1
|
McKeon PN, Bunce GW, Patton MH, Chen R, Mathur BN. Cortical control of striatal fast-spiking interneuron synchrony. J Physiol 2022; 600:2189-2202. [PMID: 35332539 PMCID: PMC9058232 DOI: 10.1113/jp282850] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/16/2022] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Electrical synapses between striatal fast-spiking interneurons in adult mice occur in ∼8% of assayed pairs. Coincident, convergent cortical input onto fast-spiking interneurons significantly contributes to fast-spiking interneuron synchrony Electrical synapses between fast-spiking interneurons provide only minor enhancement of fast-spiking interneuron synchrony. These results suggest a mechanism by which adult mouse fast-spiking interneurons of the striatum synchronize in the face of declining expression of the electrical synapse-forming connexin-36 protein. ABSTRACT Inhibitory fast-spiking interneurons in the dorsal striatum regulate actions and action strategies, including habits. Fast-spiking interneurons are widely believed to synchronize their firing due to the electrical synapses formed between these neurons. However, neuronal modeling data suggest convergent cortical input may also drive synchrony in fast-spiking interneuron networks. To better understand how fast-spiking interneuron synchrony arises, we performed dual whole-cell patch clamp electrophysiology experiments to inform a simple Bayesian network modeling cortico-fast-spiking interneuron circuitry. Dual whole-cell patch clamp electrophysiology revealed that while responsivity to corticostriatal input activation was high in fast-spiking interneurons, few of these neurons exhibited electrical coupling in adult mice. In simulations of a cortico-fast-spiking interneuron network informed by these data, the degree of glutamatergic cortical convergence onto fast-spiking interneurons significantly increased fast-spiking interneuron synchronization while manipulations of electrical coupling between these neurons exerted relatively little impact. These results suggest that the primary source of functional coordination of fast-spiking interneuron activity in adulthood arises from convergent corticostriatal input activation. Abstract figure legend Dual whole-cell patch clamp recordings of dorsal striatal fast-spiking interneurons (FSIs; red circles) rarely (8 percentage) form electrical synapses with other FSIs in adult mouse. In a two-layer in silico model of cortical pyramidal neuron (gray triangles) input to FSIs using empirically defined cortico-FSI synaptic weights, synchronous FSI-FSI activity (in the absence of abundant electrical synapses) is achievable by convergent cortical pyramidal excitation of FSIs. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Paige N McKeon
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Garrett W Bunce
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mary H Patton
- St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Rong Chen
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Brian N Mathur
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
2
|
Zavalin K, Hassan A, Fu C, Delpire E, Lagrange AH. Loss of KCC2 in GABAergic Neurons Causes Seizures and an Imbalance of Cortical Interneurons. Front Mol Neurosci 2022; 15:826427. [PMID: 35370549 PMCID: PMC8966887 DOI: 10.3389/fnmol.2022.826427] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/21/2022] [Indexed: 11/24/2022] Open
Abstract
K-Cl transporter KCC2 is an important regulator of neuronal development and neuronal function at maturity. Through its canonical transporter role, KCC2 maintains inhibitory responses mediated by γ-aminobutyric acid (GABA) type A receptors. During development, late onset of KCC2 transporter activity defines the period when depolarizing GABAergic signals promote a wealth of developmental processes. In addition to its transporter function, KCC2 directly interacts with a number of proteins to regulate dendritic spine formation, cell survival, synaptic plasticity, neuronal excitability, and other processes. Either overexpression or loss of KCC2 can lead to abnormal circuit formation, seizures, or even perinatal death. GABA has been reported to be especially important for driving migration and development of cortical interneurons (IN), and we hypothesized that properly timed onset of KCC2 expression is vital to this process. To test this hypothesis, we created a mouse with conditional knockout of KCC2 in Dlx5-lineage neurons (Dlx5 KCC2 cKO), which targets INs and other post-mitotic GABAergic neurons in the forebrain starting during embryonic development. While KCC2 was first expressed in the INs of layer 5 cortex, perinatal IN migrations and laminar localization appeared to be unaffected by the loss of KCC2. Nonetheless, the mice had early seizures, failure to thrive, and premature death in the second and third weeks of life. At this age, we found an underlying change in IN distribution, including an excess number of somatostatin neurons in layer 5 and a decrease in parvalbumin-expressing neurons in layer 2/3 and layer 6. Our research suggests that while KCC2 expression may not be entirely necessary for early IN migration, loss of KCC2 causes an imbalance in cortical interneuron subtypes, seizures, and early death. More work will be needed to define the specific cellular basis for these findings, including whether they are due to abnormal circuit formation versus the sequela of defective IN inhibition.
Collapse
Affiliation(s)
- Kirill Zavalin
- Department of Neurology, School of Medicine, Vanderbilt University, Nashville, TN, United States
| | - Anjana Hassan
- Department of Neurology, School of Medicine, Vanderbilt University, Nashville, TN, United States
| | - Cary Fu
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Eric Delpire
- Department of Anesthesiology, School of Medicine, Vanderbilt University, Nashville, TN, United States
| | - Andre H. Lagrange
- Department of Neurology, School of Medicine, Vanderbilt University, Nashville, TN, United States,Department of Neurology, Tennessee Valley Healthcare – Veterans Affairs (TVH VA), Medical Center, Nashville, TN, United States,*Correspondence: Andre H. Lagrange,
| |
Collapse
|
3
|
Godoy LD, Prizon T, Rossignoli MT, Leite JP, Liberato JL. Parvalbumin Role in Epilepsy and Psychiatric Comorbidities: From Mechanism to Intervention. Front Integr Neurosci 2022; 16:765324. [PMID: 35250498 PMCID: PMC8891758 DOI: 10.3389/fnint.2022.765324] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 01/24/2022] [Indexed: 12/22/2022] Open
Abstract
Parvalbumin is a calcium-binding protein present in inhibitory interneurons that play an essential role in regulating many physiological processes, such as intracellular signaling and synaptic transmission. Changes in parvalbumin expression are deeply related to epilepsy, which is considered one of the most disabling neuropathologies. Epilepsy is a complex multi-factor group of disorders characterized by periods of hypersynchronous activity and hyperexcitability within brain networks. In this scenario, inhibitory neurotransmission dysfunction in modulating excitatory transmission related to the loss of subsets of parvalbumin-expressing inhibitory interneuron may have a prominent role in disrupted excitability. Some studies also reported that parvalbumin-positive interneurons altered function might contribute to psychiatric comorbidities associated with epilepsy, such as depression, anxiety, and psychosis. Understanding the epileptogenic process and comorbidities associated with epilepsy have significantly advanced through preclinical and clinical investigation. In this review, evidence from parvalbumin altered function in epilepsy and associated psychiatric comorbidities were explored with a translational perspective. Some advances in potential therapeutic interventions are highlighted, from current antiepileptic and neuroprotective drugs to cutting edge modulation of parvalbumin subpopulations using optogenetics, designer receptors exclusively activated by designer drugs (DREADD) techniques, transcranial magnetic stimulation, genome engineering, and cell grafting. Creating new perspectives on mechanisms and therapeutic strategies is valuable for understanding the pathophysiology of epilepsy and its psychiatric comorbidities and improving efficiency in clinical intervention.
Collapse
Affiliation(s)
- Lívea Dornela Godoy
- Department of Psychology, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Tamiris Prizon
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Matheus Teixeira Rossignoli
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - João Pereira Leite
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- João Pereira Leite,
| | - José Luiz Liberato
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- *Correspondence: José Luiz Liberato,
| |
Collapse
|
4
|
Assous M. Striatal cholinergic transmission. Focus on nicotinic receptors' influence in striatal circuits. Eur J Neurosci 2021; 53:2421-2442. [PMID: 33529401 PMCID: PMC8161166 DOI: 10.1111/ejn.15135] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 01/25/2021] [Accepted: 01/27/2021] [Indexed: 12/11/2022]
Abstract
The critical role of acetylcholine (ACh) in the basal ganglia is evident from the effect of cholinergic agents in patients suffering from several related neurological disorders, such as Parkinson's disease, Tourette syndrome, or dystonia. The striatum possesses the highest density of ACh markers in the basal ganglia underlying the importance of ACh in this structure. Striatal cholinergic interneurons (CINs) are responsible for the bulk of striatal ACh, although extrinsic cholinergic afferents from brainstem structures may also play a role. CINs are tonically active, and synchronized pause in their activity occurs following the presentation of salient stimuli during behavioral conditioning. However, the synaptic mechanisms involved are not fully understood in this physiological response. ACh modulates striatal circuits by acting on muscarinic and nicotinic receptors existing in several combinations both presynaptically and postsynaptically. While the effects of ACh in the striatum through muscarinic receptors have received particular attention, nicotinic receptors function has been less studied. Here, after briefly reviewing relevant results regarding muscarinic receptors expression and function, I will focus on striatal nicotinic receptor expressed presynaptically on glutamatergic and dopaminergic afferents and postsynaptically on diverse striatal interneurons populations. I will also review recent evidence suggesting the involvement of different GABAergic sources in two distinct nicotinic-receptor-mediated striatal circuits: the disynaptic inhibition of striatal projection neurons and the recurrent inhibition among CINs. A better understanding of striatal nicotinic receptors expression and function may help to develop targeted pharmacological interventions to treat brain disorders such as Parkinson's disease, Tourette syndrome, dystonia, or nicotine addiction.
Collapse
Affiliation(s)
- Maxime Assous
- Center for Molecular and Behavioral Neuroscience, Rutgers, the State University of New Jersey, Newark, NJ, USA
| |
Collapse
|
5
|
Patton MS, Heckman M, Kim C, Mu C, Mathur BN. Compulsive alcohol consumption is regulated by dorsal striatum fast-spiking interneurons. Neuropsychopharmacology 2021; 46:351-359. [PMID: 32663841 PMCID: PMC7852608 DOI: 10.1038/s41386-020-0766-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 06/22/2020] [Accepted: 07/08/2020] [Indexed: 11/09/2022]
Abstract
Compulsive alcohol consumption is a core, treatment-resistant feature of alcohol use disorder. The dorsomedial and dorsolateral striatum support goal-directed and habitual action strategies, respectively. How ethanol targets dorsolateral striatum to drive compulsive consumption is poorly understood. Parvalbumin-expressing striatal fast-spiking interneurons comprise ~1% of the total neuronal striatal population, are enriched dorsolaterally and are functionally modulated by ethanol. To test whether fast-spiking interneurons are necessary for the development of compulsive ethanol consumption, we selectively ablated these neurons in adult male and female C57BL/6 J mice undergoing a voluntary chronic intermittent ethanol consumption paradigm followed by a compulsive ethanol drinking assay. Fast-spiking interneuron ablation curtailed the development of organized ethanol lick sequence behavior, reduced ethanol consumption, and abrogated compulsive consumption of ethanol with the added bitterant quinine. In contrast, fast-spiking interneuron ablation did not affect any index of water or sucrose consumption. These data causally implicate the minority striatal fast-spiking interneuron population as a key component of compulsive ethanol consumption.
