1
|
Yonk AJ, Linares-García I, Pasternak L, Juliani SE, Gradwell MA, George AJ, Margolis DJ. Role of Posterior Medial Thalamus in the Modulation of Striatal Circuitry and Choice Behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.21.586152. [PMID: 38585753 PMCID: PMC10996534 DOI: 10.1101/2024.03.21.586152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
The posterior medial (POm) thalamus is heavily interconnected with sensory and motor circuitry and is likely involved in behavioral modulation and sensorimotor integration. POm provides axonal projections to the dorsal striatum, a hotspot of sensorimotor processing, yet the role of POm-striatal projections has remained undetermined. Using optogenetics with slice electrophysiology, we found that POm provides robust synaptic input to direct and indirect pathway striatal spiny projection neurons (D1- and D2-SPNs, respectively) and parvalbumin-expressing fast spiking interneurons (PVs). During the performance of a whisker-based tactile discrimination task, POm-striatal projections displayed learning-related activation correlating with anticipatory, but not reward-related, pupil dilation. Inhibition of POm-striatal axons across learning caused slower reaction times and an increase in the number of training sessions for expert performance. Our data indicate that POm-striatal inputs provide a behaviorally relevant arousal-related signal, which may prime striatal circuitry for efficient integration of subsequent choice-related inputs.
Collapse
Affiliation(s)
- Alex J. Yonk
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, 08854, USA
| | - Ivan Linares-García
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, 08854, USA
| | - Logan Pasternak
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, 08854, USA
| | - Sofia E. Juliani
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, 08854, USA
| | - Mark A. Gradwell
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, 08854, USA
| | - Arlene J. George
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, 08854, USA
| | - David J. Margolis
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, 08854, USA
| |
Collapse
|
2
|
Gonzalo-Martín E, Alonso-Martínez C, Sepúlveda LP, Clasca F. Micropopulation mapping of the mouse parafascicular nucleus connections reveals diverse input-output motifs. Front Neuroanat 2024; 17:1305500. [PMID: 38260117 PMCID: PMC10800635 DOI: 10.3389/fnana.2023.1305500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 11/10/2023] [Indexed: 01/24/2024] Open
Abstract
Introduction In primates, including humans, the centromedian/parafascicular (CM-Pf) complex is a key thalamic node of the basal ganglia system. Deep brain stimulation in CM-Pf has been applied for the treatment of motor disorders such as Parkinson's disease or Tourette syndrome. Rodents have become widely used models for the study of the cellular and genetic mechanisms of these and other motor disorders. However, the equivalence between the primate CM-Pf and the nucleus regarded as analogous in rodents (Parafascicular, Pf) remains unclear. Methods Here, we analyzed the neurochemical architecture and carried out a brain-wide mapping of the input-output motifs in the mouse Pf at micropopulation level using anterograde and retrograde labeling methods. Specifically, we mapped and quantified the sources of cortical and subcortical input to different Pf subregions, and mapped and compared the distribution and terminal structure of their axons. Results We found that projections to Pf arise predominantly (>75%) from the cerebral cortex, with an unusually strong (>45%) Layer 5b component, which is, in part, contralateral. The intermediate layers of the superior colliculus are the main subcortical input source to Pf. On its output side, Pf neuron axons predominantly innervate the striatum. In a sparser fashion, they innervate other basal ganglia nuclei, including the subthalamic nucleus (STN), and the cerebral cortex. Differences are evident between the lateral and medial portions of Pf, both in chemoarchitecture and in connectivity. Lateral Pf axons innervate territories of the striatum, STN and cortex involved in the sensorimotor control of different parts of the contralateral hemibody. In contrast, the mediodorsal portion of Pf innervates oculomotor-limbic territories in the above three structures. Discussion Our data thus indicate that the mouse Pf consists of several neurochemically and connectively distinct domains whose global organization bears a marked similarity to that described in the primate CM-Pf complex.
Collapse
Affiliation(s)
| | | | | | - Francisco Clasca
- Department of Anatomy and Neuroscience, Autónoma de Madrid University, Madrid, Spain
| |
Collapse
|
3
|
Schiff ND, Giacino JT, Butson CR, Choi EY, Baker JL, O'Sullivan KP, Janson AP, Bergin M, Bronte-Stewart HM, Chua J, DeGeorge L, Dikmen S, Fogarty A, Gerber LM, Krel M, Maldonado J, Radovan M, Shah SA, Su J, Temkin N, Tourdias T, Victor JD, Waters A, Kolakowsky-Hayner SA, Fins JJ, Machado AG, Rutt BK, Henderson JM. Thalamic deep brain stimulation in traumatic brain injury: a phase 1, randomized feasibility study. Nat Med 2023; 29:3162-3174. [PMID: 38049620 PMCID: PMC11087147 DOI: 10.1038/s41591-023-02638-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 10/10/2023] [Indexed: 12/06/2023]
Abstract
Converging evidence indicates that impairments in executive function and information-processing speed limit quality of life and social reentry after moderate-to-severe traumatic brain injury (msTBI). These deficits reflect dysfunction of frontostriatal networks for which the central lateral (CL) nucleus of the thalamus is a critical node. The primary objective of this feasibility study was to test the safety and efficacy of deep brain stimulation within the CL and the associated medial dorsal tegmental (CL/DTTm) tract.Six participants with msTBI, who were between 3 and 18 years post-injury, underwent surgery with electrode placement guided by imaging and subject-specific biophysical modeling to predict activation of the CL/DTTm tract. The primary efficacy measure was improvement in executive control indexed by processing speed on part B of the trail-making test.All six participants were safely implanted. Five participants completed the study and one was withdrawn for protocol non-compliance. Processing speed on part B of the trail-making test improved 15% to 52% from baseline, exceeding the 10% benchmark for improvement in all five cases.CL/DTTm deep brain stimulation can be safely applied and may improve executive control in patients with msTBI who are in the chronic phase of recovery.ClinicalTrials.gov identifier: NCT02881151 .
Collapse
Affiliation(s)
- Nicholas D Schiff
- Feil Family Brain Mind Institute, Weill Cornell Medicine, New York, NY, USA.
- Department of Neurology, Weill Cornell Medicine, New York, NY, USA.
| | - Joseph T Giacino
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Boston, MA, USA
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA, USA
| | - Christopher R Butson
- Scientific Computing and Imaging Institute Department of Bioengineering, University of Utah, Salt Lake City, UT, USA
- Norman Fixel Institute for Neurological Diseases Departments of Neurology and Neurosurgery, University of Florida, Gainesville, FL, USA
| | - Eun Young Choi
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
| | - Jonathan L Baker
- Feil Family Brain Mind Institute, Weill Cornell Medicine, New York, NY, USA
| | - Kyle P O'Sullivan
- Scientific Computing and Imaging Institute Department of Bioengineering, University of Utah, Salt Lake City, UT, USA
| | - Andrew P Janson
- Scientific Computing and Imaging Institute Department of Bioengineering, University of Utah, Salt Lake City, UT, USA
- Radiology and Radiological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Michael Bergin
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Boston, MA, USA
| | | | - Jason Chua
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Laurel DeGeorge
- Feil Family Brain Mind Institute, Weill Cornell Medicine, New York, NY, USA
| | - Sureyya Dikmen
- Department of Rehabilitation Medicine, University of Washington, Seattle, WA, USA
| | - Adam Fogarty
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
| | - Linda M Gerber
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Mark Krel
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
| | - Jose Maldonado
- Department of Psychiatry, Stanford University, Stanford, CA, USA
| | - Matthew Radovan
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Sudhin A Shah
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Jason Su
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA
| | - Nancy Temkin
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - Thomas Tourdias
- Department of Neuroimaging, University of Bordeaux, Nouvelle-Aquitaine, France
| | - Jonathan D Victor
- Feil Family Brain Mind Institute, Weill Cornell Medicine, New York, NY, USA
- Department of Neurology, Weill Cornell Medicine, New York, NY, USA
| | - Abigail Waters
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Boston, MA, USA
| | | | - Joseph J Fins
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Andre G Machado
- Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Brian K Rutt
- Department of Radiology, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Bio-X Program, Stanford University, Stanford, CA, USA
| | - Jaimie M Henderson
- Department of Neurosurgery, Stanford University, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
- Bio-X Program, Stanford University, Stanford, CA, USA.
| |
Collapse
|
4
|
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
|
5
|
Padilla-Orozco M, Duhne M, Fuentes-Serrano A, Ortega A, Galarraga E, Bargas J, Lara-González E. Synaptic determinants of cholinergic interneurons hyperactivity during parkinsonism. Front Synaptic Neurosci 2022; 14:945816. [PMID: 36147730 PMCID: PMC9485566 DOI: 10.3389/fnsyn.2022.945816] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 07/21/2022] [Indexed: 11/25/2022] Open
Abstract
Parkinson’s disease is a neurodegenerative ailment generated by the loss of dopamine in the basal ganglia, mainly in the striatum. The disease courses with increased striatal levels of acetylcholine, disrupting the balance among these modulatory transmitters. These modifications disturb the excitatory and inhibitory balance in the striatal circuitry, as reflected in the activity of projection striatal neurons. In addition, changes in the firing pattern of striatal tonically active interneurons during the disease, including cholinergic interneurons (CINs), are being searched. Dopamine-depleted striatal circuits exhibit pathological hyperactivity as compared to controls. One aim of this study was to show how striatal CINs contribute to this hyperactivity. A second aim was to show the contribution of extrinsic synaptic inputs to striatal CINs hyperactivity. Electrophysiological and calcium imaging recordings in Cre-mice allowed us to evaluate the activity of dozens of identified CINs with single-cell resolution in ex vivo brain slices. CINs show hyperactivity with bursts and silences in the dopamine-depleted striatum. We confirmed that the intrinsic differences between the activity of control and dopamine-depleted CINs are one source of their hyperactivity. We also show that a great part of this hyperactivity and firing pattern change is a product of extrinsic synaptic inputs, targeting CINs. Both glutamatergic and GABAergic inputs are essential to sustain hyperactivity. In addition, cholinergic transmission through nicotinic receptors also participates, suggesting that the joint activity of CINs drives the phenomenon; since striatal CINs express nicotinic receptors, not expressed in striatal projection neurons. Therefore, CINs hyperactivity is the result of changes in intrinsic properties and excitatory and inhibitory inputs, in addition to the modification of local circuitry due to cholinergic nicotinic transmission. We conclude that CINs are the main drivers of the pathological hyperactivity present in the striatum that is depleted of dopamine, and this is, in part, a result of extrinsic synaptic inputs. These results show that CINs may be a main therapeutic target to treat Parkinson’s disease by intervening in their synaptic inputs.
