez-Zaidi AE, Lakshminarasimhah H, Surmeier DJ. Cholinergic modulation of striatal nitric oxide-producing interneurons. Eur J Neurosci 2019; 50:3713-3731. [PMID: 31340071 DOI: 10.1111/ejn.14528] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 07/07/2019] [Accepted: 07/15/2019] [Indexed: 01/17/2023]
Abstract
Striatal GABAergic interneurons that express nitric oxide synthase-so-called low-threshold spike interneurons (LTSIs)-play several key roles in the striatum. But what drives the activity of these interneurons is less well defined. To fill this gap, a combination of monosynaptic rabies virus mapping (msRVm), electrophysiological and optogenetic approaches were used in transgenic mice in which LTSIs expressed either Cre recombinase or a fluorescent reporter. The rabies virus studies revealed a striking similarity in the afferent connectomes of LTSIs and neighboring cholinergic interneurons, particularly regarding connections arising from the parafascicular nucleus of the thalamus and cingulate cortex. While optogenetic stimulation of cingulate inputs excited both cholinergic interneurons and LTSIs, thalamic stimulation excited cholinergic interneurons, but inhibited LTSIs. This inhibition was dependent on cholinergic interneurons and had two components: a previously described GABAergic element and one that was mediated by M4 muscarinic acetylcholine receptors. In addition to this phasic signal, cholinergic interneurons tonically excited LTSIs through a distinct, M1 muscarinic acetylcholine receptor pathway. This coordinated cholinergic modulation of LTSIs predisposed them to rhythmically burst in response to phasic thalamic activity, potentially reconfiguring striatal circuitry in response to salient environmental stimuli.
Collapse
Affiliation(s)
- Alexandria E Melendez-Zaidi
- Department of Physiology, Feinberg School of Medicine, Chicago, IL, USA.,Medical Scientist Training Program, Feinberg School of Medicine, Chicago, IL, USA.,Department of Pediatrics, Division of Neurology and Developmental Neuroscience, Texas Children's Hospital/Baylor College of Medicine, Houston, TX, USA
| | | | | |
Collapse
|
48
|
Modulation and functions of dopamine receptor heteromers in drugs of abuse-induced adaptations. Neuropharmacology 2019; 152:42-50. [DOI: 10.1016/j.neuropharm.2018.12.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 11/27/2018] [Accepted: 12/03/2018] [Indexed: 12/18/2022]
|
49
|
A striatal interneuron circuit for continuous target pursuit. Nat Commun 2019; 10:2715. [PMID: 31222009 PMCID: PMC6586681 DOI: 10.1038/s41467-019-10716-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 05/28/2019] [Indexed: 12/19/2022] Open
Abstract
Most adaptive behaviors require precise tracking of targets in space. In pursuit behavior with a moving target, mice use distance to target to guide their own movement continuously. Here, we show that in the sensorimotor striatum, parvalbumin-positive fast-spiking interneurons (FSIs) can represent the distance between self and target during pursuit behavior, while striatal projection neurons (SPNs), which receive FSI projections, can represent self-velocity. FSIs are shown to regulate velocity-related SPN activity during pursuit, so that movement velocity is continuously modulated by distance to target. Moreover, bidirectional manipulation of FSI activity can selectively disrupt performance by increasing or decreasing the self-target distance. Our results reveal a key role of the FSI-SPN interneuron circuit in pursuit behavior and elucidate how this circuit implements distance to velocity transformation required for the critical underlying computation. Many natural behaviours involve tracking of a target in space. Here, the authors describe a task to assess this behaviour in mice and use in vivo electrophysiology, calcium imaging, optogenetics, and chemogenetics to investigate the role of the striatum in target pursuit.
Collapse
|
50
|
Kato S, Fukabori R, Nishizawa K, Okada K, Yoshioka N, Sugawara M, Maejima Y, Shimomura K, Okamoto M, Eifuku S, Kobayashi K. Action Selection and Flexible Switching Controlled by the Intralaminar Thalamic Neurons. Cell Rep 2019; 22:2370-2382. [PMID: 29490273 DOI: 10.1016/j.celrep.2018.02.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 09/19/2017] [Accepted: 02/02/2018] [Indexed: 01/01/2023] Open
Abstract
Learning processes contributing to appropriate selection and flexible switching of behaviors are mediated through the dorsal striatum, a key structure of the basal ganglia circuit. The major inputs to striatal subdivisions are provided from the intralaminar thalamic nuclei, including the central lateral nucleus (CL) and parafascicular nucleus (PF). Thalamostriatal neurons in the PF modulate the acquisition and performance of stimulus-response learning. Here, we address the roles of the CL thalamostriatal neurons in learning processes by using a selective neural pathway targeting technique. We show that the CL neurons are essential for the performance of stimulus-response learning and for behavioral flexibility, including reversal and attentional set-shifting of learned responses. In addition, chemogenetic suppression of neural activity supports the requirements of these neurons for behavioral flexibility. Our results suggest that the main contribution of the CL thalamostriatal neurons is functional control of the basal ganglia circuit linked to the prefrontal cortex.
Collapse
Affiliation(s)
- Shigeki Kato
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Ryoji Fukabori
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Kayo Nishizawa
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Kana Okada
- Department of Neurophysiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Nozomu Yoshioka
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Masateru Sugawara
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Yuko Maejima
- Department of Pharmacology, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Kenju Shimomura
- Department of Pharmacology, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Masahiro Okamoto
- Department of Systems Neuroscience, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Satoshi Eifuku
- Department of Systems Neuroscience, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Kazuto Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan; Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Kawaguchi 332-0012, Japan.
| |
Collapse
|