Collapse
Affiliation(s)
- Michael S. Patton
- grid.411024.20000 0001 2175 4264Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Morgan Heckman
- grid.411024.20000 0001 2175 4264Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Cecelia Kim
- grid.411024.20000 0001 2175 4264Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Chaoqi Mu
- grid.411024.20000 0001 2175 4264Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Brian N. Mathur
- grid.411024.20000 0001 2175 4264Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| |
Collapse
|
6
|
Prior Cocaine Use Alters the Normal Evolution of Information Coding in Striatal Ensembles during Value-Guided Decision-Making. J Neurosci 2020; 41:342-353. [PMID: 33219006 DOI: 10.1523/jneurosci.1755-20.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 11/11/2020] [Accepted: 11/16/2020] [Indexed: 12/31/2022] Open
Abstract
Substance use disorders (SUDs) are characterized by maladaptive behavior. The ability to properly adjust behavior according to changes in environmental contingencies necessitates the interlacing of existing memories with updated information. This can be achieved by assigning learning in different contexts to compartmentalized "states." Though not often framed this way, the maladaptive behavior observed in individuals with SUDs may result from a failure to properly encode states because of drug-induced neural alterations. Previous studies found that the dorsomedial striatum (DMS) is important for behavioral flexibility and state encoding, suggesting the DMS may be an important substrate for these effects. Here, we recorded DMS neural activity in cocaine-experienced male rats during a decision-making task where blocks of trials represented distinct states to probe whether the encoding of state and state-related information is affected by prior drug exposure. We found that DMS medium spiny neurons (MSNs) and fast-spiking interneurons (FSIs) encoded such information and that prior cocaine experience disrupted the evolution of representations both within trials and across recording sessions. Specifically, DMS MSNs and FSIs from cocaine-experienced rats demonstrated higher classification accuracy of trial-specific rules, defined by response direction and value, compared with those drawn from sucrose-experienced rats, and these overly strengthened trial-type representations were related to slower switching behavior and reaction times. These data show that prior cocaine experience paradoxically increases the encoding of state-specific information and rules in the DMS and suggest a model in which abnormally specific and persistent representation of rules throughout trials in DMS slows value-based decision-making in well trained subjects.SIGNIFICANCE STATEMENT Substance use disorders (SUDs) may result from a failure to properly encode rules guiding situationally appropriate behavior. The dorsomedial striatum (DMS) is thought to be important for such behavioral flexibility and encoding that defines the situation or "state." This suggests that the DMS may be an important substrate for the maladaptive behavior observed in SUDs. In the current study, we show that prior cocaine experience results in over-encoding of state-specific information and rules in the DMS, which may impair normal adaptive decision-making in the task, akin to what is observed in SUDs.
Collapse
|
7
|
Hjorth JJJ, Kozlov A, Carannante I, Frost Nylén J, Lindroos R, Johansson Y, Tokarska A, Dorst MC, Suryanarayana SM, Silberberg G, Hellgren Kotaleski J, Grillner S. The microcircuits of striatum in silico. Proc Natl Acad Sci U S A 2020; 117:9554-9565. [PMID: 32321828 PMCID: PMC7197017 DOI: 10.1073/pnas.2000671117] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The basal ganglia play an important role in decision making and selection of action primarily based on input from cortex, thalamus, and the dopamine system. Their main input structure, striatum, is central to this process. It consists of two types of projection neurons, together representing 95% of the neurons, and 5% of interneurons, among which are the cholinergic, fast-spiking, and low threshold-spiking subtypes. The membrane properties, soma-dendritic shape, and intrastriatal and extrastriatal synaptic interactions of these neurons are quite well described in the mouse, and therefore they can be simulated in sufficient detail to capture their intrinsic properties, as well as the connectivity. We focus on simulation at the striatal cellular/microcircuit level, in which the molecular/subcellular and systems levels meet. We present a nearly full-scale model of the mouse striatum using available data on synaptic connectivity, cellular morphology, and electrophysiological properties to create a microcircuit mimicking the real network. A striatal volume is populated with reconstructed neuronal morphologies with appropriate cell densities, and then we connect neurons together based on appositions between neurites as possible synapses and constrain them further with available connectivity data. Moreover, we simulate a subset of the striatum involving 10,000 neurons, with input from cortex, thalamus, and the dopamine system, as a proof of principle. Simulation at this biological scale should serve as an invaluable tool to understand the mode of operation of this complex structure. This platform will be updated with new data and expanded to simulate the entire striatum.
Collapse
Affiliation(s)
- J J Johannes Hjorth
- Science for Life Laboratory, School of Electrical Engeneering and Computer Science, Royal Institute of Technology, SE-10044 Stockholm, Sweden
| | - Alexander Kozlov
- Science for Life Laboratory, School of Electrical Engeneering and Computer Science, Royal Institute of Technology, SE-10044 Stockholm, Sweden
- Department of Neuroscience, Karolinska Institutet, SE-17165 Stockholm
| | - Ilaria Carannante
- Science for Life Laboratory, School of Electrical Engeneering and Computer Science, Royal Institute of Technology, SE-10044 Stockholm, Sweden
| | | | - Robert Lindroos
- Department of Neuroscience, Karolinska Institutet, SE-17165 Stockholm
| | - Yvonne Johansson
- Department of Neuroscience, Karolinska Institutet, SE-17165 Stockholm
| | - Anna Tokarska
- Department of Neuroscience, Karolinska Institutet, SE-17165 Stockholm
| | - Matthijs C Dorst
- Department of Neuroscience, Karolinska Institutet, SE-17165 Stockholm
| | | | - Gilad Silberberg
- Department of Neuroscience, Karolinska Institutet, SE-17165 Stockholm
| | - Jeanette Hellgren Kotaleski
- Science for Life Laboratory, School of Electrical Engeneering and Computer Science, Royal Institute of Technology, SE-10044 Stockholm, Sweden;
- Department of Neuroscience, Karolinska Institutet, SE-17165 Stockholm
| | - Sten Grillner
- Department of Neuroscience, Karolinska Institutet, SE-17165 Stockholm
| |
Collapse
|
8
|
Duhne M, Lara‐González E, Laville A, Padilla‐Orozco M, Ávila‐Cascajares F, Arias‐García M, Galarraga E, Bargas J. Activation of parvalbumin‐expressing neurons reconfigures neuronal ensembles in murine striatal microcircuits. Eur J Neurosci 2020; 53:2149-2164. [DOI: 10.1111/ejn.14670] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 12/23/2019] [Accepted: 01/02/2020] [Indexed: 12/30/2022]
Affiliation(s)
- Mariana Duhne
- División Neurociencias Instituto de Fisiología Celular Universidad Nacional Autónoma de México México City Mexico
| | - Esther Lara‐González
- División Neurociencias Instituto de Fisiología Celular Universidad Nacional Autónoma de México México City Mexico
- Facultad de Ciencias Químicas Benemérita Universidad Autónoma de Puebla Puebla Mexico
| | - Antonio Laville
- División Neurociencias Instituto de Fisiología Celular Universidad Nacional Autónoma de México México City Mexico
| | - Montserrat Padilla‐Orozco
- División Neurociencias Instituto de Fisiología Celular Universidad Nacional Autónoma de México México City Mexico
| | - Fatima Ávila‐Cascajares
- División Neurociencias Instituto de Fisiología Celular Universidad Nacional Autónoma de México México City Mexico
| | - Mario Arias‐García
- División Neurociencias Instituto de Fisiología Celular Universidad Nacional Autónoma de México México City Mexico
| | - Elvira Galarraga
- División Neurociencias Instituto de Fisiología Celular Universidad Nacional Autónoma de México México City Mexico
| | - José Bargas
- División Neurociencias Instituto de Fisiología Celular Universidad Nacional Autónoma de México México City Mexico
| |
Collapse
|
9
|
Baaske MK, Kramer ER, Meka DP, Engler G, Engel AK, Moll CKE. Parkin deficiency perturbs striatal circuit dynamics. Neurobiol Dis 2020; 137:104737. [PMID: 31923460 DOI: 10.1016/j.nbd.2020.104737] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 12/16/2019] [Accepted: 01/05/2020] [Indexed: 01/09/2023] Open
Abstract
Loss-of-function mutations in the parkin-encoding PARK2 gene are a frequent cause of young-onset, autosomal recessive Parkinson's disease (PD). Parkin knockout mice have no nigro-striatal neuronal loss but exhibit abnormalities of striatal dopamine transmission and cortico-striatal synaptic function. How these predegenerative changes observed in vitro affect neural dynamics at the intact circuit level, however, remains hitherto elusive. Here, we recorded from motor cortex, striatum and globus pallidus (GP) of anesthetized parkin-deficient mice to assess cortex-basal ganglia circuit dynamics and to dissect cell type-specific functional connectivity in the presymptomatic phase of genetic PD. While ongoing activity of presumed striatal spiny projection neurons and their downstream counterparts in the GP was not different from controls, parkin deficiency had a differential impact on striatal interneurons: In parkin-mutant mice, tonically active neurons displayed elevated activity levels. Baseline firing rates of transgenic striatal fast spiking interneurons (FSI), on the contrary, were reduced and the correlational structure of the FSI microcircuitry was disrupted. The entire transgenic striatal microcircuit showed enhanced and phase-shifted phase coupling to slow (1-3 Hz) cortical population oscillations. Unexpectedly, local field potentials recorded from striatum and GP of parkin-mutant mice robustly displayed amplified beta oscillations (~22 Hz), phase-coupled to cortex. Parkin deficiency selectively increased spike-field coupling of FSIs to beta oscillations. Our findings suggest that loss of parkin function leads to amplifications of synchronized cortico-striatal oscillations and an intrastriatal reconfiguration of interneuronal circuits. This presymptomatic disarrangement of dynamic functional connectivity may precede nigro-striatal neurodegeneration and predispose to imbalance of striatal outflow accompanying symptomatic PD.
Collapse
Affiliation(s)
- Magdalena K Baaske
- Department of Neurophysiology and Pathophysiology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; Institute of Neurogenetics, University of Lübeck, 23562 Lübeck, Germany; Department of Neurology, University of Lübeck, 23538 Lübeck, Germany.
| | - Edgar R Kramer
- Center of Molecular Neurobiology, 20251 Hamburg, Germany; Institute of Translational and Stratified Medicine, University of Plymouth, Plymouth PL6 8BU, UK
| | | | - Gerhard Engler
- Department of Neurophysiology and Pathophysiology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Andreas K Engel
- Department of Neurophysiology and Pathophysiology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Christian K E Moll
- Department of Neurophysiology and Pathophysiology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| |
Collapse
|
10
|
Nucleus Accumbens Fast-Spiking Interneurons Constrain Impulsive Action. Biol Psychiatry 2019; 86:836-847. [PMID: 31471038 PMCID: PMC6823148 DOI: 10.1016/j.biopsych.2019.07.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 07/02/2019] [Accepted: 07/02/2019] [Indexed: 11/23/2022]
Abstract
BACKGROUND The nucleus accumbens (NAc) controls multiple facets of impulsivity but is a heterogeneous brain region with diverse microcircuitry. Prior literature links impulsive behavior in rodents to gamma-aminobutyric acid signaling in the NAc. Here, we studied the regulation of impulsive behavior by fast-spiking interneurons (FSIs), a strong source of gamma-aminobutyric acid-mediated synaptic inhibition in the NAc. METHODS Male and female transgenic mice expressing Cre recombinase in FSIs allowed us to identify these sparsely distributed cells in the NAc. We used a 5-choice serial reaction time task to measure both impulsive action and sustained attention. During the 5-choice serial reaction time task, we monitored FSI activity with fiber photometry calcium imaging and manipulated FSI activity with chemogenetic and optogenetic methodology. We used electrophysiology, optogenetics, and fluorescent in situ hybridization to confirm these methods were robust and specific to FSIs. RESULTS In mice performing the 5-choice serial reaction time task, NAc FSIs showed sustained activity on trials ending with correct responses, but FSI activity declined over time on trials ending with premature responses. The number of premature responses increased significantly after sustained chemogenetic inhibition or temporally delimited optogenetic inhibition of NAc FSIs, without any changes in response latencies or general locomotor activity. CONCLUSIONS These experiments provide strong evidence that NAc FSIs constrain impulsive actions, most likely through gamma-aminobutyric acid-mediated synaptic inhibition of medium spiny projection neurons. Our findings may provide insight into the pathophysiology of disorders associated with impulsivity and may inform the development of circuit-based therapeutic interventions.