Collapse
Affiliation(s)
- Montserrat Padilla-Orozco
- División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Mariana Duhne
- División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Department of Neurology, University of California, San Francisco, San Francisco, CA, United States
| | - Alejandra Fuentes-Serrano
- División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Aidán Ortega
- División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Elvira Galarraga
- División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - José Bargas
- División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
- *Correspondence: José Bargas,
| | - Esther Lara-González
- División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Esther Lara-González,
| |
Collapse
|
6
|
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: 6.0] [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
|
7
|
Aïssa HB, Sala RW, Georgescu Margarint EL, Frontera JL, Varani AP, Menardy F, Pelosi A, Hervé D, Léna C, Popa D. Functional abnormalities in the cerebello-thalamic pathways in a mouse model of DYT25 dystonia. eLife 2022; 11:79135. [PMID: 35699413 PMCID: PMC9197392 DOI: 10.7554/elife.79135] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/27/2022] [Indexed: 11/18/2022] Open
Abstract
Dystonia is often associated with functional alterations in the cerebello-thalamic pathways, which have been proposed to contribute to the disorder by propagating pathological firing patterns to the forebrain. Here, we examined the function of the cerebello-thalamic pathways in a model of DYT25 dystonia. DYT25 (Gnal+/−) mice carry a heterozygous knockout mutation of the Gnal gene, which notably disrupts striatal function, and systemic or striatal administration of oxotremorine to these mice triggers dystonic symptoms. Our results reveal an increased cerebello-thalamic excitability in the presymptomatic state. Following the first dystonic episode, Gnal+/- mice in the asymptomatic state exhibit a further increase of the cerebello-thalamo-cortical excitability, which is maintained after θ-burst stimulations of the cerebellum. When administered in the symptomatic state induced by a cholinergic activation, these stimulations decreased the cerebello-thalamic excitability and reduced dystonic symptoms. In agreement with dystonia being a multiregional circuit disorder, our results suggest that the increased cerebello-thalamic excitability constitutes an early endophenotype, and that the cerebellum is a gateway for corrective therapies via the depression of cerebello-thalamic pathways.
Collapse
Affiliation(s)
- Hind Baba Aïssa
- Neurophysiology of Brain Circuits Team, Institut de biologie de l'Ecole normale supérieure (IBENS), Ecole normale supérieure, CNRS, INSERM, PSL Research University, Paris, France
| | - Romain W Sala
- Neurophysiology of Brain Circuits Team, Institut de biologie de l'Ecole normale supérieure (IBENS), Ecole normale supérieure, CNRS, INSERM, PSL Research University, Paris, France
| | - Elena Laura Georgescu Margarint
- Neurophysiology of Brain Circuits Team, Institut de biologie de l'Ecole normale supérieure (IBENS), Ecole normale supérieure, CNRS, INSERM, PSL Research University, Paris, France
| | - Jimena Laura Frontera
- Neurophysiology of Brain Circuits Team, Institut de biologie de l'Ecole normale supérieure (IBENS), Ecole normale supérieure, CNRS, INSERM, PSL Research University, Paris, France
| | - Andrés Pablo Varani
- Neurophysiology of Brain Circuits Team, Institut de biologie de l'Ecole normale supérieure (IBENS), Ecole normale supérieure, CNRS, INSERM, PSL Research University, Paris, France
| | - Fabien Menardy
- Neurophysiology of Brain Circuits Team, Institut de biologie de l'Ecole normale supérieure (IBENS), Ecole normale supérieure, CNRS, INSERM, PSL Research University, Paris, France
| | - Assunta Pelosi
- Inserm UMR-S 1270, Paris, France.,Sorbonne Université, Sciences and Technology Faculty, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Denis Hervé
- Inserm UMR-S 1270, Paris, France.,Sorbonne Université, Sciences and Technology Faculty, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Clément Léna
- Neurophysiology of Brain Circuits Team, Institut de biologie de l'Ecole normale supérieure (IBENS), Ecole normale supérieure, CNRS, INSERM, PSL Research University, Paris, France
| | - Daniela Popa
- Neurophysiology of Brain Circuits Team, Institut de biologie de l'Ecole normale supérieure (IBENS), Ecole normale supérieure, CNRS, INSERM, PSL Research University, Paris, France
| |
Collapse
|
8
|
Tanimura A, Shen W, Wokosin D, Surmeier DJ. Pathway-Specific Remodeling of Thalamostriatal Synapses in a Mouse Model of Parkinson's Disease. Mov Disord 2022; 37:1164-1174. [PMID: 35485341 PMCID: PMC9232945 DOI: 10.1002/mds.29030] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 03/21/2022] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The network pathophysiology underlying the motor symptoms of Parkinson's disease (PD) is poorly understood. In models of late-stage PD, there is significant cell-specific remodeling of corticostriatal, axospinous glutamatergic synapses on principal spiny projection neurons (SPNs). Neurons in the centrolateral nucleus (CLN) of the thalamus that relay cerebellar activity to the striatum also make axospinous synapses on SPNs, but the extent to which they are affected in PD has not been definitively characterized. OBJECTIVE To fill this gap, transgenic mice in which CLN neurons express Cre recombinase were used in conjunction with optogenetic and circuit mapping approaches to determine changes in the CLN projection to SPNs in a unilateral 6-hydroxydopamine (6-OHDA) model of late-stage PD. METHODS Adeno-associated virus vectors carrying Cre-dependent opsin expression constructs were stereotaxically injected into the CLN of Grp-KH288 mice in which CLN, but not parafascicular nucleus neurons, expressed Cre recombinase. The properties of this projection to identify direct pathway spiny projection neurons (dSPNs) and indirect pathway spiny projection neurons (iSPNs) were then studied in ex vivo brain slices of the dorsolateral striatum from control and 6-OHDA lesioned mice using anatomic, optogenetic, and electrophysiological approaches. RESULTS Optogenetically evoked excitatory synaptic currents in both iSPNs and dSPNs were reduced in lesioned mice; however, the reduction was significantly greater in dSPNs. In iSPNs, the reduction in evoked responses was attributable to synaptic pruning, because synaptic channelrhodopsin assisted circuit mapping (sCRACm) revealed fewer synapses per cell after lesioning. In contrast, sCRACm mapping of CLN inputs to dSPNs failed to detect any change in synapse abundance in lesioned mice. However, the ratio of currents through α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors to those through N-methyl-D-aspartate receptors was significantly reduced in dSPNs. Moreover, the distribution of currents evoked by optical stimulation of individual synapses shifted toward smaller amplitudes by lesioning, suggesting that they had undergone long-term depression. CONCLUSIONS Taken together, our results demonstrate that the CLN projection to the striatum undergoes a pathway-specific remodeling that could contribute to the circuit imbalance thought to drive the hypokinetic features of PD. © 2022 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Asami Tanimura
- Department of Neuroscience, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
- Department of BiomedicineAarhus UniversityAarhusDenmark
| | - Weixing Shen
- Department of Neuroscience, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - David Wokosin
- Department of Neuroscience, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - D. James Surmeier
- Department of Neuroscience, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| |
Collapse
|
9
|
Striatal synaptic adaptations in Parkinson's disease. Neurobiol Dis 2022; 167:105686. [PMID: 35272023 DOI: 10.1016/j.nbd.2022.105686] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 02/16/2022] [Accepted: 03/03/2022] [Indexed: 01/02/2023] Open
Abstract
The striatum is densely innervated by mesencephalic dopaminergic neurons that modulate acquisition and vigor of goal-directed actions and habits. This innervation is progressively lost in Parkinson's disease (PD), contributing to the defining movement deficits of the disease. Although boosting dopaminergic signaling with levodopa early in the course of the disease alleviates these deficits, later this strategy leads to the emergence of debilitating dyskinesia. Here, recent advances in our understanding of how striatal cells and circuits adapt to this progressive de-innervation and to levodopa therapy are discussed. First, we discuss how dopamine (DA) depletion triggers cell type-specific, homeostatic changes in spiny projection neurons (SPNs) that tend to normalize striatal activity but also lead to disruption of the synaptic architecture sculpted by experience. Second, we discuss the roles played by cholinergic and nitric oxide-releasing interneurons in these adaptations. Third, we examine recent work in freely moving mice suggesting that alterations in the spatiotemporal dynamics of striatal ensembles contributes to PD movement deficits. Lastly, we discuss recently published evidence from a progressive model of PD suggesting that contrary to the classical model, striatal pathway imbalance is necessary but not sufficient to produce frank parkinsonism.