Collapse
|
11
|
Pettibone JR, Yu JY, Derman RC, Faust TW, Hughes ED, Filipiak WE, Saunders TL, Ferrario CR, Berke JD. Knock-In Rat Lines with Cre Recombinase at the Dopamine D1 and Adenosine 2a Receptor Loci. eNeuro 2019; 6:ENEURO.0163-19.2019. [PMID: 31451604 PMCID: PMC6776791 DOI: 10.1523/eneuro.0163-19.2019] [Citation(s) in RCA: 12] [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: 05/01/2019] [Revised: 06/14/2019] [Accepted: 06/19/2019] [Indexed: 12/31/2022] Open
Abstract
Genetically modified mice have become standard tools in neuroscience research. Our understanding of the basal ganglia in particular has been greatly assisted by BAC mutants with selective transgene expression in striatal neurons forming the direct or indirect pathways. However, for more sophisticated behavioral tasks and larger intracranial implants, rat models are preferred. Furthermore, BAC lines can show variable expression patterns depending upon genomic insertion site. We therefore used CRISPR/Cas9 to generate two novel knock-in rat lines specifically encoding Cre recombinase immediately after the dopamine D1 receptor (Drd1a) or adenosine 2a receptor (Adora2a) loci. Here, we validate these lines using in situ hybridization and viral vector mediated transfection to demonstrate selective, functional Cre expression in the striatal direct and indirect pathways, respectively. We used whole-genome sequencing to confirm the lack of off-target effects and established that both rat lines have normal locomotor activity and learning in simple instrumental and Pavlovian tasks. We expect these new D1-Cre and A2a-Cre rat lines will be widely used to study both normal brain functions and neurological and psychiatric pathophysiology.
Collapse
Affiliation(s)
| | - Jai Y Yu
- Department of Physiology, University of California, San Francisco 94143, CA
| | - Rifka C Derman
- Neuroscience Graduate Program, University of Michigan, Ann Arbor 48109, MI
| | - Thomas W Faust
- Department of Neurology, University of California, San Francisco 94143, CA
| | - Elizabeth D Hughes
- Transgenic Animal Model Core, University of Michigan, Ann Arbor 48109, MI
| | - Wanda E Filipiak
- Transgenic Animal Model Core, University of Michigan, Ann Arbor 48109, MI
| | - Thomas L Saunders
- Transgenic Animal Model Core, University of Michigan, Ann Arbor 48109, MI
- Department of Internal Medicine, University of Michigan, Ann Arbor 48109, MI
| | - Carrie R Ferrario
- Neuroscience Graduate Program, University of Michigan, Ann Arbor 48109, MI
- Department of Pharmacology, University of Michigan, Ann Arbor 48109, MI
| | - Joshua D Berke
- Department of Neurology, University of California, San Francisco 94143, CA
- Department of Psychiatry, University of California, San Francisco 94143, CA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco 94143, CA
- Weill Institute for Neurosciences, University of California, San Francisco 94143, CA
| |
Collapse
|
12
|
Ensemble encoding of action speed by striatal fast-spiking interneurons. Brain Struct Funct 2019; 224:2567-2576. [PMID: 31243530 DOI: 10.1007/s00429-019-01908-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 06/08/2019] [Indexed: 10/26/2022]
Abstract
Striatal fast-spiking interneurons (FSIs) potently inhibit the output neurons of the striatum and, as such, powerfully modulate action learning. Through electrical synaptic coupling, FSIs are theorized to temporally coordinate their activity. This has important implications for their ability to temporally summate inhibition on downstream striatal projection neurons. While some in vivo single-unit electrophysiological recordings of putative FSIs support coordinated firing, others do not. Moreover, it is unclear as to what aspect of action FSIs encode. To address this, we used in vivo calcium imaging of genetically identified FSIs in freely moving mice and applied machine learning analyses to decipher the relationship between FSI activity and movement. We report that FSIs exhibit ensemble activity that encodes the speed of action sub-components, including ambulation and head movements. These results suggest FSI population dynamics fit within a Hebbian model for ensemble inhibition of striatal output guiding action.
Collapse
|
13
|
Corbit VL, Manning EE, Gittis AH, Ahmari SE. Strengthened Inputs from Secondary Motor Cortex to Striatum in a Mouse Model of Compulsive Behavior. J Neurosci 2019; 39:2965-2975. [PMID: 30737313 PMCID: PMC6462450 DOI: 10.1523/jneurosci.1728-18.2018] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 11/19/2018] [Accepted: 12/19/2018] [Indexed: 12/15/2022] Open
Abstract
Hyperactivity in striatum is associated with compulsive behaviors in obsessive-compulsive disorder (OCD) and related illnesses, but it is unclear whether this hyperactivity is due to intrinsic striatal dysfunction or abnormalities in corticostriatal inputs. Understanding the cellular and circuit properties underlying striatal hyperactivity could help inform the optimization of targeted stimulation treatments for compulsive behavior disorders. To investigate the cellular and synaptic abnormalities that may underlie corticostriatal dysfunction relevant to OCD, we used the Sapap3 knock-out (Sapap3-KO) mouse model of compulsive behaviors, which also exhibits hyperactivity in central striatum. Ex vivo electrophysiology in double-transgenic mice was used to assess intrinsic excitability and functional synaptic input in spiny projection neurons (SPNs) and fast-spiking interneurons (FSIs) in central striatum of Sapap3-KOs and wild-type (WT) littermates. While we found no differences in intrinsic excitability of SPNs or FSIs between Sapap3-KOs and WTs, excitatory drive to FSIs was significantly increased in KOs. Contrary to predictions, lateral orbitofrontal cortex-striatal synapses were not responsible for this increased drive; optogenetic stimulation revealed that lateral orbitofrontal cortex input to SPNs was reduced in KOs (∼3-fold) and unchanged in FSIs. However, secondary motor area (M2) postsynaptic responses in central striatum were significantly increased (∼6-fold) in strength and reliability in KOs relative to WTs. These results suggest that increased M2-striatal drive may contribute to both in vivo striatal hyperactivity and compulsive behaviors, and support a potential role for presupplementary/supplementary motor cortical regions in the pathology and treatment of compulsive behavior disorders.SIGNIFICANCE STATEMENT These findings highlight an unexpected contribution of M2 projections to striatal dysfunction in the Sapap3-KO obsessive-compulsive disorder (OCD)-relevant mouse model, with M2 inputs strengthened by at least sixfold onto both spiny projection neurons and fast-spiking interneurons in central striatum. Because M2 is thought to be homologous to presupplementary/supplementary motor areas (pre-SMA/SMA) in humans, regions important for movement preparation and behavioral sequencing, these data are consistent with a model in which increased drive from M2 leads to excessive selection of sequenced motor patterns. Together with observations of hyperactivity in pre-SMA/SMA in both OCD and Tourette syndrome, and evidence that pre-SMA is a potential target for repetitive transcranial magnetic stimulation treatment in OCD, these results support further dissection of the role of M2 in compulsivity.
Collapse
Affiliation(s)
- Victoria L Corbit
- Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, and
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania 15219
| | - Elizabeth E Manning
- Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania 15219
| | - Aryn H Gittis
- Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, and
| | - Susanne E Ahmari
- Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213,
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania 15219
| |
Collapse
|
14
|
Cortico-Striatal Cross-Frequency Coupling and Gamma Genesis Disruptions in Huntington's Disease Mouse and Computational Models. eNeuro 2018; 5:eN-NWR-0210-18. [PMID: 30627632 PMCID: PMC6325534 DOI: 10.1523/eneuro.0210-18.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 10/19/2018] [Accepted: 11/12/2018] [Indexed: 12/16/2022] Open
Abstract
Abnormal gamma band power across cortex and striatum is an important phenotype of Huntington's disease (HD) in both patients and animal models, but neither the origin nor the functional relevance of this phenotype is well understood. Here, we analyzed local field potential (LFP) activity in freely behaving, symptomatic R6/2 and Q175 mouse models and corresponding wild-type (WT) controls. We focused on periods of quiet rest, which show strong γ activity in HD mice. Simultaneous recording from motor cortex and its target area in dorsal striatum in the R6/2 model revealed exaggerated functional coupling over that observed in WT between the phase of delta frequencies (1-4 Hz) in cortex and striatum and striatal amplitude modulation of low γ frequencies (25-55 Hz; i.e., phase-amplitude coupling, PAC), but no evidence that abnormal cortical activity alone can account for the increase in striatal γ power. Both HD mouse models had stronger coupling of γ amplitude to δ phase and more unimodal phase distributions than their WT counterparts. To assess the possible role of striatal fast-spiking interneurons (FSIs) in these phenomena, we developed a computational model based on additional striatal recordings from Q175 mice. Changes in peak γ frequency and power ratio were readily reproduced by our computational model, accounting for several experimental findings reported in the literature. Our results suggest that HD is characterized by both a reorganization of cortico-striatal drive and specific population changes related to intrastriatal synaptic coupling.
Collapse
|
15
|
Fino E, Vandecasteele M, Perez S, Saudou F, Venance L. Region-specific and state-dependent action of striatal GABAergic interneurons. Nat Commun 2018; 9:3339. [PMID: 30131490 PMCID: PMC6104028 DOI: 10.1038/s41467-018-05847-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 07/31/2018] [Indexed: 11/09/2022] Open
Abstract
Striatum processes a wide range of functions including goal-directed behavior and habit formation, respectively encoded by the dorsomedial striatum (DMS) and dorsolateral striatum (DLS). GABAergic feedforward inhibition is known to control the integration of cortical information by striatal projection neurons (SPNs). Here we questioned whether this control is specific between distinct striatal functional territories. Using opto-activation and opto-inhibition of identified GABAergic interneurons, we found that different circuits are engaged in DLS and DMS, both ex vivo and in vivo: while parvalbumin interneurons efficiently control SPNs in DLS, somatostatin interneurons control SPNs in DMS. Moreover, both parvalbumin and somatostatin interneurons use a dual hyperpolarizing/depolarizing effect to control cortical input integration depending on SPN activity state: GABAergic interneurons potently inhibit spiking SPNs while in resting SPNs, they favor cortical activity summation via a depolarizing effect. Our findings establish that striatal GABAergic interneurons exert efficient territory-specific and state-dependent control of SPN activity and functional output.
Collapse
Affiliation(s)
- Elodie Fino
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS UMR7241, INSERM U1050, Paris, 75005, France. .,Université Pierre et Marie Curie, ED 158, Paris Sciences et Lettres, Paris, 75005, France. .,INSERM U1216, Grenoble, 38000, France. .,Grenoble Institute of Neuroscience, Université Grenoble Alpes, Grenoble, 38000, France.
| | - Marie Vandecasteele
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS UMR7241, INSERM U1050, Paris, 75005, France.,Université Pierre et Marie Curie, ED 158, Paris Sciences et Lettres, Paris, 75005, France
| | - Sylvie Perez
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS UMR7241, INSERM U1050, Paris, 75005, France.,Université Pierre et Marie Curie, ED 158, Paris Sciences et Lettres, Paris, 75005, France
| | - Frédéric Saudou
- INSERM U1216, Grenoble, 38000, France.,Grenoble Institute of Neuroscience, Université Grenoble Alpes, Grenoble, 38000, France.,CHU Grenoble Alpes, Grenoble, 38000, France
| | - Laurent Venance
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS UMR7241, INSERM U1050, Paris, 75005, France.,Université Pierre et Marie Curie, ED 158, Paris Sciences et Lettres, Paris, 75005, France
| |
Collapse
|
16
|
Neuronal activity pattern defects in the striatum in awake mouse model of Parkinson’s disease. Behav Brain Res 2018; 341:135-145. [DOI: 10.1016/j.bbr.2017.12.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 12/12/2017] [Accepted: 12/12/2017] [Indexed: 11/23/2022]
|
17
|
Assous M, Tepper JM. Excitatory extrinsic afferents to striatal interneurons and interactions with striatal microcircuitry. Eur J Neurosci 2018; 49:593-603. [PMID: 29480942 DOI: 10.1111/ejn.13881] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 02/20/2018] [Accepted: 02/20/2018] [Indexed: 01/24/2023]
Abstract
The striatum constitutes the main input structure of the basal ganglia and receives two major excitatory glutamatergic inputs, from the cortex and the thalamus. Excitatory cortico- and thalamostriatal connections innervate the principal neurons of the striatum, the spiny projection neurons (SPNs), which constitute the main cellular input as well as the only output of the striatum. In addition, corticostriatal and thalamostriatal inputs also innervate striatal interneurons. Some of these inputs have been very well studied, for example the thalamic innervation of cholinergic interneurons and the cortical innervation of striatal fast-spiking interneurons, but inputs to most other GABAergic interneurons remain largely unstudied, due in part to the relatively recent identification and characterization of many of these interneurons. In this review, we will discuss and reconcile some older as well as more recent data on the extrinsic excitatory inputs to striatal interneurons. We propose that the traditional feed-forward inhibitory model of the cortical input to the fast-spiking interneuron then inhibiting the SPN, often assumed to be the prototype of the main functional organization of striatal interneurons, is incomplete. We provide evidence that the extrinsic innervation of striatal interneurons is not uniform but shows great cell-type specificity. In addition, we will review data showing that striatal interneurons are themselves interconnected in a highly cell-type-specific manner. These data suggest that the impact of the extrinsic inputs on striatal activity critically depends on synaptic interactions within interneuronal circuitry.