Collapse
|
10
|
Wolff SBE, Ko R, Ölveczky BP. Distinct roles for motor cortical and thalamic inputs to striatum during motor skill learning and execution. SCIENCE ADVANCES 2022; 8:eabk0231. [PMID: 35213216 PMCID: PMC8880788 DOI: 10.1126/sciadv.abk0231] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 01/03/2022] [Indexed: 05/11/2023]
Abstract
The acquisition and execution of motor skills are mediated by a distributed motor network, spanning cortical and subcortical brain areas. The sensorimotor striatum is an important cog in this network, yet the roles of its two main inputs, from motor cortex and thalamus, remain largely unknown. To address this, we silenced the inputs in rats trained on a task that results in highly stereotyped and idiosyncratic movement patterns. While striatal-projecting motor cortex neurons were critical for learning these skills, silencing this pathway after learning had no effect on performance. In contrast, silencing striatal-projecting thalamus neurons disrupted the execution of the learned skills, causing rats to revert to species-typical pressing behaviors and preventing them from relearning the task. These results show distinct roles for motor cortex and thalamus in the learning and execution of motor skills and suggest that their interaction in the striatum underlies experience-dependent changes in subcortical motor circuits.
Collapse
Affiliation(s)
| | - Raymond Ko
- Department of Organismic and Evolutionary Biology and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | | |
Collapse
|
11
|
Janson AP, Baker JL, Sani I, Purpura KP, Schiff ND, Butson CR. Selective activation of central thalamic fiber pathway facilitates behavioral performance in healthy non-human primates. Sci Rep 2021; 11:23054. [PMID: 34845232 PMCID: PMC8630225 DOI: 10.1038/s41598-021-02270-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 11/09/2021] [Indexed: 01/28/2023] Open
Abstract
Central thalamic deep brain stimulation (CT-DBS) is an investigational therapy to treat enduring cognitive dysfunctions in structurally brain injured (SBI) patients. However, the mechanisms of CT-DBS that promote restoration of cognitive functions are unknown, and the heterogeneous etiology and recovery profiles of SBI patients contribute to variable outcomes when using conventional DBS strategies,which may result in off-target effects due to activation of multiple pathways. To disambiguate the effects of stimulation of two adjacent thalamic pathways, we modeled and experimentally compared conventional and novel 'field-shaping' methods of CT-DBS within the central thalamus of healthy non-human primates (NHP) as they performed visuomotor tasks. We show that selective activation of the medial dorsal thalamic tegmental tract (DTTm), but not of the adjacent centromedian-parafascicularis (CM-Pf) pathway, results in robust behavioral facilitation. Our predictive modeling approach in healthy NHPs directly informs ongoing and future clinical investigations of conventional and novel methods of CT-DBS for treating cognitive dysfunctions in SBI patients, for whom no therapy currently exists.
Collapse
Affiliation(s)
- A. P. Janson
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT USA
- Scientific Computing and Imaging Institute, Salt Lake City, UT USA
- Departments of Neurology and Neurosurgery, Vanderbilt University Medical Center, Nashville, TN USA
| | - J. L. Baker
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY USA
| | - I. Sani
- The Rockefeller University, New York, NY USA
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - K. P. Purpura
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY USA
| | - N. D. Schiff
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY USA
| | - C. R. Butson
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT USA
- Scientific Computing and Imaging Institute, Salt Lake City, UT USA
- Departments of Neurology, Neurosurgery, and Psychiatry, Salt Lake City, UT USA
- Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL USA
| |
Collapse
|
12
|
Kourosh-Arami M, Hosseini N, Komaki A. Brain is modulated by neuronal plasticity during postnatal development. J Physiol Sci 2021; 71:34. [PMID: 34789147 PMCID: PMC10716960 DOI: 10.1186/s12576-021-00819-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 10/27/2021] [Indexed: 11/10/2022]
Abstract
Neuroplasticity is referred to the ability of the nervous system to change its structure or functions as a result of former stimuli. It is a plausible mechanism underlying a dynamic brain through adaptation processes of neural structure and activity patterns. Nevertheless, it is still unclear how the plastic neural systems achieve and maintain their equilibrium. Additionally, the alterations of balanced brain dynamics under different plasticity rules have not been explored either. Therefore, the present article primarily aims to review recent research studies regarding homosynaptic and heterosynaptic neuroplasticity characterized by the manipulation of excitatory and inhibitory synaptic inputs. Moreover, it attempts to understand different mechanisms related to the main forms of synaptic plasticity at the excitatory and inhibitory synapses during the brain development processes. Hence, this study comprised surveying those articles published since 1988 and available through PubMed, Google Scholar and science direct databases on a keyword-based search paradigm. All in all, the study results presented extensive and corroborative pieces of evidence for the main types of plasticity, including the long-term potentiation (LTP) and long-term depression (LTD) of the excitatory and inhibitory postsynaptic potentials (EPSPs and IPSPs).
Collapse
Affiliation(s)
- Masoumeh Kourosh-Arami
- Department of Neuroscience, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Nasrin Hosseini
- Neuroscience Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Alireza Komaki
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
13
|
Kato S, Nishizawa K, Kobayashi K. Thalamostriatal System Controls the Acquisition, Performance, and Flexibility of Learning Behavior. Front Syst Neurosci 2021; 15:729389. [PMID: 34733142 PMCID: PMC8558393 DOI: 10.3389/fnsys.2021.729389] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 09/29/2021] [Indexed: 11/16/2022] Open
Abstract
The dorsal striatum (DS) is a key structure of the basal ganglia circuitry, which regulates various types of learning processes and flexible switching of behavior. Intralaminar thalamic nuclei (ILNs) provide the main source of thalamostriatal inputs to the DS and constitute multiple nuclear groups, each of which innervates specific subdivisions of the striatum. Although the anatomical and electrophysiological properties of thalamostriatal neurons have been previously characterized, the behavioral and physiological functions of these neurons remain unclarified. Two representative thalamostriatal cell groups in the parafascicular nucleus (PF) and the central lateral nucleus (CL) are located in the caudal and rostral regions of the ILNs in rodents. Recently, the behavioral roles of these thalamostriatal cell groups have been investigated by the use of genetic and pharmacological manipulation techniques. In the current review, we summarize behavioral studies on thalamostriatal neurons, showing the key roles of these neurons in different learning processes, such as the acquisition, performance, and flexibility of behavior.
Collapse
Affiliation(s)
- Shigeki Kato
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Kayo Nishizawa
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Kazuto Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan
| |
Collapse
|
14
|
Cover KK, Mathur BN. Rostral Intralaminar Thalamus Engagement in Cognition and Behavior. Front Behav Neurosci 2021; 15:652764. [PMID: 33935663 PMCID: PMC8082140 DOI: 10.3389/fnbeh.2021.652764] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 03/22/2021] [Indexed: 11/25/2022] Open
Abstract
The thalamic rostral intralaminar nuclei (rILN) are a contiguous band of neurons that include the central medial, paracentral, and central lateral nuclei. The rILN differ from both thalamic relay nuclei, such as the lateral geniculate nucleus, and caudal intralaminar nuclei, such as the parafascicular nucleus, in afferent and efferent connectivity as well as physiological and synaptic properties. rILN activity is associated with a range of neural functions and behaviors, including arousal, pain, executive function, and action control. Here, we review this evidence supporting a role for the rILN in integrating arousal, executive and motor feedback information. In light of rILN projections out to the striatum, amygdala, and sensory as well as executive cortices, we propose that such a function enables the rILN to modulate cognitive and motor resources to meet task-dependent behavioral engagement demands.
Collapse
Affiliation(s)
- Kara K Cover
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Brian N Mathur
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
15
|
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: 0.5] [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
|
16
|
Firing Differences Between Adult Intralaminar Thalamo-striatal Neurons. Neuroscience 2021; 458:153-165. [PMID: 33428968 DOI: 10.1016/j.neuroscience.2020.12.032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 12/08/2020] [Accepted: 12/27/2020] [Indexed: 11/23/2022]
Abstract
Differences in the intrinsic properties of intralaminar thalamo-striatal neurons such as expressing low-threshold-spikes (LTS) or after hyperpolarizing potentials (AHPs) of different duration have been attributed to different maturation stages. However, two morphological types: "diffuse" and "bushy" have been described. Therefore, we explored whether electrophysiological differences persist in adult mice using whole cell recordings. Some recorded neurons were identified by intracellular labeling with biocytin and double labeling with retrograde or anterograde tracings using Cre-mice. We classified these neurons by their AHPs during spontaneous firing. Neurons with long duration AHPs, with fast and slow components, were mostly found in the parafascicular (Pf) nucleus. Neurons with brief AHPs were mainly found in the central lateral (CL) nucleus. However, neurons with both AHPs were found in both nuclei in different proportions. Firing frequency adaptation differed between these neuron classes: those with prolonged AHPs exhibited firing frequency adaptation with fast and slow time constants whereas those with brief AHPs were slow adapters. Neurons with more prolonged AHPs had significant higher input resistances than neurons with brief AHPs. Both cell classes could fire in two modes: trains of single action potentials at depolarized potentials or high frequency bursts on top of LTS at more hyperpolarized potentials. LTS were probably generated by Cav3 calcium channels since they were blocked by the selective antagonist TTA-P2. About 11% of neurons with brief AHPs and 55% of neurons with prolonged AHPs do not show LTS and bursts, even when potassium currents are blocked.