Collapse
Affiliation(s)
- Maxime Assous
- Center for Molecular and Behavioral Neuroscience, Rutgers, the State University of New Jersey, 197 University Avenue, Newark, NJ, 07102, USA
| | - James M Tepper
- Center for Molecular and Behavioral Neuroscience, Rutgers, the State University of New Jersey, 197 University Avenue, Newark, NJ, 07102, USA
| |
Collapse
|
18
|
Loss of Balance between Striatal Feedforward Inhibition and Corticostriatal Excitation Leads to Tremor. J Neurosci 2018; 38:1699-1710. [PMID: 29330326 DOI: 10.1523/jneurosci.2821-17.2018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 11/30/2017] [Accepted: 01/05/2018] [Indexed: 11/21/2022] Open
Abstract
Fast-spiking interneurons (FSIs) exert powerful inhibitory control over the striatum and are hypothesized to balance the massive excitatory cortical and thalamic input to this structure. We recorded neuronal activity in the dorsolateral striatum and globus pallidus (GP) concurrently with the detailed movement kinematics of freely behaving female rats before and after selective inhibition of FSI activity using IEM-1460 microinjections. The inhibition led to the appearance of episodic rest tremor in the body part that depended on the somatotopic location of the injection within the striatum. The tremor was accompanied by coherent oscillations in the local field potential (LFP). Individual neuron activity patterns became oscillatory and coherent in the tremor frequency. Striatal neurons, but not GP neurons, displayed additional temporal, nonoscillatory correlations. The subsequent reduction in the corticostriatal input following muscimol injection to the corresponding somatotopic location in the primary motor cortex led to disruption of the tremor and a reduction of the LFP oscillations and individual neuron's phase-locked activity. The breakdown of the normal balance of excitation and inhibition in the striatum has been shown previously to be related to different motor abnormalities. Our results further indicate that the balance between excitatory corticostriatal input and feedforward FSI inhibition is sufficient to break down the striatal decorrelation process and generate oscillations resulting in rest tremor typical of multiple basal ganglia disorders.SIGNIFICANCE STATEMENT Fast-spiking interneurons (FSIs) play a key role in normal striatal processing by exerting powerful inhibitory control over the network. FSI malfunctions have been associated with abnormal processing of information within the striatum that leads to multiple movement disorders. Here, we study the changes in neuronal activity and movement kinematics following selective inhibition of these neurons. The injections led to the appearance of episodic rest tremor, accompanied by coherent oscillations in neuronal activity, which was reversed following corticostriatal inhibition. These results suggest that the balance between corticostriatal excitation and feedforward FSI inhibition is crucial for maintaining the striatal decorrelation process, and that its breakdown leads to the formation of oscillations resulting in rest tremor typical of multiple basal ganglia disorders.
Collapse
|
19
|
Rangel-Barajas C, Rebec GV. Dysregulation of Corticostriatal Connectivity in Huntington's Disease: A Role for Dopamine Modulation. J Huntingtons Dis 2017; 5:303-331. [PMID: 27983564 PMCID: PMC5181679 DOI: 10.3233/jhd-160221] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Aberrant communication between striatum, the main information processing unit of the basal ganglia, and cerebral cortex plays a critical role in the emergence of Huntington’s disease (HD), a fatal monogenetic condition that typically strikes in the prime of life. Although both striatum and cortex undergo substantial cell loss over the course of HD, corticostriatal circuits become dysfunctional long before neurons die. Understanding the dysfunction is key to developing effective strategies for treating a progressively worsening triad of motor, cognitive, and psychiatric symptoms. Cortical output neurons drive striatal activity through the release of glutamate, an excitatory amino acid. Striatal outputs, in turn, release γ-amino butyric acid (GABA) and exert inhibitory control over downstream basal ganglia targets. Ample evidence from transgenic rodent models points to dysregulation of corticostriatal glutamate transmission along with corresponding changes in striatal GABA release as underlying factors in the HD behavioral phenotype. Another contributor is dysregulation of dopamine (DA), a modulator of both glutamate and GABA transmission. In fact, pharmacological manipulation of DA is the only currently available treatment for HD symptoms. Here, we review data from animal models and human patients to evaluate the role of DA in HD, including DA interactions with glutamate and GABA within the context of dysfunctional corticostriatal circuitry.
Collapse
Affiliation(s)
| | - George V. Rebec
- Correspondence to: George V. Rebec, PhD, Department of Psychological and Brain Sciences, Program in
Neuroscience, Indiana University, 1101 E. 10th Street, Bloomington, IN 47405-7007, USA. Tel.: +1 812 855 4832;
Fax: +1 812 855 4520; E-mail:
| |
Collapse
|
20
|
Zheng P, Kozloski J. Striatal Network Models of Huntington's Disease Dysfunction Phenotypes. Front Comput Neurosci 2017; 11:70. [PMID: 28798680 PMCID: PMC5529396 DOI: 10.3389/fncom.2017.00070] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 07/13/2017] [Indexed: 11/17/2022] Open
Abstract
We present a network model of striatum, which generates "winnerless" dynamics typical for a network of sparse, unidirectionally connected inhibitory units. We observe that these dynamics, while interesting and a good match to normal striatal electrophysiological recordings, are fragile. Specifically, we find that randomly initialized networks often show dynamics more resembling "winner-take-all," and relate this "unhealthy" model activity to dysfunctional physiological and anatomical phenotypes in the striatum of Huntington's disease animal models. We report plasticity as a potent mechanism to refine randomly initialized networks and create a healthy winnerless dynamic in our model, and we explore perturbations to a healthy network, modeled on changes observed in Huntington's disease, such as neuron cell death and increased bidirectional connectivity. We report the effect of these perturbations on the conversion risk of the network to an unhealthy state. Finally we discuss the relationship between structural and functional phenotypes observed at the level of simulated network dynamics as a promising means to model disease progression in different patient populations.
Collapse
Affiliation(s)
| | - James Kozloski
- Computational Neuroscience and Multiscale Brain Modeling, Computational Biology Center, IBM Research Division, IBM T. J. Watson Research CenterNew York, NY, United States
| |
Collapse
|
21
|
Buxton D, Bracci E, Overton PG, Gurney K. Striatal Neuropeptides Enhance Selection and Rejection of Sequential Actions. Front Comput Neurosci 2017; 11:62. [PMID: 28798678 PMCID: PMC5529366 DOI: 10.3389/fncom.2017.00062] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 06/27/2017] [Indexed: 12/05/2022] Open
Abstract
The striatum is the primary input nucleus for the basal ganglia, and receives glutamatergic afferents from the cortex. Under the hypothesis that basal ganglia perform action selection, these cortical afferents encode potential “action requests.” Previous studies have suggested the striatum may utilize a mutually inhibitory network of medium spiny neurons (MSNs) to filter these requests so that only those of high salience are selected. However, the mechanisms enabling the striatum to perform clean, rapid switching between distinct actions that form part of a learned action sequence are still poorly understood. Substance P (SP) and enkephalin are neuropeptides co-released with GABA in MSNs preferentially expressing D1 or D2 dopamine receptors respectively. SP has a facilitatory effect on subsequent glutamatergic inputs to target MSNs, while enkephalin has an inhibitory effect. Blocking the action of SP in the striatum is also known to affect behavioral transitions. We constructed phenomenological models of the effects of SP and enkephalin, and integrated these into a hybrid model of basal ganglia comprising a spiking striatal microcircuit and rate–coded populations representing other major structures. We demonstrated that diffuse neuropeptide connectivity enhanced the selection of unordered action requests, and that for true action sequences, where action semantics define a fixed structure, a patterning of the SP connectivity reflecting this ordering enhanced selection of actions presented in the correct sequential order and suppressed incorrect ordering. We also showed that selective pruning of SP connections allowed context–sensitive inhibition of specific undesirable requests that otherwise interfered with selection of an action group. Our model suggests that the interaction of SP and enkephalin enhances the contrast between selection and rejection of action requests, and that patterned SP connectivity in the striatum allows the “chunking” of actions and improves selection of sequences. Efficient execution of action sequences may therefore result from a combination of ordered cortical inputs and patterned neuropeptide connectivity within striatum.
Collapse
Affiliation(s)
- David Buxton
- Adaptive Behaviour Research Group, Department of Psychology, The University of SheffieldSheffield, United Kingdom
| | - Enrico Bracci
- Adaptive Behaviour Research Group, Department of Psychology, The University of SheffieldSheffield, United Kingdom
| | - Paul G Overton
- Adaptive Behaviour Research Group, Department of Psychology, The University of SheffieldSheffield, United Kingdom
| | - Kevin Gurney
- Adaptive Behaviour Research Group, Department of Psychology, The University of SheffieldSheffield, United Kingdom
| |
Collapse
|
22
|
Background activity and visual responsiveness of caudate nucleus neurons in halothane anesthetized and in awake, behaving cats. Neuroscience 2017; 356:182-192. [PMID: 28546109 DOI: 10.1016/j.neuroscience.2017.05.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 05/11/2017] [Accepted: 05/15/2017] [Indexed: 11/20/2022]
Abstract
This study focuses on the important question whether brain activity recorded from anesthetized, paralyzed animals is comparable to that recorded from awake, behaving ones. We compared neuronal activity recorded from the caudate nucleus (CN) of two halothane-anesthetized, paralyzed and two awake, behaving cats. In both models, extracellular recordings were made from the CN during static and dynamic visual stimulation. The anesthesia was maintained during the recordings by a gaseous mixture of air and halothane (1.0%). The behaving animals were trained to perform a visual fixation task. Based on their electrophysiological properties, the recorded CN neurons were separated into three different classes: phasically active (PANs), high firing (HFNs), and tonically active (TANs) neurons. Halothane anesthesia significantly decreased the background activity of the CN neurons in all three classes. The anesthesia had the most remarkable suppressive effect on PANs, where the background activity was consistently under 1 spike/s. The analysis of these responses was almost impossible due to the extremely low activity. The evoked responses during both static and dynamic visual stimulation were obvious in the behaving cats. On the other hand, only weak visual responses were found in some neurons of halothane anesthetized cats. These results show that halothane gas anesthesia has a marked suppressive effect on the feline CN. We suggest that for the purposes of the visual and related multisensory/sensorimotor electrophysiological exploration of the CN, behaving animal models are preferable over anesthetized ones.
Collapse
|
23
|
Belić JJ, Kumar A, Hellgren Kotaleski J. Interplay between periodic stimulation and GABAergic inhibition in striatal network oscillations. PLoS One 2017; 12:e0175135. [PMID: 28384268 PMCID: PMC5383243 DOI: 10.1371/journal.pone.0175135] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 03/21/2017] [Indexed: 11/19/2022] Open
Abstract
Network oscillations are ubiquitous across many brain regions. In the basal ganglia, oscillations are also present at many levels and a wide range of characteristic frequencies have been reported to occur during both health and disease. The striatum, the main input nucleus of the basal ganglia, receives massive glutamatergic inputs from the cortex and is highly susceptible to external oscillations. However, there is limited knowledge about the exact nature of this routing process and therefore, it is of key importance to understand how time-dependent, external stimuli propagate through the striatal circuitry. Using a network model of the striatum and corticostriatal projections, we try to elucidate the importance of specific GABAergic neurons and their interactions in shaping striatal oscillatory activity. Here, we propose that fast-spiking interneurons can perform an important role in transferring cortical oscillations to the striatum especially to those medium spiny neurons that are not directly driven by the cortical oscillations. We show how the activity levels of different populations, the strengths of different inhibitory synapses, degree of outgoing projections of striatal cells, ongoing activity and synchronicity of inputs can influence network activity. These results suggest that the propagation of oscillatory inputs into the medium spiny neuron population is most efficient, if conveyed via the fast-spiking interneurons. Therefore, pharmaceuticals that target fast-spiking interneurons may provide a novel treatment for regaining the spectral characteristics of striatal activity that correspond to the healthy state.
Collapse
Affiliation(s)
- Jovana J. Belić
- Science for Life Laboratory, School of Computer Science and Communication, KTH Royal Institute of Technology, Stockholm, Sweden
- Department of Computational Science and Technology, School of Computer Science and Communication, KTH Royal Institute of Technology, Stockholm, Sweden
- Bernstein Center Freiburg, University of Freiburg, Freiburg, Germany
| | - Arvind Kumar
- Department of Computational Science and Technology, School of Computer Science and Communication, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Jeanette Hellgren Kotaleski
- Science for Life Laboratory, School of Computer Science and Communication, KTH Royal Institute of Technology, Stockholm, Sweden
- Department of Computational Science and Technology, School of Computer Science and Communication, KTH Royal Institute of Technology, Stockholm, Sweden
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
24
|
A Feedforward Inhibitory Circuit Mediated by CB1-Expressing Fast-Spiking Interneurons in the Nucleus Accumbens. Neuropsychopharmacology 2017; 42:1146-1156. [PMID: 27929113 PMCID: PMC5506784 DOI: 10.1038/npp.2016.275] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Revised: 11/18/2016] [Accepted: 12/03/2016] [Indexed: 11/08/2022]
Abstract
The nucleus accumbens (NAc) gates motivated behaviors through the functional output of principle medium spiny neurons (MSNs), whereas dysfunctional output of NAc MSNs contributes to a variety of psychiatric disorders. Fast-spiking interneurons (FSIs) are sparsely distributed throughout the NAc, forming local feedforward inhibitory circuits. It remains elusive how FSI-based feedforward circuits regulate the output of NAc MSNs. Here, we investigated a distinct subpopulation of NAc FSIs that express the cannabinoid receptor type-1 (CB1). Using a combination of paired electrophysiological recordings and pharmacological approaches, we characterized and compared feedforward inhibition of NAc MSNs from CB1+ FSIs and lateral inhibition from recurrent MSN collaterals. We observed that CB1+ FSIs exerted robust inhibitory control over a large percentage of nearby MSNs in contrast to local MSN collaterals that provided only sparse and weak inhibitory input to their neighboring MSNs. Furthermore, CB1+ FSI-mediated feedforward inhibition was preferentially suppressed by endocannabinoid (eCB) signaling, whereas MSN-mediated lateral inhibition was unaffected. Finally, we demonstrated that CB1+ FSI synapses onto MSNs are capable of undergoing experience-dependent long-term depression in a voltage- and eCB-dependent manner. These findings demonstrated that CB1+ FSIs are a major source of local inhibitory control of MSNs and a critical component of the feedforward inhibitory circuits regulating the output of the NAc.
Collapse
|
25
|
Lee K, Holley SM, Shobe JL, Chong NC, Cepeda C, Levine MS, Masmanidis SC. Parvalbumin Interneurons Modulate Striatal Output and Enhance Performance during Associative Learning. Neuron 2017; 93:1451-1463.e4. [PMID: 28334608 PMCID: PMC5386608 DOI: 10.1016/j.neuron.2017.02.033] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 01/09/2017] [Accepted: 02/15/2017] [Indexed: 01/13/2023]
Abstract
The prevailing view is that striatal parvalbumin (PV)-positive interneurons primarily function to downregulate medium spiny projection neuron (MSN) activity via monosynaptic inhibitory signaling. Here, by combining in vivo neural recordings and optogenetics, we unexpectedly find that both suppressing and over-activating PV cells attenuates spontaneous MSN activity. To account for this, we find that, in addition to monosynaptic coupling, PV-MSN interactions are mediated by a competing disynaptic inhibitory circuit involving a variety of neuropeptide Y-expressing interneurons. Next we use optogenetic and chemogenetic approaches to show that dorsolateral striatal PV interneurons influence the initial expression of reward-conditioned responses but that their contribution to performance declines with experience. Consistent with this, we observe with large-scale recordings in behaving animals that the relative contribution of PV cells on MSN activity diminishes with training. Together, this work provides a possible mechanism by which PV interneurons modulate striatal output and selectively enhance performance early in learning.
Collapse
Affiliation(s)
- Kwang Lee
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Sandra M Holley
- Intellectual and Developmental Disabilities Research Center, Brain Research Institute, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Justin L Shobe
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Natalie C Chong
- Neuroscience Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Carlos Cepeda
- Intellectual and Developmental Disabilities Research Center, Brain Research Institute, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Michael S Levine
- Intellectual and Developmental Disabilities Research Center, Brain Research Institute, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Sotiris C Masmanidis
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Brain Research Institute, Integrative Center for Learning and Memory, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
26
|
Representation of the body in the lateral striatum of the freely moving rat: Fast Spiking Interneurons respond to stimulation of individual body parts. Brain Res 2016; 1657:101-108. [PMID: 27914882 DOI: 10.1016/j.brainres.2016.11.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 11/22/2016] [Accepted: 11/28/2016] [Indexed: 11/22/2022]
Abstract
Numerous studies have shown that certain types of striatal interneurons play a crucial role in selection and regulation of striatal output. Striatal Fast-Spiking Interneurons (FSIs) are parvalbumin positive, GABAergic interneurons that constitute less than 1% of the total striatal population. It is becoming increasingly evident that these sparsely distributed neurons exert a strong inhibitory effect on Medium Spiny projection Neurons (MSNs). MSNs in lateral striatum receive direct synaptic input from regions of cortex representing discrete body parts, and show phasic increases in activity during touch or movement of specific body parts. In the present study, we sought to determine whether lateral striatal FSIs identified by their electrophysiological properties, i.e., short-duration spike and fast firing rate (FR), display body part sensitivity similar to that exhibited by MSNs. During video recorded somatosensorimotor exams, each individual body part was stimulated and responses of single neurons were observed and quantified. Individual FSIs displayed patterns of activity related selectively to stimulation of a discrete body part. Most patterns of activity were similar to those exhibited by typical MSNs, but some phasic decreases were observed. These results serve as evidence that some striatal FSIs process information related to discrete body parts and participate in sensorimotor processing by striatal networks that contribute to motor output. STATEMENT OF SIGNIFICANCE Parvalbumin positive, striatal FSIs are hypothesized to play an important role in behavior by inhibiting MSNs. We asked a fundamental question regarding information processed during behavior by FSIs: whether FSIs, which preferentially occupy the sensorimotor portion of the striatum, process activity of discrete body parts. Our finding that they do, in a selective manner similar to MSNs, begins to reveal the types of phasic signals that FSI feed forward to projection neurons during striatal processing of cortical input regarding a specific sensorimotor event. These findings suggest new avenues for testing feed-forward inhibition theory as applied to striatum in naturalistic conditions, such as whether FSI decreases facilitate excitation of MSNs related to the current movement while FSI increases silence MSNs unrelated to the current movement.
Collapse
|
27
|
Garas FN, Shah RS, Kormann E, Doig NM, Vinciati F, Nakamura KC, Dorst MC, Smith Y, Magill PJ, Sharott A. Secretagogin expression delineates functionally-specialized populations of striatal parvalbumin-containing interneurons. eLife 2016; 5. [PMID: 27669410 PMCID: PMC5036963 DOI: 10.7554/elife.16088] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 08/25/2016] [Indexed: 11/13/2022] Open
Abstract
Corticostriatal afferents can engage parvalbumin-expressing (PV+) interneurons to rapidly curtail the activity of striatal projection neurons (SPNs), thus shaping striatal output. Schemes of basal ganglia circuit dynamics generally consider striatal PV+ interneurons to be homogenous, despite considerable heterogeneity in both form and function. We demonstrate that the selective co-expression of another calcium-binding protein, secretagogin (Scgn), separates PV+ interneurons in rat and primate striatum into two topographically-, physiologically- and structurally-distinct cell populations. In rats, these two interneuron populations differed in their firing rates, patterns and relationships with cortical oscillations in vivo. Moreover, the axons of identified PV+/Scgn+ interneurons preferentially targeted the somata of SPNs of the so-called 'direct pathway', whereas PV+/Scgn- interneurons preferentially targeted 'indirect pathway' SPNs. These two populations of interneurons could therefore provide a substrate through which either of the striatal output pathways can be rapidly and selectively inhibited to subsequently mediate the expression of behavioral routines.
Collapse
Affiliation(s)
- Farid N Garas
- Medical Research Council Brain Network Dynamics Unit, Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Rahul S Shah
- Medical Research Council Brain Network Dynamics Unit, Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Eszter Kormann
- Medical Research Council Brain Network Dynamics Unit, Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Natalie M Doig
- Medical Research Council Brain Network Dynamics Unit, Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Federica Vinciati
- Medical Research Council Brain Network Dynamics Unit, Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Kouichi C Nakamura
- Medical Research Council Brain Network Dynamics Unit, Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Matthijs C Dorst
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Yoland Smith
- Yerkes National Primate Research Center, Department of Neurology, Emory University, Atlanta, United States.,Udall Center of Excellence for Parkinson's Disease Research, Emory University, Atlanta, United States
| | - Peter J Magill
- Medical Research Council Brain Network Dynamics Unit, Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Andrew Sharott
- Medical Research Council Brain Network Dynamics Unit, Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
28
|
Patton MH, Roberts BM, Lovinger DM, Mathur BN. Ethanol Disinhibits Dorsolateral Striatal Medium Spiny Neurons Through Activation of A Presynaptic Delta Opioid Receptor. Neuropsychopharmacology 2016; 41:1831-40. [PMID: 26758662 PMCID: PMC4869052 DOI: 10.1038/npp.2015.353] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 11/14/2015] [Accepted: 12/01/2015] [Indexed: 11/09/2022]
Abstract
The dorsolateral striatum mediates habit formation, which is expedited by exposure to alcohol. Across species, alcohol exposure disinhibits the DLS by dampening GABAergic transmission onto this structure's principal medium spiny projection neurons (MSNs), providing a potential mechanistic basis for habitual alcohol drinking. However, the molecular and circuit components underlying this disinhibition remain unknown. To examine this, we used a combination of whole-cell patch-clamp recordings and optogenetics to demonstrate that ethanol potently depresses both MSN- and fast-spiking interneuron (FSI)-MSN GABAergic synaptic transmission in the DLS. Concentrating on the powerfully inhibitory FSI-MSN synapse, we further show that acute exposure of ethanol (50 mM) to striatal slices activates delta opioid receptors that reside on FSI axon terminals and negatively couple to adenylyl cyclase to induce a long-term depression of GABA release onto both direct and indirect pathway MSNs. These findings elucidate a mechanism through which ethanol may globally disinhibit the DLS.