Collapse
|
17
|
Chuhma N. Functional Connectome Analysis of the Striatum with Optogenetics. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1293:417-428. [PMID: 33398830 DOI: 10.1007/978-981-15-8763-4_27] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Neural circuit function is determined not only by anatomical connections but also by the strength and nature of the connections, that is functional or physiological connectivity. To elucidate functional connectivity, selective stimulation of presynaptic terminals of an identified neuronal population is crucial. However, in the central nervous system, intermingled input fibers make selective electrical stimulation impossible. With optogenetics, this becomes possible, and enables the comprehensive study of functional synaptic connections between an identified population of neurons and defined postsynaptic targets to determine the functional connectome. By stimulating convergent synaptic inputs impinging on individual postsynaptic neurons, low frequency and small amplitude synaptic connections can be detected. Further, the optogenetic approach enables the measurement of cotransmission and its relative strength. Recently, optogenetic methods have been more widely used to study synaptic connectivity and revealed novel synaptic connections and revised connectivity of known projections. In this chapter, I focus on functional synaptic connectivity in the striatum, the main input structure of the basal ganglia, involved in the motivated behavior, cognition, and motor control, and its disruption in a range of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Nao Chuhma
- Department of Psychiatry, Columbia University, New York, NY, USA. .,Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA.
| |
Collapse
|
18
|
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: 9.0] [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
|
19
|
Kato S, Kobayashi K. Pseudotyped lentiviral vectors for tract-targeting and application for the functional control of selective neural circuits. J Neurosci Methods 2020; 344:108854. [PMID: 32663549 DOI: 10.1016/j.jneumeth.2020.108854] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/03/2020] [Accepted: 07/04/2020] [Indexed: 12/13/2022]
Abstract
A lentiviral vector strategy for efficient gene transfer through retrograde axonal transport provides a powerful approach for studying the neural circuit mechanisms that mediate higher level functions of the central nervous system. Pseudotyping of human immunodeficiency virus type-1 with different types of fusion glycoproteins (FuGs), which are composed of segments of rabies virus glycoprotein (RV-G) and vesicular stomatitis virus glycoprotein (VSV-G), enhances the efficiency of retrograde gene transfer in both rodent and non-human primate brains. These pseudotyped lentiviral vectors are classified into two groups, highly efficient retrograde gene transfer (HiRet) and neuron-specific retrograde gene transfer (NeuRet) vectors, based on their properties of gene transduction. Combinatorial use of the pseudotyped vectors with various molecular tools for manipulating neural circuit functions (such as the cell targeting, synaptic silencing, and optogenetic or chemogenetic approaches) enables us to control the function of specific neural circuits, thus leading to a deeper understanding of the mechanism underlying various nervous system functions.
Collapse
Affiliation(s)
- Shigeki Kato
- Department of Molecular Genetics, Institute of Biomedical Sciences, 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.
| |
Collapse
|
20
|
Grillner S, Robertson B, Kotaleski JH. Basal Ganglia—A Motion Perspective. Compr Physiol 2020; 10:1241-1275. [DOI: 10.1002/cphy.c190045] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
21
|
Intrinsic electrophysiological properties predict variability in morphology and connectivity among striatal Parvalbumin-expressing Pthlh-cells. Sci Rep 2020; 10:15680. [PMID: 32973206 PMCID: PMC7518419 DOI: 10.1038/s41598-020-72588-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 09/02/2020] [Indexed: 12/27/2022] Open
Abstract
Determining the cellular content of the nervous system in terms of cell types and the rules of their connectivity represents a fundamental challenge to the neurosciences. The recent advent of high-throughput techniques, such as single-cell RNA-sequencing has allowed for greater resolution in the identification of cell types and/or states. Although most of the current neuronal classification schemes comprise discrete clusters, several recent studies have suggested that, perhaps especially, within the striatum, neuronal populations exist in continua, with regards to both their molecular and electrophysiological properties. Whether these continua are stable properties, established during development, or if they reflect acute differences in activity-dependent regulation of critical genes is currently unknown. We set out to determine whether gradient-like molecular differences in the recently described Pthlh-expressing inhibitory interneuron population, which contains the Pvalb-expressing cells, correlate with differences in morphological and connectivity properties. We show that morphology and long-range inputs correlate with a spatially organized molecular and electrophysiological gradient of Pthlh-interneurons, suggesting that the processing of different types of information (by distinct anatomical striatal regions) has different computational requirements.
Collapse
|
22
|
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: 2.2] [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
|
23
|
Tubert C, Murer MG. What’s wrong with the striatal cholinergic interneurons in Parkinson’s disease? Focus on intrinsic excitability. Eur J Neurosci 2020; 53:2100-2116. [DOI: 10.1111/ejn.14742] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 04/04/2020] [Accepted: 04/05/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Cecilia Tubert
- Instituto de Fisiología y Biofísica “Bernardo Houssay”, (IFIBIO‐Houssay) Grupo de Neurociencia de Sistemas Universidad de Buenos Aires y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Buenos Aires Argentina
| | - Mario Gustavo Murer
- Instituto de Fisiología y Biofísica “Bernardo Houssay”, (IFIBIO‐Houssay) Grupo de Neurociencia de Sistemas Universidad de Buenos Aires y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Buenos Aires Argentina
| |
Collapse
|
24
|
Cover KK, Gyawali U, Kerkhoff WG, Patton MH, Mu C, White MG, Marquardt AE, Roberts BM, Cheer JF, Mathur BN. Activation of the Rostral Intralaminar Thalamus Drives Reinforcement through Striatal Dopamine Release. Cell Rep 2020; 26:1389-1398.e3. [PMID: 30726725 PMCID: PMC6402336 DOI: 10.1016/j.celrep.2019.01.044] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 10/29/2018] [Accepted: 01/09/2019] [Indexed: 11/29/2022] Open
Abstract
Glutamatergic projections of the thalamic rostral intralaminar nuclei of the thalamus (rILN) innervate the dorsal striatum (DS) and are implicated in dopamine (DA)-dependent incubation of drug seeking. However, the mechanism by which rILN signaling modulates reward seeking and striatal DA release is unknown. We find that activation of rILN inputs to the DS drives cholinergic interneuron burst-firing behavior and DA D2 receptor-dependent post-burst pauses in cholinergic interneuron firing. In vivo, optogenetic activation of this pathway drives reinforcement in a DA D1 receptor-dependent manner, and chemogenetic suppression of the rILN reduces dopaminergic nigrostriatal terminal activity as measured by fiber photometry. Altogether, these data provide evidence that the rILN activates striatal cholinergic interneurons to enhance the pursuit of reward through local striatal DA release and introduce an additional level of complexity in our understanding of striatal DA signaling. Cover et al. identify a glutamatergic thalamostriatal pathway that locally elicits striatal dopamine release to drive reward-related behavior in mice.
Collapse
Affiliation(s)
- Kara K Cover
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Utsav Gyawali
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Willa G Kerkhoff
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Mary H Patton
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Chaoqi Mu
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Michael G White
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Ashley E Marquardt
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Bradley M Roberts
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Joseph F Cheer
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Brian N Mathur
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
25
|
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: 1.8] [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
|
26
|
Crevier-Sorbo G, Rymar VV, Crevier-Sorbo R, Sadikot AF. Thalamostriatal degeneration contributes to dystonia and cholinergic interneuron dysfunction in a mouse model of Huntington's disease. Acta Neuropathol Commun 2020; 8:14. [PMID: 32033588 PMCID: PMC7007676 DOI: 10.1186/s40478-020-0878-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 01/03/2020] [Indexed: 01/18/2023] Open
Abstract
Huntington’s disease (HD) is an autosomal dominant trinucleotide repeat disorder characterized by choreiform movements, dystonia and striatal neuronal loss. Amongst multiple cellular processes, abnormal neurotransmitter signalling and decreased trophic support from glutamatergic cortical afferents are major mechanisms underlying striatal degeneration. Recent work suggests that the thalamostriatal (TS) system, another major source of glutamatergic input, is abnormal in HD although its phenotypical significance is unknown. We hypothesized that TS dysfunction plays an important role in generating motor symptoms and contributes to degeneration of striatal neuronal subtypes. Our results using the R6/2 mouse model of HD indicate that neurons of the parafascicular nucleus (PF), the main source of TS afferents, degenerate at an early stage. PF lesions performed prior to motor dysfunction or striatal degeneration result in an accelerated dystonic phenotype and are associated with premature loss of cholinergic interneurons. The progressive loss of striatal medium spiny neurons and parvalbumin-positive interneurons observed in R6/2 mice is unaltered by PF lesions. Early striatal cholinergic ablation using a mitochondrial immunotoxin provides evidence for increased cholinergic vulnerability to cellular energy failure in R6/2 mice, and worsens the dystonic phenotype. The TS system therefore contributes to trophic support of striatal interneuron subtypes in the presence of neurodegenerative stress, and TS deafferentation may be a novel cell non-autonomous mechanism contributing to the pathogenesis of HD. Furthermore, behavioural experiments demonstrate that the TS system and striatal cholinergic interneurons are key motor-network structures involved in the pathogenesis of dystonia. This work suggests that treatments aimed at rescuing the TS system may preserve important elements of striatal structure and function and provide symptomatic relief in HD.