Collapse
Affiliation(s)
- Mary H Patton
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Bradley M Roberts
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - David M Lovinger
- Section on Synaptic Pharmacology, Laboratory for Integrative Neuroscience, Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, US National Institutes of Health, Bethesda, MD, USA
| | - Brian N Mathur
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA,Department of Pharmacology, University of Maryland School of Medicine, BRB RM 4011, 655 West Baltimore Street, Baltimore, MD 21201, USA, Tel: +410 706 8239, Fax: +410 706 8341, E-mail:
| |
Collapse
|
29
|
|
30
|
Mintzopoulos D, Gillis TE, Robertson HR, Dalia T, Feng G, Rauch SL, Kaufman MJ. Striatal magnetic resonance spectroscopy abnormalities in young adult SAPAP3 knockout mice. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2016; 1:39-48. [PMID: 26858992 PMCID: PMC4742338 DOI: 10.1016/j.bpsc.2015.10.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Obsessive compulsive disorder (OCD) is a debilitating condition with lifetime prevalence of 1-3%. OCD typically arises in youth but delays in diagnosis impede optimal treatment and developmental studies of the disorder. Research using genetically modified rodents may provide models of etiology that enable earlier detection and intervention. The SAPAP3 knockout (KO) transgenic mouse was developed as an animal model of OCD and related disorders (OCRD). KO mice exhibit compulsive self-grooming behavior analogous to behaviors found in people with OCRD. Striatal hyperactivity has been reported in these mice and in humans with OCD. METHODS Striatal and medial frontal cortex 9.4 Tesla proton spectra were acquired from young adult SAPAP3 KO and wild-type control mice to determine whether KO mice have metabolic and neurochemical abnormalities. RESULTS Young adult KO mice had lower striatal lactate (P=0.006) and glutathione (P=0.039) levels. Among all mice, striatal lactate and glutathione levels were associated (R=0.73, P=0.007). We found no group differences in medial frontal cortex metabolites. At the age range studied, only 1 of 8 KO mice had skin lesions indicative of severe compulsive grooming. CONCLUSION Young adult SAPAP3 KO mice have striatal but not medial frontal cortex MRS abnormalities that may reflect striatal hypermetabolism accompanied by oxidative stress. These abnormalities typically preceded the onset of severe compulsive grooming. Our findings are consistent with striatal hypermetabolism in OCD. Together, these results suggest that striatal MRS measures of lactate or glutathione might be useful biomarkers for early detection of risk for developing compulsive behavior disorders.
Collapse
|
31
|
Nagypál T, Gombkötő P, Barkóczi B, Benedek G, Nagy A. Activity of Caudate Nucleus Neurons in a Visual Fixation Paradigm in Behaving Cats. PLoS One 2015; 10:e0142526. [PMID: 26544604 PMCID: PMC4636356 DOI: 10.1371/journal.pone.0142526] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 10/22/2015] [Indexed: 11/25/2022] Open
Abstract
Beside its motor functions, the caudate nucleus (CN), the main input structure of the basal ganglia, is also sensitive to various sensory modalities. The goal of the present study was to investigate the effects of visual stimulation on the CN by using a behaving, head-restrained, eye movement-controlled feline model developed recently for this purpose. Extracellular multielectrode recordings were made from the CN of two cats in a visual fixation paradigm applying static and dynamic stimuli. The recorded neurons were classified in three groups according to their electrophysiological properties: phasically active (PAN), tonically active (TAN) and high-firing (HFN) neurons. The response characteristics were investigated according to this classification. The PAN and TAN neurons were sensitive primarily to static stimuli, while the HFN neurons responded primarily to changes in the visual environment i.e. to optic flow and the offset of the stimuli. The HFNs were the most sensitive to visual stimulation; their responses were stronger than those of the PANs and TANs. The majority of the recorded units were insensitive to the direction of the optic flow, regardless of group, but a small number of direction-sensitive neurons were also found. Our results demonstrate that both the static and the dynamic components of the visual information are represented in the CN. Furthermore, these results provide the first piece of evidence on optic flow processing in the CN, which, in more general terms, indicates the possible role of this structure in dynamic visual information processing.
Collapse
Affiliation(s)
- Tamás Nagypál
- Department of Physiology, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Péter Gombkötő
- Center for Molecular and Behavioral Neuroscience Rutgers University, Newark, New Jersey, United States of America
| | - Balázs Barkóczi
- Department of Physiology, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - György Benedek
- Department of Physiology, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Attila Nagy
- Department of Physiology, Faculty of Medicine, University of Szeged, Szeged, Hungary
- * E-mail:
| |
Collapse
|
32
|
Dopaminergic Regulation of Striatal Interneurons in Reward and Addiction: Focus on Alcohol. Neural Plast 2015; 2015:814567. [PMID: 26246915 PMCID: PMC4515529 DOI: 10.1155/2015/814567] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 06/09/2015] [Indexed: 12/13/2022] Open
Abstract
Corticobasal ganglia networks coursing through the striatum are key structures for reward-guided behaviors. The ventral striatum (nucleus accumbens (nAc)) and its reciprocal connection with the ventral tegmental area (VTA) represent a primary component of the reward system, but reward-guided learning also involves the dorsal striatum and dopaminergic inputs from the substantia nigra. The majority of neurons in the striatum (>90%) are GABAergic medium spiny neurons (MSNs), but both the input to and the output from these neurons are dynamically controlled by striatal interneurons. Dopamine is a key neurotransmitter in reward and reward-guided learning, and the physiological activity of GABAergic and cholinergic interneurons is regulated by dopaminergic transmission in a complex manner. Here we review the role of striatal interneurons in modulating striatal output during drug reward, with special emphasis on alcohol.
Collapse
|
33
|
Perk CG, Wickens JR, Hyland BI. Differing properties of putative fast-spiking interneurons in the striatum of two rat strains. Neuroscience 2015; 294:215-26. [PMID: 25758937 DOI: 10.1016/j.neuroscience.2015.02.051] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Revised: 02/05/2015] [Accepted: 02/27/2015] [Indexed: 11/25/2022]
Abstract
Local circuits within the striatum of the basal ganglia include a small number of γ-aminobutyric acid (GABA)-ergic fast-spiking interneurons (FSI). The number of these cells is reduced in disorders of behavioral control, but it is unknown whether this is accompanied by altered electrophysiological properties. The genetically hypertensive (GH) rat strain exhibits impulsiveness and hyperactivity. We investigated if resting-state FSI activity is affected in this strain using extracellular recordings. We also examined the effect of systemic amphetamine (AMPH), a stimulant drug used in the treatment of these particular behavioral deficits. Putative FSI (pFSI) were encountered less often in GH rats compared to the Wistar control strain. pFSI in GH rats also exhibited a higher mean firing rate, higher intraburst firing rate, lower interburst interval, and shorter bursts compared to controls. AMPH increased the mean overall firing rate of Wistar rat pFSI but did not significantly alter the firing properties of this subtype in GH rats. These differences in the resting-state electrophysiological activity of pFSI in GH rats point to them as a cell type of particular interest in understanding striatal functioning across different strains.
Collapse
Affiliation(s)
- C G Perk
- Department of Physiology, Otago School of Medical Sciences, Brain Health Research Centre, and Brain Research New Zealand, University of Otago, Dunedin 9054, New Zealand; Department of Anatomy, Otago School of Medical Sciences, University of Otago, Dunedin 9054, New Zealand.
| | - J R Wickens
- Department of Anatomy, Otago School of Medical Sciences, University of Otago, Dunedin 9054, New Zealand; Neurobiology Research Unit, Okinawa Institute of Science and Technology, 1919-1, Tancha, Onna-Son, Kunigami, Okinawa 904-0412, Japan
| | - B I Hyland
- Department of Physiology, Otago School of Medical Sciences, Brain Health Research Centre, and Brain Research New Zealand, University of Otago, Dunedin 9054, New Zealand
| |
Collapse
|
34
|
Moyer JT, Halterman BL, Finkel LH, Wolf JA. Lateral and feedforward inhibition suppress asynchronous activity in a large, biophysically-detailed computational model of the striatal network. Front Comput Neurosci 2014; 8:152. [PMID: 25505406 PMCID: PMC4243567 DOI: 10.3389/fncom.2014.00152] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 11/04/2014] [Indexed: 12/03/2022] Open
Abstract
Striatal medium spiny neurons (MSNs) receive lateral inhibitory projections from other MSNs and feedforward inhibitory projections from fast-spiking, parvalbumin-containing striatal interneurons (FSIs). The functional roles of these connections are unknown, and difficult to study in an experimental preparation. We therefore investigated the functionality of both lateral (MSN-MSN) and feedforward (FSI-MSN) inhibition using a large-scale computational model of the striatal network. The model consists of 2744 MSNs comprised of 189 compartments each and 121 FSIs comprised of 148 compartments each, with dendrites explicitly represented and almost all known ionic currents included and strictly constrained by biological data as appropriate. Our analysis of the model indicates that both lateral inhibition and feedforward inhibition function at the population level to limit non-ensemble MSN spiking while preserving ensemble MSN spiking. Specifically, lateral inhibition enables large ensembles of MSNs firing synchronously to strongly suppress non-ensemble MSNs over a short time-scale (10–30 ms). Feedforward inhibition enables FSIs to strongly inhibit weakly activated, non-ensemble MSNs while moderately inhibiting activated ensemble MSNs. Importantly, FSIs appear to more effectively inhibit MSNs when FSIs fire asynchronously. Both types of inhibition would increase the signal-to-noise ratio of responding MSN ensembles and contribute to the formation and dissolution of MSN ensembles in the striatal network.
Collapse
Affiliation(s)
- Jason T Moyer
- Department of Bioengineering, University of Pennsylvania Philadelphia, PA, USA
| | | | - Leif H Finkel
- Department of Bioengineering, University of Pennsylvania Philadelphia, PA, USA
| | - John A Wolf
- Department of Neurosurgery, University of Pennsylvania Philadelphia, PA, USA
| |
Collapse
|
35
|
Garcia-Munoz M, Lopez-Huerta VG, Carrillo-Reid L, Arbuthnott GW. Extrasynaptic glutamate NMDA receptors: key players in striatal function. Neuropharmacology 2014; 89:54-63. [PMID: 25239809 DOI: 10.1016/j.neuropharm.2014.09.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 08/26/2014] [Accepted: 09/06/2014] [Indexed: 10/24/2022]
Abstract
N-methyl-D-aspartate receptors (NMDAR) are crucial for the function of excitatory neurotransmission and are present at the synapse and on the extrasynaptic membrane. The major nucleus of the basal ganglia, striatum, receives a large glutamatergic excitatory input carrying information about movements and associated sensory stimulation for its proper function. Such bombardment of glutamate synaptic release results in a large extracellular concentration of glutamate that can overcome the neuronal and glial uptake homeostatic systems therefore allowing the stimulation of extrasynaptic glutamate receptors. Here we have studied the participation of their extrasynaptic type in cortically evoked responses or in the presence of NMDARs stimulation. We report that extrasynaptic NMDAR blocker memantine, reduced in a dose-dependent manner cortically induced NMDA excitatory currents in striatal neurons (recorded in zero-Mg(++) plus DNQX 10 μM). Moreover, memantine (2-4 μM) significantly reduced the NMDAR-dependent membrane potential oscillations called up and down states. Recordings of neuronal striatal networks with a fluorescent calcium indicator or with multielectrode arrays (MEA) also showed that memantine reduced in a dose-dependent manner, NMDA-induced excitatory currents and network behavior. We used multielectrode arrays (MEA) to grow segregated cortical and striatal neurons. Once synaptic contacts were developed (>21DIV) recordings of extracellular activity confirmed the cortical drive of spontaneous synchronous discharges in both compartments. After severing connections between compartments, active striatal neurons in the presence of memantine (1 μM) and CNQX (10 μM) were predominantly fast spiking interneurons (FSI). The significance of extrasynaptic receptors in the regulation of striatal function and neuronal network activity is evident.