Collapse
|
27
|
Liu J, Shelkar GP, Gandhi PJ, Gawande DY, Hoover A, Villalba RM, Pavuluri R, Smith Y, Dravid SM. Striatal glutamate delta-1 receptor regulates behavioral flexibility and thalamostriatal connectivity. Neurobiol Dis 2020; 137:104746. [PMID: 31945419 PMCID: PMC7204410 DOI: 10.1016/j.nbd.2020.104746] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 12/19/2019] [Accepted: 01/12/2020] [Indexed: 10/31/2022] Open
Abstract
Impaired behavioral flexibility and repetitive behavior is a common phenotype in autism and other neuropsychiatric disorders, but the underlying synaptic mechanisms are poorly understood. The trans-synaptic glutamate delta (GluD)-Cerebellin 1-Neurexin complex, critical for synapse formation/maintenance, represents a vulnerable axis for neuropsychiatric diseases. We have previously found that GluD1 deletion results in reversal learning deficit and repetitive behavior. In this study, we show that selective ablation of GluD1 from the dorsal striatum impairs behavioral flexibility in a water T-maze task. We further found that striatal GluD1 is preferentially found in dendritic shafts, and more frequently associated with thalamic than cortical glutamatergic terminals suggesting localization to projections from the thalamic parafascicular nucleus (Pf). Conditional deletion of GluD1 from the striatum led to a selective loss of thalamic, but not cortical, terminals, and reduced glutamatergic neurotransmission. Optogenetic studies demonstrated functional changes at thalamostriatal synapses from the Pf, but no effect on the corticostriatal system, upon ablation of GluD1 in the dorsal striatum. These studies suggest a novel molecular mechanism by which genetic variations associated with neuropsychiatric disorders may impair behavioral flexibility, and reveal a unique principle by which GluD1 subunit regulates forebrain circuits.
Collapse
Affiliation(s)
- Jinxu Liu
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Gajanan P Shelkar
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Pauravi J Gandhi
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Dinesh Y Gawande
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Andrew Hoover
- Yerkes National Primate Research Center, Atlanta, GA 30329, USA; UDALL Center of Excellence for Parkinson's Disease, Atlanta, GA 30329, USA
| | - Rosa M Villalba
- Yerkes National Primate Research Center, Atlanta, GA 30329, USA; UDALL Center of Excellence for Parkinson's Disease, Atlanta, GA 30329, USA
| | - Ratnamala Pavuluri
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Yoland Smith
- Yerkes National Primate Research Center, Atlanta, GA 30329, USA; UDALL Center of Excellence for Parkinson's Disease, Atlanta, GA 30329, USA; Dept. Neurology, Emory University, Atlanta, GA 30329, USA
| | - Shashank M Dravid
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA.
| |
Collapse
|
28
|
Assous M, Tepper JM. Cortical and thalamic inputs exert cell type-specific feedforward inhibition on striatal GABAergic interneurons. J Neurosci Res 2019; 97:1491-1502. [PMID: 31102306 PMCID: PMC6801038 DOI: 10.1002/jnr.24444] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 04/24/2019] [Accepted: 04/29/2019] [Indexed: 12/12/2022]
Abstract
The classical view of striatal GABAergic interneuron function has been that they operate as largely independent, parallel, feedforward inhibitory elements providing inhibitory inputs to spiny projection neurons (SPNs). Much recent evidence has shown that the extrinsic innervation of striatal interneurons is not indiscriminate but rather very specific, and that striatal interneurons are themselves interconnected in a cell type-specific manner. This suggests that the ultimate effect of extrinsic inputs on striatal neuronal activity depends critically on synaptic interactions within interneuronal circuitry. Here, we compared the cortical and thalamic input to two recently described subtypes of striatal GABAergic interneurons, tyrosine hydroxylase-expressing interneurons (THINs), and spontaneously active bursty interneurons (SABIs) using transgenic TH-Cre and Htr3a-Cre mice of both sexes. Our results show that both THINs and SABIs receive strong excitatory input from the motor cortex and the thalamic parafascicular nucleus. Cortical optogenetic stimulation also evokes disynaptic inhibitory GABAergic responses in THINs but not in SABIs. In contrast, optogenetic stimulation of the parafascicular nucleus induces disynaptic inhibitory responses in both interneuron populations. However, the short-term plasticity of these disynaptic inhibitory responses is different suggesting the involvement of different intrastriatal microcircuits. Altogether, our results point to highly specific interneuronal circuits that are selectively engaged by different excitatory inputs.
Collapse
Affiliation(s)
- Maxime Assous
- Center for Molecular and Behavioral Neuroscience, Rutgers University, Newark, NJ 07102
| | - James M. Tepper
- Center for Molecular and Behavioral Neuroscience, Rutgers University, Newark, NJ 07102
| |
Collapse
|
29
|
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: 2.7] [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
|
30
|
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: 5.3] [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
|
31
|
Fujiyama F, Unzai T, Karube F. Thalamostriatal projections and striosome-matrix compartments. Neurochem Int 2019; 125:67-73. [DOI: 10.1016/j.neuint.2019.01.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 01/18/2019] [Accepted: 01/27/2019] [Indexed: 12/11/2022]
|
32
|
Mandelbaum G, Taranda J, Haynes TM, Hochbaum DR, Huang KW, Hyun M, Umadevi Venkataraju K, Straub C, Wang W, Robertson K, Osten P, Sabatini BL. Distinct Cortical-Thalamic-Striatal Circuits through the Parafascicular Nucleus. Neuron 2019; 102:636-652.e7. [PMID: 30905392 DOI: 10.1016/j.neuron.2019.02.035] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 01/03/2019] [Accepted: 02/20/2019] [Indexed: 01/08/2023]
Abstract
The thalamic parafascicular nucleus (PF), an excitatory input to the basal ganglia, is targeted with deep-brain stimulation to alleviate a range of neuropsychiatric symptoms. Furthermore, PF lesions disrupt the execution of correct motor actions in uncertain environments. Nevertheless, the circuitry of the PF and its contribution to action selection are poorly understood. We find that, in mice, PF has the highest density of striatum-projecting neurons among all sub-cortical structures. This projection arises from transcriptionally and physiologically distinct classes of PF neurons that are also reciprocally connected with functionally distinct cortical regions, differentially innervate striatal neurons, and are not synaptically connected in PF. Thus, mouse PF contains heterogeneous neurons that are organized into parallel and independent associative, limbic, and somatosensory circuits. Furthermore, these subcircuits share motifs of cortical-PF-cortical and cortical-PF-striatum organization that allow each PF subregion, via its precise connectivity with cortex, to coordinate diverse inputs to striatum.
Collapse
Affiliation(s)
- Gil Mandelbaum
- Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Julian Taranda
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Trevor M Haynes
- Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Daniel R Hochbaum
- Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA; Society of Fellows, Harvard University, Cambridge, MA 02138, USA
| | - Kee Wui Huang
- Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Minsuk Hyun
- Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | | | - Christoph Straub
- Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Wengang Wang
- Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Keiramarie Robertson
- Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Pavel Osten
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Bernardo L Sabatini
- Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
33
|
Choi K, Holly EN, Davatolhagh MF, Beier KT, Fuccillo MV. Integrated anatomical and physiological mapping of striatal afferent projections. Eur J Neurosci 2019; 49:623-636. [PMID: 29359830 PMCID: PMC6056328 DOI: 10.1111/ejn.13829] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 12/15/2017] [Accepted: 01/08/2018] [Indexed: 12/31/2022]
Abstract
The dorsomedial striatum, a key site of reward-sensitive motor output, receives extensive afferent input from cortex, thalamus and midbrain. These projections are integrated by striatal microcircuits containing both spiny projection neurons and local circuit interneurons. To explore target cell specificity of these projections, we compared inputs onto D1-dopamine receptor-positive spiny neurons, parvalbumin-positive fast-spiking interneurons and somatostatin-positive low-threshold-spiking interneurons, using cell type-specific rabies virus tracing and optogenetic-mediated projection neuron recruitment in mice. While the relative proportion of retrogradely labelled projection neurons was similar between target cell types, the convergence of inputs was systematically higher for projections onto fast-spiking interneurons. Rabies virus is frequently used to assess cell-specific anatomical connectivity but it is unclear how this correlates to synaptic connectivity and efficacy. To test this, we compared tracing data with target cell-specific measures of synaptic efficacy for anterior cingulate cortex and parafascicular thalamic projections using novel quantitative optogenetic measures. We found that target-specific patterns of convergence were extensively modified according to region of projection neuron origin and postsynaptic cell type. Furthermore, we observed significant divergence between cell type-specific anatomical connectivity and measures of excitatory synaptic strength, particularly for low-threshold-spiking interneurons. Taken together, this suggests a basic uniform connectivity map for striatal afferent inputs upon which presynaptic-postsynaptic interactions impose substantial diversity of physiological connectivity.
Collapse
Affiliation(s)
- Kyuhyun Choi
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Clinical Research Building, Room 226, Philadelphia, PA, 19104, USA
| | - Elizabeth N Holly
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Clinical Research Building, Room 226, Philadelphia, PA, 19104, USA
| | - M Felicia Davatolhagh
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Clinical Research Building, Room 226, Philadelphia, PA, 19104, USA
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kevin T Beier
- Department of Psychiatry and Behavioral Sciences, Stanford University Medical School, Stanford, CA, USA
| | - Marc V Fuccillo
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Clinical Research Building, Room 226, Philadelphia, PA, 19104, USA
| |
Collapse
|
34
|
Aceves Buendia JDJ, Tiroshi L, Chiu W, Goldberg JA. Selective remodeling of glutamatergic transmission to striatal cholinergic interneurons after dopamine depletion. Eur J Neurosci 2019; 49:824-833. [PMID: 28922504 PMCID: PMC6519226 DOI: 10.1111/ejn.13715] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 09/13/2017] [Accepted: 09/13/2017] [Indexed: 01/12/2023]
Abstract
The widely held view that the pathophysiology of Parkinson's disease arises from an under-activation of the direct pathway striatal spiny neurons (dSPNs) has gained support from a recently described weakening of the glutamatergic projection from the parafascicular nucleus (PfN) to dSPNs in experimental parkinsonism. However, the impact of the remodeling of the thalamostriatal projection cannot be fully appreciated without considering its impact on cholinergic interneurons (ChIs) that themselves preferentially activate indirect pathway spiny neurons (iSPNs). To study this thalamostriatal projection, we virally transfected with Cre-dependent channelrhodopsin-2 (ChR2) the PfN of Vglut2-Cre mice that were dopamine-depleted with 6-hydroxydopamine (6-OHDA). In parallel, we studied the corticostriatal projection to ChIs in 6-OHDA-treated transgenic mice expressing ChR2 under the Thy1 promoter. We found the 6-OHDA lesions failed to affect short-term synaptic plasticity or the size of unitary responses evoked optogenetically in either of these projections. However, we found that NMDA-to-AMPA ratios at PfN synapses-that were significantly larger than NMDA-to-AMPA ratios at cortical synapses-were reduced by 6-OHDA treatment, thereby impairing synaptic integration at PfN synapses onto ChIs. Finally, we found that application of an agonist of the D5 dopamine receptors on ChIs potentiated NMDA currents without affecting AMPA currents or short-term plasticity selectively at PfN synapses. We propose that dopamine depletion leads to an effective de-potentiation of NMDA currents at PfN synapses onto ChIs which degrades synaptic integration. This selective remodeling of NMDA currents at PfN synapses may counter the selective weakening of PfN synapses onto dSPNs in parkinsonism.
Collapse
Affiliation(s)
- Jose de Jesus Aceves Buendia
- Department of Medical NeurobiologyInstitute of Medical Research Israel – CanadaThe Faculty of MedicineThe Hebrew University of Jerusalem9112102JerusalemIsrael
| | - Lior Tiroshi
- Department of Medical NeurobiologyInstitute of Medical Research Israel – CanadaThe Faculty of MedicineThe Hebrew University of Jerusalem9112102JerusalemIsrael
| | - Wei‐Hua Chiu
- Department of Medical NeurobiologyInstitute of Medical Research Israel – CanadaThe Faculty of MedicineThe Hebrew University of Jerusalem9112102JerusalemIsrael
| | - Joshua A. Goldberg
- Department of Medical NeurobiologyInstitute of Medical Research Israel – CanadaThe Faculty of MedicineThe Hebrew University of Jerusalem9112102JerusalemIsrael
| |
Collapse
|
35
|
Smith PH, Uhlrich DJ, Manning KA. Evaluation of medial division of the medial geniculate (MGM) and posterior intralaminar nucleus (PIN) inputs to the rat auditory cortex, amygdala, and striatum. J Comp Neurol 2019; 527:1478-1494. [PMID: 30689207 DOI: 10.1002/cne.24644] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 01/10/2019] [Accepted: 01/16/2019] [Indexed: 12/18/2022]
Abstract
The medial division of the medial geniculate (MGM) and the posterior intralaminar nucleus (PIN) are association nuclei of the auditory thalamus. We made tracer injections in these nuclei to evaluate/compare their presynaptic terminal and postsynaptic target features in auditory cortex, amygdala and striatum, at the light and electron microscopic levels. Cortical labeling was concentrated in Layer 1 but in other layers distribution was location-dependent. In cortical areas designated dorsal, primary and ventral (AuD, Au1, AuV) terminals deep to Layer 1 were concentrated in infragranular layers and sparser in the supragranular and middle layers. In ectorhinal cortex (Ect), distributions below Layer 1 changed with concentrations in supragranular and middle layers. In temporal association cortex (TeA) terminal distributions below Layer 1 was intermediate between AuV/1/D and Ect. In amygdala and striatum, terminal concentrations were higher in striatum but not as dense as in cortical Layer 1. Ultrastructurally, presynaptic terminal size was similar in amygdala, striatum or cortex and in all cortical layers. Postsynaptically MGM/PIN terminals everywhere synapsed on spines or small distal dendrites but as a population the postsynaptic structures in cortex were larger than those in the striatum. In addition, primary cortical targets of terminals were larger in primary cortex than in area Ect. Thus, although postsynaptic size may play some role in changes in synaptic influence between areas it appears that terminal size is not a variable used for that purpose. In auditory cortex, cortical subdivision-dependent changes in the terminal distribution between cortical layers may also play a role.
Collapse
Affiliation(s)
- Philip H Smith
- Department of Neuroscience, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Daniel J Uhlrich
- Department of Neuroscience, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Karen A Manning
- Department of Neuroscience, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| |
Collapse
|
36
|
Tanimura A, Du Y, Kondapalli J, Wokosin DL, Surmeier DJ. Cholinergic Interneurons Amplify Thalamostriatal Excitation of Striatal Indirect Pathway Neurons in Parkinson’s Disease Models. Neuron 2019; 101:444-458.e6. [DOI: 10.1016/j.neuron.2018.12.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 09/13/2018] [Accepted: 12/04/2018] [Indexed: 12/16/2022]
|
37
|
Balbinot G, Schuch CP. Compensatory Relearning Following Stroke: Cellular and Plasticity Mechanisms in Rodents. Front Neurosci 2019; 12:1023. [PMID: 30766468 PMCID: PMC6365459 DOI: 10.3389/fnins.2018.01023] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 12/18/2018] [Indexed: 11/13/2022] Open
Abstract
von Monakow’s theory of diaschisis states the functional ‘standstill’ of intact brain regions that are remote from a damaged area, often implied in recovery of function. Accordingly, neural plasticity and activity patterns related to recovery are also occurring at the same regions. Recovery relies on plasticity in the periinfarct and homotopic contralesional regions and involves relearning to perform movements. Seeking evidence for a relearning mechanism following stroke, we found that rodents display many features that resemble classical learning and memory mechanisms. Compensatory relearning is likely to be accompanied by gradual shaping of these regions and pathways, with participating neurons progressively adapting cortico-striato-thalamic activity and synaptic strengths at different cortico-thalamic loops – adapting function relayed by the striatum. Motor cortex functional maps are progressively reinforced and shaped by these loops as the striatum searches for different functional actions. Several cortical and striatal cellular mechanisms that influence motor learning may also influence post-stroke compensatory relearning. Future research should focus on how different neuromodulatory systems could act before, during or after rehabilitation to improve stroke recovery.
Collapse
Affiliation(s)
- Gustavo Balbinot
- Brain Institute, Universidade Federal do Rio Grande do Norte, Natal, Brazil
| | - Clarissa Pedrini Schuch
- Graduate Program in Rehabilitation Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
| |
Collapse
|
38
|
Activity Patterns in the Neuropil of Striatal Cholinergic Interneurons in Freely Moving Mice Represent Their Collective Spiking Dynamics. eNeuro 2019; 6:eN-NWR-0351-18. [PMID: 30671536 PMCID: PMC6338468 DOI: 10.1523/eneuro.0351-18.2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 12/24/2018] [Accepted: 12/27/2018] [Indexed: 01/25/2023] Open
Abstract
Cholinergic interneurons (CINs) are believed to form synchronous cell assemblies that modulate the striatal microcircuitry and possibly orchestrate local dopamine release. We expressed GCaMP6s, a genetically encoded calcium indicator (GECIs), selectively in CINs, and used microendoscopes to visualize the putative CIN assemblies in the dorsal striatum of freely moving mice. The GECI fluorescence signal from the dorsal striatum was composed of signals from individual CIN somata that were engulfed by a widespread fluorescent neuropil. Bouts of synchronous activation of the cholinergic neuropil revealed patterns of activity that preceded the signal from individual somata. To investigate the nature of the neuropil signal and why it precedes the somatic signal, we target-patched GECI-expressing CINs in acute striatal slices in conjunction with multiphoton imaging or wide-field imaging that emulates the microendoscopes' specifications. The ability to detect fluorescent transients associated with individual action potential was constrained by the long decay constant of GECIs (relative to common inorganic dyes) to slowly firing (<2 spikes/s) CINs. The microendoscopes' resolving power and sampling rate further diminished this ability. Additionally, we found that only back-propagating action potentials but not synchronous optogenetic activation of thalamic inputs elicited observable calcium transients in CIN dendrites. Our data suggest that only bursts of CIN activity (but not their tonic firing) are visible using endoscopic imaging, and that the neuropil patterns are a physiological measure of the collective recurrent CIN network spiking activity.
Collapse
|
39
|
Wang JB, Aryal M, Zhong Q, Vyas DB, Airan RD. Noninvasive Ultrasonic Drug Uncaging Maps Whole-Brain Functional Networks. Neuron 2018; 100:728-738.e7. [PMID: 30408444 PMCID: PMC6274638 DOI: 10.1016/j.neuron.2018.10.042] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 09/13/2018] [Accepted: 10/24/2018] [Indexed: 01/06/2023]
Abstract
Being able to noninvasively modulate brain activity, where and when an experimenter desires, with an immediate path toward human translation is a long-standing goal for neuroscience. To enable robust perturbation of brain activity while leveraging the ability of focused ultrasound to deliver energy to any point of the brain noninvasively, we have developed biocompatible and clinically translatable nanoparticles that allow ultrasound-induced uncaging of neuromodulatory drugs. Utilizing the anesthetic propofol, together with electrophysiological and imaging assays, we show that the neuromodulatory effect of ultrasonic drug uncaging is limited spatially and temporally by the size of the ultrasound focus, the sonication timing, and the pharmacokinetics of the uncaged drug. Moreover, we see secondary effects in brain regions anatomically distinct from and functionally connected to the sonicated region, indicating that ultrasonic drug uncaging could noninvasively map the changes in functional network connectivity associated with pharmacologic action at a particular brain target.