Collapse
Affiliation(s)
- Marianela Garcia-Munoz
- Brain Mechanisms for Behaviour Unit, Okinawa Institute of Science and Technology Graduate University, Japan.
| | - Violeta G Lopez-Huerta
- Brain Mechanisms for Behaviour Unit, Okinawa Institute of Science and Technology Graduate University, Japan.
| | - Luis Carrillo-Reid
- Brain Mechanisms for Behaviour Unit, Okinawa Institute of Science and Technology Graduate University, Japan; Department of Biological Sciences, Columbia University, NY, USA.
| | - Gordon W Arbuthnott
- Brain Mechanisms for Behaviour Unit, Okinawa Institute of Science and Technology Graduate University, Japan.
| |
Collapse
|
36
|
Kim N, Barter JW, Sukharnikova T, Yin HH. Striatal firing rate reflects head movement velocity. Eur J Neurosci 2014; 40:3481-90. [PMID: 25209171 DOI: 10.1111/ejn.12722] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 08/09/2014] [Accepted: 08/12/2014] [Indexed: 01/25/2023]
Abstract
Although the basal ganglia have long been implicated in the initiation of actions, their contribution to movement remains a matter of dispute. Using wireless multi-electrode recording and motion tracking, we examined the relationship between single-unit activity in the sensorimotor striatum and movement kinematics. We recorded single-unit activity from medium spiny projection neurons and fast-spiking interneurons while monitoring the movements of mice using motion tracking. In Experiment 1, we trained mice to generate movements reliably by water-depriving them and giving them periodic cued sucrose rewards. We found high correlations between single-unit activity and movement velocity in particular directions. This correlation was found in both putative medium spiny projection neurons and fast-spiking interneurons. In Experiment 2, to rule out the possibility that the observed correlations were due to reward expectancy, we repeated the same procedure but added trials in which sucrose delivery was replaced by an aversive air puff stimulus. The air puff generated avoidance movements that were clearly different from movements on rewarded trials, but the same neurons that showed velocity correlation on reward trials exhibited a similar correlation on air puff trials. These experiments show for the first time that the firing rate of striatal neurons reflects movement velocity for different types of movements, whether to seek rewards or to avoid harm.
Collapse
Affiliation(s)
- Namsoo Kim
- Department of Psychology and Neuroscience, Duke University, Box 91050, Durham, NC, 27708, USA
| | | | | | | |
Collapse
|
37
|
Muñoz-Manchado AB, Foldi C, Szydlowski S, Sjulson L, Farries M, Wilson C, Silberberg G, Hjerling-Leffler J. Novel Striatal GABAergic Interneuron Populations Labeled in the 5HT3a(EGFP) Mouse. Cereb Cortex 2014; 26:96-105. [PMID: 25146369 DOI: 10.1093/cercor/bhu179] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Histological and morphological studies indicate that approximately 5% of striatal neurons are cholinergic or γ-aminobutyric acidergic (GABAergic) interneurons (gINs). However, the number of striatal neurons expressing known interneuron markers is too small to account for the entire interneuron population. We therefore studied the serotonin (5HT) receptor 3a-enhanced green fluorescent protein (5HT3a(EGFP)) mouse, in which we found that a large number of striatal gINs are labeled. Roughly 20% of 5HT3a(EGFP)-positive cells co-express parvalbumin and exhibit fast-spiking (FS) electrophysiological properties. However, the majority of labeled neurons do not overlap with known molecular interneuron markers. Intrinsic electrical properties reveal at least 2 distinct novel subtypes: a late-spiking (LS) neuropeptide-Y (NPY)-negative neurogliaform (NGF) interneuron, and a large heterogeneous population with several features resembling low-threshold-spiking (LTS) interneurons that do not express somatostatin, NPY, or neuronal nitric oxide synthase. Although the 5HT3a(EGFP) NGF and LTS-like interneurons have electrophysiological properties similar to previously described populations, they are pharmacologically distinct. In direct contrast to previously described NPY(+) LTS and NGF cells, LTS-like 5HT3a(EGFP) cells show robust responses to nicotine administration, while the 5HT3a(EGFP) NGF cell type shows little or no response. By constructing a molecular map of the overlap between these novel populations and existing interneuron populations, we are able to reconcile the morphological and molecular estimates of striatal interneuron numbers.
Collapse
Affiliation(s)
| | - C Foldi
- Department of Medical Biochemistry and Biophysics
| | - S Szydlowski
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - L Sjulson
- Department of Psychiatry.,Department of Neuroscience and Physiology, NYU Neuroscience Institute, NYU Langone Medical Center, New York, NY, USA
| | - M Farries
- Department of Biology, University of Texas at San Antonio, San Antonio, TX, USA
| | - C Wilson
- Department of Biology, University of Texas at San Antonio, San Antonio, TX, USA
| | - G Silberberg
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
38
|
Ma Y, Feng Q, Ouyang L, Mu S, Liu B, Li Y, Chen S, Lei W. Morphological diversity of GABAergic and cholinergic interneurons in the striatal dorsolateral and ventromedial regions of rats. Cell Mol Neurobiol 2014; 34:351-9. [PMID: 24343377 DOI: 10.1007/s10571-013-0019-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Accepted: 12/09/2013] [Indexed: 10/25/2022]
Abstract
The striatum plays a fundamental role in sensorimotor and cognitive functions of the body, and different sub-regions control different physiological functions. The striatal interneurons play important roles in the striatal function, yet their specific functions are not clearly elucidated so far. The present study aimed to investigate the morphological properties of the GABAergic interneurons expressing neuropeptide Y (NPY), calretinin (Cr), and parvalbumin (Parv) as well as the cholinergic interneurons expressing choline acetyltransferase (ChAT) in the striatal dorsolateral (DL) and ventromedial (VM) regions of rats using immunohistochemistry and Western blot. The present results showed that the somatic size of Cr+ was the smallest, while ChAT+ was the largest among the four types of interneurons. There was no regional difference in neuronal somatic size of all types of interneurons. Cr+ and Parv+ neurons were differentially distributed in the striatum. Moreover, Parv+ had the longest primary dendrites in the DL region, while NPY+ had the longest ones in the VM region of striatum. But there was regional difference in the length of primary dendrites of Parv. The numbers of primary dendrites of Parv+ were the largest in both DL and VM regions of striatum. Both Cr+ and Parv+ primary dendrites displayed regional difference in the striatum. Western blot further confirmed the regional differences in the protein expression level of Cr and Parv. Hence, the present study indicates that GABAergic and cholinergic interneurons might be involved in different physiological functions based on their morphological and distributional diversity in different regions of the rat striatum.
Collapse
Affiliation(s)
- Yuxin Ma
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan Rd 2, Guangzhou, 510080, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
This chapter focuses on neurodevelopmental diseases that are tightly linked to abnormal function of the striatum and connected structures. We begin with an overview of three representative diseases in which striatal dysfunction plays a key role--Tourette syndrome and obsessive-compulsive disorder, Rett's syndrome, and primary dystonia. These diseases highlight distinct etiologies that disrupt striatal integrity and function during development, and showcase the varied clinical manifestations of striatal dysfunction. We then review striatal organization and function, including evidence for striatal roles in online motor control/action selection, reinforcement learning, habit formation, and action sequencing. A key barrier to progress has been the relative lack of animal models of these diseases, though recently there has been considerable progress. We review these efforts, including their relative merits providing insight into disease pathogenesis, disease symptomatology, and basal ganglia function.
Collapse
|
40
|
Russo G, Nieus TR, Maggi S, Taverna S. Dynamics of action potential firing in electrically connected striatal fast-spiking interneurons. Front Cell Neurosci 2013; 7:209. [PMID: 24294191 PMCID: PMC3827583 DOI: 10.3389/fncel.2013.00209] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 10/21/2013] [Indexed: 12/31/2022] Open
Abstract
Fast-spiking interneurons (FSIs) play a central role in organizing the output of striatal neural circuits, yet functional interactions between these cells are still largely unknown. Here we investigated the interplay of action potential (AP) firing between electrically connected pairs of identified FSIs in mouse striatal slices. In addition to a loose coordination of firing activity mediated by membrane potential coupling, gap junctions (GJ) induced a frequency-dependent inhibition of spike discharge in coupled cells. At relatively low firing rates (2–20 Hz), some APs were tightly synchronized whereas others were inhibited. However, burst firing at intermediate frequencies (25–60 Hz) mostly induced spike inhibition, while at frequencies >50–60 Hz FSI pairs tended to synchronize. Spike silencing occurred even in the absence of GABAergic synapses or persisted after a complete block of GABAA receptors. Pharmacological suppression of presynaptic spike afterhyperpolarization (AHP) caused postsynaptic spikelets to become more prone to trigger spikes at near-threshold potentials, leading to a mostly synchronous firing activity. The complex pattern of functional coordination mediated by GJ endows FSIs with peculiar dynamic properties that may be critical in controlling striatal-dependent behavior.
Collapse
Affiliation(s)
- Giovanni Russo
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia Genoa, Italy
| | | | | | | |
Collapse
|
41
|
Barbaresi P, Mensà E, Lariccia V, Pugnaloni A, Amoroso S, Fabri M. Differential distribution of parvalbumin- and calbindin-D28K-immunoreactive neurons in the rat periaqueductal gray matter and their colocalization with enzymes producing nitric oxide. Brain Res Bull 2013; 99:48-62. [PMID: 24107244 DOI: 10.1016/j.brainresbull.2013.09.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 09/09/2013] [Accepted: 09/17/2013] [Indexed: 10/26/2022]
Abstract
The distribution, colocalization with enzymes producing nitric oxide (NO), and the synaptic organization of neurons containing two calcium-binding proteins (CaBPs) - parvalbumin (Parv) and calbindin-D28K (Calb) - were investigated in the rat periaqueductal gray matter (PAG). Parv-immunopositive (ParvIP) neurons were detected in the mesencephalic nucleus and rarely in the PAG. CalbIP neurons were found both in the dorsolateral (PAG-dl) and ventrolateral PAG (PAG-vl); their size ranged from 112.96 μm(2) (PAG-dl) to 125.13 μm(2) (PAG-vl). Ultrastructurally Parv and Calb immunoreactivity was mostly found in dendritic profiles. Axon terminals containing each of the two CaBPs formed symmetric synapses. Moreover both Parv and Calb were used to label a subpopulation of NO-producing neurons. Colocalization was investigated using two protocols: (i) a combination of Calb and Parv immunocytochemistry (Icc) with nicotinamide adenine dinucleotide phosphate diaphorase (NADPH-d) histochemistry (Hi) and (ii) neuronal NO synthase-Icc (nNOS) (immunofluorescence). Both techniques demonstrated a complete lack of colocalization of Parv and NADPH-d/nNOS in PAG neurons. Double-labeled (DL) neurons (Calb-NADPH-d; Calb-nNOS) were detected in PAG-dl. NADPH-d-Hi/Calb-Icc indicated that 41-47% of NADPH-d-positive neurons contained Calb, whereas 17-23% of CalbIP cells contained NADPH-d. Two-color immunofluorescence revealed that 53-66% of nNOSIP cells colocalized with Calb and 24-34% of CalbIP neurons contained nNOS. DL neuron size was 104.44 μm(2); neurons labeled only with NADPH-d or Calb measured 89.793 μm(2) and 113.48 μm(2), respectively. Together with previous findings (Barbaresi et al. [2012]) these data suggest that: Therefore the important aspect of the PAG intrinsic organization emerging from this and previous double-labeling studies is the chemical diversity of NO-synthesizing neurons, which is likely related to the different functions in which these neurons are involved.
Collapse
Affiliation(s)
- Paolo Barbaresi
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Marche Polytechnic University, I-60020 Ancona, Italy.
| | | | | | | | | | | |
Collapse
|
42
|
Abstract
How does the ventral striatum (VS) prioritize and process afferent input? In this issue, Calhoon and O'Donnell (2013) demonstrate that cortical projections to the VS can attenuate hippocampal and thalamic VS input, suggesting that the cortex can uniquely control VS circuit dynamics.