Collapse
Affiliation(s)
- Jeffrey B Wang
- Department of Radiology, Neuroradiology Division, Stanford University, Stanford, CA 94305, USA
| | - Muna Aryal
- Department of Radiology, Neuroradiology Division, Stanford University, Stanford, CA 94305, USA
| | - Qian Zhong
- Department of Radiology, Neuroradiology Division, Stanford University, Stanford, CA 94305, USA
| | - Daivik B Vyas
- Department of Radiology, Neuroradiology Division, Stanford University, Stanford, CA 94305, USA
| | - Raag D Airan
- Department of Radiology, Neuroradiology Division, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
40
|
Perrin E, Venance L. Bridging the gap between striatal plasticity and learning. Curr Opin Neurobiol 2018; 54:104-112. [PMID: 30321866 DOI: 10.1016/j.conb.2018.09.007] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 09/19/2018] [Accepted: 09/25/2018] [Indexed: 12/28/2022]
Abstract
The striatum, the main input nucleus of the basal ganglia, controls goal-directed behavior and procedural learning. Striatal projection neurons integrate glutamatergic inputs from cortex and thalamus together with neuromodulatory systems, and are subjected to plasticity. Striatal projection neurons exhibit bidirectional plasticity (LTP and LTD) when exposed to Hebbian paradigms. Importantly, correlative and even causal links between procedural learning and striatal plasticity have recently been shown. This short review summarizes the current view on striatal plasticity (with a focus on spike-timing-dependent plasticity), recent studies aiming at bridging in vivo skill acquisition and striatal plasticity, the temporal credit-assignment problem, and the gaps that remain to be filled.
Collapse
Affiliation(s)
- Elodie Perrin
- Center for Interdisciplinary Research in Biology, Collège de France, INSERM U1050, CNRS UMR7241, Labex Memolife, 75005 Paris, France; Université Pierre et Marie Curie, ED 158, Paris, France
| | - Laurent Venance
- Center for Interdisciplinary Research in Biology, Collège de France, INSERM U1050, CNRS UMR7241, Labex Memolife, 75005 Paris, France; Université Pierre et Marie Curie, ED 158, Paris, France.
| |
Collapse
|
41
|
"Shake it off" Versus "In Your Wildest Dreams": Thalamus as a Consciousness Gate for Temporal Lobe Seizures. Epilepsy Curr 2018; 18:248-250. [PMID: 30254522 DOI: 10.5698/1535-7597.18.4.248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
42
|
Cavaccini A, Gritti M, Giorgi A, Locarno A, Heck N, Migliarini S, Bertero A, Mereu M, Margiani G, Trusel M, Catelani T, Marotta R, De Luca MA, Caboche J, Gozzi A, Pasqualetti M, Tonini R. Serotonergic Signaling Controls Input-Specific Synaptic Plasticity at Striatal Circuits. Neuron 2018; 98:801-816.e7. [DOI: 10.1016/j.neuron.2018.04.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 02/15/2018] [Accepted: 04/05/2018] [Indexed: 10/17/2022]
|
43
|
Unzai T, Kuramoto E, Kaneko T, Fujiyama F. Quantitative Analyses of the Projection of Individual Neurons from the Midline Thalamic Nuclei to the Striosome and Matrix Compartments of the Rat Striatum. Cereb Cortex 2018; 27:1164-1181. [PMID: 26672610 DOI: 10.1093/cercor/bhv295] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
A fundamental organizing principle of the striatum is the striosome/matrix system that is defined by inputs/outputs and neurochemical markers. The thalamostriatal projection is highly heterogeneous originating in many subnuclei of the thalamus including the midline (ML) and intralaminar (IL) nuclei. We examined the dendritic morphology and axonal trajectory of 15 ML and 11 IL neurons by single-neuron labeling with viral vectors in combination with mu-opioid receptor immunostaining in rat brains. Dendritic and axonal morphology defined ML neurons as type II cells consisting of at least two subclasses according to the presence or absence of striatal axon collaterals. In the striatum, ML neurons preferentially innervated striosomes, whereas parafascicular neurons preferentially innervated the matrix. Almost all single thalamostriatal neurons favoring striosome or matrix compartments also innervated the cerebral cortical areas that supplied cortical input to the same striatal compartment. We thus revealed that thalamostriatal projections are highly organized 1) by the similarity in morphological characteristics and 2) their preference for the striatal compartments and cortical areas. These findings demonstrate that the functional properties of striatal compartments are influenced by both their cortical and thalamic afferents presumably with a different time latency and support selective dynamics for the striosome and matrix compartments.
Collapse
Affiliation(s)
- Tomo Unzai
- Laboratory of Neural Circuitry, Graduate School of Brain Science, Doshisha University, Kyoto 619-0394, Japan.,Department of Morphological Brain Science, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan.,Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Tokyo 102-0076, Japan
| | - Eriko Kuramoto
- Department of Morphological Brain Science, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan.,Department of Oral Anatomy and Cell Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | - Takeshi Kaneko
- Department of Morphological Brain Science, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Fumino Fujiyama
- Laboratory of Neural Circuitry, Graduate School of Brain Science, Doshisha University, Kyoto 619-0394, Japan.,Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Tokyo 102-0076, Japan
| |
Collapse
|
44
|
Nakano Y, Karube F, Hirai Y, Kobayashi K, Hioki H, Okamoto S, Kameda H, Fujiyama F. Parvalbumin-producing striatal interneurons receive excitatory inputs onto proximal dendrites from the motor thalamus in male mice. J Neurosci Res 2018; 96:1186-1207. [PMID: 29314192 DOI: 10.1002/jnr.24214] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 12/12/2017] [Accepted: 12/12/2017] [Indexed: 01/09/2023]
Abstract
In rodents, the dorsolateral striatum regulates voluntary movement by integrating excitatory inputs from the motor-related cerebral cortex and thalamus to produce contingent inhibitory output to other basal ganglia nuclei. Striatal parvalbumin (PV)-producing interneurons receiving this excitatory input then inhibit medium spiny neurons (MSNs) and modify their outputs. To understand basal ganglia function in motor control, it is important to reveal the precise synaptic organization of motor-related cortical and thalamic inputs to striatal PV interneurons. To examine which domains of the PV neurons receive these excitatory inputs, we used male bacterial artificial chromosome transgenic mice expressing somatodendritic membrane-targeted green fluorescent protein in PV neurons. An anterograde tracing study with the adeno-associated virus vector combined with immunodetection of pre- and postsynaptic markers visualized the distribution of the excitatory appositions on PV dendrites. Statistical analysis revealed that the density of thalamostriatal appositions along the dendrites was significantly higher on the proximal than distal dendrites. In contrast, there was no positional preference in the density of appositions from axons of the dorsofrontal cortex. Population observations of thalamostriatal and corticostriatal appositions by immunohistochemistry for pathway-specific vesicular glutamate transporters confirmed that thalamic inputs preferentially, and cortical ones less preferentially, made apposition on proximal dendrites of PV neurons. This axodendritic organization suggests that PV neurons produce fast and reliable inhibition of MSNs in response to thalamic inputs and process excitatory inputs from motor cortices locally and plastically, possibly together with other GABAergic and dopaminergic dendritic inputs, to modulate MSN inhibition.
Collapse
Affiliation(s)
- Yasutake Nakano
- Laboratory of Neural Circuitry, Graduate School of Brain Science, Doshisha University, Kyotanabe, Japan
| | - Fuyuki Karube
- Laboratory of Neural Circuitry, Graduate School of Brain Science, Doshisha University, Kyotanabe, Japan
| | - Yasuharu Hirai
- Laboratory of Neural Circuitry, Graduate School of Brain Science, Doshisha University, Kyotanabe, Japan
| | - Kenta Kobayashi
- Section of Viral Vector Development, National Institute for Physiological Sciences, Okazaki, Japan
| | - Hiroyuki Hioki
- Department of Morphological Brain Science, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shinichiro Okamoto
- Department of Morphological Brain Science, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroshi Kameda
- Department of Physiology, Teikyo University School of Medicine, Tokyo, Japan
| | - Fumino Fujiyama
- Laboratory of Neural Circuitry, Graduate School of Brain Science, Doshisha University, Kyotanabe, Japan
| |
Collapse
|
45
|
Astrocytes and presynaptic plasticity in the striatum: Evidence and unanswered questions. Brain Res Bull 2018; 136:17-25. [DOI: 10.1016/j.brainresbull.2017.01.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 12/30/2016] [Accepted: 01/02/2017] [Indexed: 02/03/2023]
|
46
|
Burke DA, Rotstein HG, Alvarez VA. Striatal Local Circuitry: A New Framework for Lateral Inhibition. Neuron 2017; 96:267-284. [PMID: 29024654 DOI: 10.1016/j.neuron.2017.09.019] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 08/09/2017] [Accepted: 09/12/2017] [Indexed: 12/01/2022]
Abstract
This Perspective will examine the organization of intrastriatal circuitry, review recent findings in this area, and discuss how the pattern of connectivity between striatal neurons might give rise to the behaviorally observed synergism between the direct/indirect pathway neurons. The emphasis of this Perspective is on the underappreciated role of lateral inhibition between striatal projection cells in controlling neuronal firing and shaping the output of this circuit. We review some classic studies in combination with more recent anatomical and functional findings to lay out a framework for an updated model of the intrastriatal lateral inhibition, where we explore its contribution to the formation of functional units of processing and the integration and filtering of inputs to generate motor patterns and learned behaviors.
Collapse
Affiliation(s)
- Dennis A Burke
- Laboratory on Neurobiology of Compulsive Behaviors, Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA; Department of Neuroscience, Brown University, Providence, Providence, RI 02912, USA
| | - Horacio G Rotstein
- Federated Department of Biological Sciences, New Jersey Institute of Technology and Rutgers University, Newark, NJ 07102, USA; Institute for Brain and Neuroscience Research, New Jersey Institute of Technology, Newark, NJ 07102, USA
| | - Veronica A Alvarez
- Laboratory on Neurobiology of Compulsive Behaviors, Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA; Intramural Research Program, National Institute on Drug Abuse, NIH, Baltimore, MD 21224, USA.
| |
Collapse
|
47
|
Zhai S, Tanimura A, Graves SM, Shen W, Surmeier DJ. Striatal synapses, circuits, and Parkinson's disease. Curr Opin Neurobiol 2017; 48:9-16. [PMID: 28843800 DOI: 10.1016/j.conb.2017.08.004] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 08/03/2017] [Indexed: 11/30/2022]
Abstract
The striatum is a hub in the basal ganglia circuitry controlling goal directed actions and habits. The loss of its dopaminergic (DAergic) innervation in Parkinson's disease (PD) disrupts the ability of the two principal striatal projection systems to respond appropriately to cortical and thalamic signals, resulting in the hypokinetic features of the disease. New tools to study brain circuitry have led to significant advances in our understanding of striatal circuits and how they adapt in PD models. This short review summarizes some of these recent studies and the gaps that remain to be filled.
Collapse
Affiliation(s)
- Shenyu Zhai
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Asami Tanimura
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Steven M Graves
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Weixing Shen
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - D James Surmeier
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
48
|
Assous M, Kaminer J, Shah F, Garg A, Koós T, Tepper JM. Differential processing of thalamic information via distinct striatal interneuron circuits. Nat Commun 2017; 8:15860. [PMID: 28604688 PMCID: PMC5477498 DOI: 10.1038/ncomms15860] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 05/03/2017] [Indexed: 11/23/2022] Open
Abstract
Recent discoveries of striatal GABAergic interneurons require a new conceptualization of the organization of intrastriatal circuitry and their cortical and thalamic inputs. We investigated thalamic inputs to the two populations of striatal neuropeptide Y (NPY) interneurons, plateau low threshold spike (PLTS) and NPY-neurogliaform (NGF) cells. Optogenetic activation of parafascicular inputs evokes suprathreshold monosynaptic glutamatergic excitation in NGF interneurons and a disynaptic, nicotinic excitation through cholinergic interneurons. In contrast, the predominant response of PLTS interneurons is a disynaptic inhibition dependent on thalamic activation of striatal tyrosine hydroxylase interneurons (THINs). In contrast, THINs do not innervate NGF or fast spiking interneurons, showing significant specificity in THINs outputs. Chemospecific ablation of THINs impairs prepulse inhibition of the acoustic startle response suggesting an important behavioural role of this disynaptic pathway. Our findings demonstrate that the impact of the parafascicular nucleus on striatal activity and some related behaviour critically depend on synaptic interactions within interneuronal circuits. The responses of striatal GABAergic interneurons to thalamic inputs are not well characterised. Here, the authors demonstrate that complex intrastriatal circuitry is responsible for thalamic-evoked monosynaptic and disynaptic excitation in NPY-NGF interneurons but a disynaptic inhibition in the NPY-PLTS.
Collapse
Affiliation(s)
- Maxime Assous
- Center for Molecular and Behavioral Neuroscience, Rutgers University, Newark, New Jersey 07102, USA
| | - Jaime Kaminer
- Center for Molecular and Behavioral Neuroscience, Rutgers University, Newark, New Jersey 07102, USA
| | - Fulva Shah
- Center for Molecular and Behavioral Neuroscience, Rutgers University, Newark, New Jersey 07102, USA
| | - Arpan Garg
- Center for Molecular and Behavioral Neuroscience, Rutgers University, Newark, New Jersey 07102, USA
| | - Tibor Koós
- Center for Molecular and Behavioral Neuroscience, Rutgers University, Newark, New Jersey 07102, USA
| | - James M Tepper
- Center for Molecular and Behavioral Neuroscience, Rutgers University, Newark, New Jersey 07102, USA
| |
Collapse
|
49
|
Yamanaka K, Hori Y, Minamimoto T, Yamada H, Matsumoto N, Enomoto K, Aosaki T, Graybiel AM, Kimura M. Roles of centromedian parafascicular nuclei of thalamus and cholinergic interneurons in the dorsal striatum in associative learning of environmental events. J Neural Transm (Vienna) 2017; 125:501-513. [PMID: 28324169 DOI: 10.1007/s00702-017-1713-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 03/15/2017] [Indexed: 12/20/2022]
Abstract
The thalamus provides a massive input to the striatum, but despite accumulating evidence, the functions of this system remain unclear. It is known, however, that the centromedian (CM) and parafascicular (Pf) nuclei of the thalamus can strongly influence particular striatal neuron subtypes, notably including the cholinergic interneurons of the striatum (CINs), key regulators of striatal function. Here, we highlight the thalamostriatal system through the CM-Pf to striatal CINs. We consider how, by virtue of the direct synaptic connections of the CM and PF, their neural activity contributes to the activity of CINs and striatal projection neurons (SPNs). CM-Pf neurons are strongly activated at sudden changes in behavioral context, such as switches in action-outcome contingency or sequence of behavioral requirements, suggesting that their activity may represent change of context operationalized as associability. Striatal CINs, on the other hand, acquire and loose responses to external events associated with particular contexts. In light of this physiological evidence, we propose a hypothesis of the CM-Pf-CINs system, suggesting that it augments associative learning by generating an associability signal and promotes reinforcement learning guided by reward prediction error signals from dopamine-containing neurons. We discuss neuronal circuit and synaptic organizations based on in vivo/in vitro studies that we suppose to underlie our hypothesis. Possible implications of CM-Pf-CINs dysfunction (or degeneration) in brain diseases are also discussed by focusing on Parkinson's disease.
Collapse
Affiliation(s)
- Ko Yamanaka
- Brain Science Institute, Tamagawa University, Machida, Tokyo, Japan.,Department of Physiology, Kyoto Prefectural University of Medicine, Kyoto, Japan.,Department of Physiology, Faculty of Health and Sports Science, Juntendo University, Chiba, Japan
| | - Yukiko Hori
- Department of Physiology, Kyoto Prefectural University of Medicine, Kyoto, Japan.,Department of Functional Brain Imaging, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Takafumi Minamimoto
- Department of Physiology, Kyoto Prefectural University of Medicine, Kyoto, Japan.,Department of Functional Brain Imaging, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Hiroshi Yamada
- Department of Physiology, Kyoto Prefectural University of Medicine, Kyoto, Japan.,Division of Biomedical Science, Faculty of Medicine, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Naoyuki Matsumoto
- Department of Physiology, Kyoto Prefectural University of Medicine, Kyoto, Japan.,Department of Food and Health Sciences, Faculty of Environmental and Symbiotic Sciences, Prefectural University of Kumamoto, Kumamoto, Japan
| | - Kazuki Enomoto
- Brain Science Institute, Tamagawa University, Machida, Tokyo, Japan.,Department of Physiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Toshihiko Aosaki
- Neurophysiology Research Group, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Ann M Graybiel
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Minoru Kimura
- Brain Science Institute, Tamagawa University, Machida, Tokyo, Japan. .,Department of Physiology, Kyoto Prefectural University of Medicine, Kyoto, Japan.
| |
Collapse
|
50
|
Tanimura A, Lim SAO, Aceves Buendia JDJ, Goldberg JA, Surmeier DJ. Cholinergic Interneurons Amplify Corticostriatal Synaptic Responses in the Q175 Model of Huntington's Disease. Front Syst Neurosci 2016; 10:102. [PMID: 28018188 PMCID: PMC5159611 DOI: 10.3389/fnsys.2016.00102] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 12/02/2016] [Indexed: 01/18/2023] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disorder characterized by deficits in movement control that are widely viewed as stemming from pathophysiological changes in the striatum. Giant, aspiny cholinergic interneurons (ChIs) are key elements in the striatal circuitry controlling movement, but whether their physiological properties are intact in the HD brain is unclear. To address this issue, the synaptic properties of ChIs were examined using optogenetic approaches in the Q175 mouse model of HD. In ex vivo brain slices, synaptic facilitation at thalamostriatal synapses onto ChIs was reduced in Q175 mice. The alteration in thalamostriatal transmission was paralleled by an increased response to optogenetic stimulation of cortical axons, enabling these inputs to more readily induce burst-pause patterns of activity in ChIs. This adaptation was dependent upon amplification of cortically evoked responses by a post-synaptic upregulation of voltage-dependent Na+ channels. This upregulation also led to an increased ability of somatic spikes to invade ChI dendrites. However, there was not an alteration in the basal pacemaking rate of ChIs, possibly due to increased availability of Kv4 channels. Thus, there is a functional "re-wiring" of the striatal networks in Q175 mice, which results in greater cortical control of phasic ChI activity, which is widely thought to shape the impact of salient stimuli on striatal action selection.
Collapse
Affiliation(s)
- Asami Tanimura
- Department of Physiology, Feinberg School of Medicine, Northwestern University Chicago, IL, USA
| | - Sean Austin O Lim
- Department of Physiology, Feinberg School of Medicine, Northwestern University Chicago, IL, USA
| | - Jose de Jesus Aceves Buendia
- Department of Medical Neurobiology, Institute of Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem Jerusalem, Israel
| | - Joshua A Goldberg
- Department of Medical Neurobiology, Institute of Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem Jerusalem, Israel
| | - D James Surmeier
- Department of Physiology, Feinberg School of Medicine, Northwestern University Chicago, IL, USA
| |
Collapse
|