Collapse
|
43
|
Song CH, Bernhard D, Bolarinwa C, Hess EJ, Smith Y, Jinnah HA. Subtle microstructural changes of the striatum in a DYT1 knock-in mouse model of dystonia. Neurobiol Dis 2013; 54:362-71. [PMID: 23336980 PMCID: PMC3628999 DOI: 10.1016/j.nbd.2013.01.008] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 01/04/2013] [Accepted: 01/10/2013] [Indexed: 11/21/2022] Open
Abstract
The dystonias are comprised of a group of disorders that share common neurological abnormalities of involuntary twisting or repetitive movements and postures. The most common inherited primary dystonia is DYT1 dystonia, which is due to loss of a GAG codon in the TOR1A gene that encodes torsinA. Autopsy studies of brains from patients with DYT1 dystonia have revealed few abnormalities, although recent neuroimaging studies have implied the existence of microstructural defects that might not be detectable with traditional histopathological methods. The current studies took advantage of a knock-in mouse model for DYT1 dystonia to search for subtle anatomical abnormalities in the striatum, a region often implicated in studies of dystonia. Multiple abnormalities were identified using a combination of quantitative stereological measures of immunohistochemical stains for specific neuronal populations, morphometric studies of Golgi-stained neurons, and immuno-electron microscopy of synaptic connectivity. In keeping with other studies, there was no obvious loss of striatal neurons in the DYT1 mutant mice. However, interneurons immunoreactive for choline acetyltransferase or parvalbumin were larger in the mutants than in control mice. In contrast, interneurons immunoreactive for neuronal nitric oxide synthase were smaller in the mutants than in controls. Golgi histochemical studies of medium spiny projection neurons in the mutant mice revealed slightly fewer and thinner dendrites, and a corresponding loss of dendritic spines. Electron microscopic studies showed a reduction in the ratio of axo-spinous to axo-dendritic synaptic inputs from glutamatergic and dopaminergic sources in mutant mice compared with controls. These results suggest specific anatomical substrates for altered signaling in the striatum and potential correlates of the abnormalities implied by human imaging studies of DYT1 dystonia.
Collapse
Affiliation(s)
- Chang-Hyun Song
- Department of Neurology, Emory University, Atlanta GA, 30322
| | | | - Caroline Bolarinwa
- Yerkes National Primate Research Center and Department of Neurology, Emory University, Atlanta GA, 30329
| | - Ellen J. Hess
- Department of Neurology, Emory University, Atlanta GA, 30322
- Department of Pharmacology, Emory University, Atlanta GA, 30322
| | - Yoland Smith
- Yerkes National Primate Research Center and Department of Neurology, Emory University, Atlanta GA, 30329
| | - H. A. Jinnah
- Departments of Neurology, Human Genetics and Pediatrics, Emory University, Atlanta GA 30322
| |
Collapse
|
44
|
Dong C, Anand KJS. Developmental neurotoxicity of ketamine in pediatric clinical use. Toxicol Lett 2013; 220:53-60. [PMID: 23566897 DOI: 10.1016/j.toxlet.2013.03.030] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 03/21/2013] [Accepted: 03/22/2013] [Indexed: 12/27/2022]
Abstract
Ketamine is widely used as an anesthetic, analgesic, and sedative in pediatric clinical practice and it is also listed as an illicit drug by most countries. Recent in vivo and in vitro animal studies have confirmed that ketamine can induce neuronal cell death in the immature brain, resulting from widespread neuronal apoptosis. These effects can disturb normal development further altering the structure and functions of the brain. Our recent studies further indicate that ketamine can alter neurogenesis from neural stem progenitor cells in the developing brain. Taken together, these findings identify a novel complication associated with ketamine use in premature infants, term newborns, and pregnant women. Recent data on the developmental neurotoxicity of ketamine are reviewed with proposed future directions for evaluating the safety of ketamine in these patient populations.
Collapse
Affiliation(s)
- Chaoxuan Dong
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, United States.
| | | |
Collapse
|
45
|
Orduz D, Bischop DP, Schwaller B, Schiffmann SN, Gall D. Parvalbumin tunes spike-timing and efferent short-term plasticity in striatal fast spiking interneurons. J Physiol 2013; 591:3215-32. [PMID: 23551945 DOI: 10.1113/jphysiol.2012.250795] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Striatal fast spiking interneurons (FSIs) modulate output of the striatum by synchronizing medium-sized spiny neurons (MSNs). Recent studies have broadened our understanding of FSIs, showing that they are implicated in severe motor disorders such as parkinsonism, dystonia and Tourette syndrome. FSIs are the only striatal neurons to express the calcium-binding protein parvalbumin (PV). This selective expression of PV raises questions about the functional role of this Ca(2+) buffer in controlling FSI Ca(2+) dynamics and, consequently, FSI spiking mode and neurotransmission. To study the functional involvement of FSIs in striatal microcircuit activity and the role of PV in FSI function, we performed perforated patch recordings on enhanced green fluorescent protein-expressing FSIs in brain slices from control and PV-/- mice. Our results revealed that PV-/- FSIs fired more regularly and were more excitable than control FSIs by a mechanism in which Ca(2+) buffering is linked to spiking activity as a result of the activation of small conductance Ca(2+)-dependent K(+) channels. A modelling approach of striatal FSIs supports our experimental results. Furthermore, PV deletion modified frequency-specific short-term plasticity at inhibitory FSI to MSN synapses. Our results therefore reinforce the hypothesis that in FSIs, PV is crucial for fine-tuning of the temporal responses of the FSI network and for the orchestration of MSN populations. This, in turn, may play a direct role in the generation and pathology-related worsening of motor rhythms.
Collapse
Affiliation(s)
- David Orduz
- Laboratory of Neurophysiology, Université Libre de Bruxelles, ULB Neuroscience Institute, Université Libre de Bruxelles, 808 route de Lennik, CP601, 1070 Brussels, Belgium.
| | | | | | | | | |
Collapse
|
46
|
The percentage of interneurons in the dorsal striatum of the rat, cat, monkey and human: A critique of the evidence. ACTA ACUST UNITED AC 2013. [DOI: 10.1016/j.baga.2012.11.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
47
|
GABAergic neuron specification in the spinal cord, the cerebellum, and the cochlear nucleus. Neural Plast 2012; 2012:921732. [PMID: 22830054 PMCID: PMC3395262 DOI: 10.1155/2012/921732] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Revised: 05/17/2012] [Accepted: 05/17/2012] [Indexed: 12/01/2022] Open
Abstract
In the nervous system, there are a wide variety of neuronal cell types that have morphologically, physiologically, and histochemically different characteristics. These various types of neurons can be classified into two groups: excitatory and inhibitory neurons. The elaborate balance of the activities of the two types is very important to elicit higher brain function, because its imbalance may cause neurological disorders, such as epilepsy and hyperalgesia. In the central nervous system, inhibitory neurons are mainly represented by GABAergic ones with some exceptions such as glycinergic. Although the machinery to specify GABAergic neurons was first studied in the telencephalon, identification of key molecules, such as pancreatic transcription factor 1a (Ptf1a), as well as recently developed genetic lineage-tracing methods led to the better understanding of GABAergic specification in other brain regions, such as the spinal cord, the cerebellum, and the cochlear nucleus.
Collapse
|
48
|
Lee H, Leamey CA, Sawatari A. Perineuronal nets play a role in regulating striatal function in the mouse. PLoS One 2012; 7:e32747. [PMID: 22427872 PMCID: PMC3299692 DOI: 10.1371/journal.pone.0032747] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Accepted: 02/01/2012] [Indexed: 11/19/2022] Open
Abstract
The striatum is the primary input nucleus of the basal ganglia, a collection of nuclei that play important roles in motor control and associative learning. We have previously reported that perineuronal nets (PNNs), aggregations of chondroitin-sulfate proteoglycans (CSPGs), form in the matrix compartment of the mouse striatum during the second postnatal week. This period overlaps with important developmental changes, including the attainment of an adult-like gait. Here, we investigate the identity of the cells encapsulated by PNNs, characterize their topographical distribution and determine their function by assessing the impact of enzymatic digestion of PNNs on two striatum-dependent behaviors: ambulation and goal-directed spatial learning. We show PNNs are more numerous caudally, and that a substantial fraction (41%) of these structures surrounds parvalbumin positive (PV+) interneurons, while approximately 51% of PV+ cells are ensheathed by PNNs. The colocalization of these structures is greatest in dorsal, lateral and caudal regions of the striatum. Bilateral digestion of striatal PNNs led to an increase in both the width and variability of hind limb gait. Intriguingly, this also resulted in an improvement in the acquisition rate of the Morris water maze. Together, these data show that PNNs are associated with specific elements of striatal circuits and play a key role in regulating the function of this important structure in the mouse.
Collapse
Affiliation(s)
| | | | - Atomu Sawatari
- Discipline of Physiology, School of Medical Sciences and the Bosch Institute, University of Sydney, Sydney, Australia
- * E-mail:
| |
Collapse
|
49
|
Kinases SPAK and OSR1 are upregulated by estradiol and activate NKCC1 in the developing hypothalamus. J Neurosci 2012; 32:593-8. [PMID: 22238094 DOI: 10.1523/jneurosci.5415-11.2012] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
In immature neurons the amino acid neurotransmitter, GABA provides the dominant mode for neuronal excitation by inducing membrane depolarization due to Cl(-) efflux through GABA(A) receptors (GABA(A)Rs). The driving force for Cl(-) is outward because the Na(+)-K(+)-2Cl(-) cotransporter (NKCC1) elevates the Cl(-) concentration in these cells. GABA-induced membrane depolarization and the resulting activation of voltage-gated Ca(2+) channels is fundamental to normal brain development, yet the mechanisms that regulate depolarizing GABA are not well understood. The neurosteroid estradiol potently augments depolarizing GABA action in the immature hypothalamus by enhancing the activity of the NKCC1 cotransporter. Understanding how estradiol controls NKCC1 activity will be essential for a complete understanding of brain development. We now report that estradiol treatment of newborn rat pups significantly increases protein levels of two kinases upstream of the NKCC1 cotransporter, SPAK (STE20/SPS1-related proline alanine rich kinase) and OSR1 (oxidative stress response kinase). The estradiol-induced increase is transcription dependent, and its time course parallels that of estradiol-enhanced phosphorylation of NKCC1. Antisense oligonucleotide-mediated knockdown of SPAK, and to a lesser degree of OSR1, precludes estradiol-mediated enhancement of NKCC1 phosphorylation. Functionally, knockdown of SPAK or OSR1 in embryonic hypothalamic cultures diminishes estradiol-enhanced Ca(2+) influx induced by GABA(A)R activation. Our data suggest that SPAK and OSR1 may be critical factors in the regulation of depolarizing GABA-mediated processes in the developing brain. It will be important to examine these kinases with respect to sex differences and developmental brain anomalies in future studies.
Collapse
|
50
|
Kreitzer AC, Berke JD. Investigating striatal function through cell-type-specific manipulations. Neuroscience 2011; 198:19-26. [PMID: 21867745 DOI: 10.1016/j.neuroscience.2011.08.018] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Revised: 08/03/2011] [Accepted: 08/06/2011] [Indexed: 12/17/2022]
Abstract
The striatum integrates convergent input from the cortex, thalamus, and midbrain, and has a powerful influence over motivated behavior via outputs to downstream basal ganglia nuclei. Although the anatomy and physiology of distinct classes of striatal neurons have been intensively studied, the specific functions of these cell subpopulations have been more difficult to address. Recently, application of new methodologies for perturbing activity and signaling in different cell types in vivo has begun to allow direct tests of the causal roles of striatal neurons in behavior.
Collapse
Affiliation(s)
- A C Kreitzer
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA.
| | | |
Collapse
|