Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.026
Filter
1.
Nature ; 621(7979): 577-585, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37557915

ABSTRACT

Striatal dopamine and acetylcholine are essential for the selection and reinforcement of motor actions and decision-making1. In vitro studies have revealed an intrastriatal circuit in which acetylcholine, released by cholinergic interneurons (CINs), drives the release of dopamine, and dopamine, in turn, inhibits the activity of CINs through dopamine D2 receptors (D2Rs). Whether and how this circuit contributes to striatal function in vivo is largely unknown. Here, to define the role of this circuit in a living system, we monitored acetylcholine and dopamine signals in the ventrolateral striatum of mice performing a reward-based decision-making task. We establish that dopamine and acetylcholine exhibit multiphasic and anticorrelated transients that are modulated by decision history and reward outcome. Dopamine dynamics and reward encoding do not require the release of acetylcholine by CINs. However, dopamine inhibits acetylcholine transients in a D2R-dependent manner, and loss of this regulation impairs decision-making. To determine how other striatal inputs shape acetylcholine signals, we assessed the contribution of cortical and thalamic projections, and found that glutamate release from both sources is required for acetylcholine release. Altogether, we uncover a dynamic relationship between dopamine and acetylcholine during decision-making, and reveal multiple modes of CIN regulation. These findings deepen our understanding of the neurochemical basis of decision-making and behaviour.


Subject(s)
Acetylcholine , Corpus Striatum , Decision Making , Dopamine , Glutamic Acid , Animals , Mice , Acetylcholine/metabolism , Corpus Striatum/cytology , Corpus Striatum/metabolism , Dopamine/metabolism , Glutamic Acid/metabolism , Neostriatum/cytology , Neostriatum/metabolism , Decision Making/physiology , Reward , Receptors, Dopamine D2/metabolism , Cholinergic Neurons/metabolism , Neural Pathways
2.
Nature ; 621(7979): 543-549, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37558873

ABSTRACT

External rewards such as food and money are potent modifiers of behaviour1,2. Pioneering studies established that these salient sensory stimuli briefly interrupt the tonic discharge of neurons that produce the neuromodulators dopamine (DA) and acetylcholine (ACh): midbrain DA neurons (DANs) fire a burst of action potentials that broadly elevates DA in the striatum3,4 at the same time that striatal cholinergic interneurons (CINs) produce a characteristic pause in firing5,6. These phasic responses are thought to create unique, temporally limited conditions that motivate action and promote learning7-11. However, the dynamics of DA and ACh outside explicitly rewarded situations remain poorly understood. Here we show that extracellular DA and ACh levels fluctuate spontaneously and periodically at a frequency of approximately 2 Hz in the dorsal striatum of mice and maintain the same temporal relationship relative to one another as that evoked by reward. We show that this neuromodulatory coordination does not arise from direct interactions between DA and ACh within the striatum. Instead, we provide evidence that periodic fluctuations in striatal DA are inherited from midbrain DANs, while striatal ACh transients are driven by glutamatergic inputs, which act to locally synchronize the spiking of CINs. Together, our findings show that striatal neuromodulatory dynamics are autonomously organized by distributed extra-striatal afferents. The dominance of intrinsic rhythms in DA and ACh offers new insights for explaining how reward-associated neural dynamics emerge and how the brain motivates action and promotes learning from within.


Subject(s)
Acetylcholine , Corpus Striatum , Dopamine , Animals , Mice , Acetylcholine/metabolism , Action Potentials , Corpus Striatum/cytology , Corpus Striatum/metabolism , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Glutamine/metabolism , Interneurons/metabolism , Motivation , Neostriatum/cytology , Neostriatum/metabolism , Reward , Afferent Pathways
3.
Nature ; 610(7931): 327-334, 2022 10.
Article in English | MEDLINE | ID: mdl-36171283

ABSTRACT

Recent studies suggested that microglia, the primary brain immune cells, can affect circuit connectivity and neuronal function1,2. Microglia infiltrate the neuroepithelium early in embryonic development and are maintained in the brain throughout adulthood3,4. Several maternal environmental factors-such as an aberrant microbiome, immune activation and poor nutrition-can influence prenatal brain development5,6. Nevertheless, it is unknown how changes in the prenatal environment instruct the developmental trajectory of infiltrating microglia, which in turn affect brain development and function. Here we show that, after maternal immune activation (MIA) in mice, microglia from the offspring have a long-lived decrease in immune reactivity (blunting) across the developmental trajectory. The blunted immune response was accompanied by changes in chromatin accessibility and reduced transcription factor occupancy of the open chromatin. Single-cell RNA-sequencing analysis revealed that MIA does not induce a distinct subpopulation but, rather, decreases the contribution to inflammatory microglia states. Prenatal replacement of microglia from MIA offspring with physiological infiltration of naive microglia ameliorated the immune blunting and restored a decrease in presynaptic vesicle release probability onto dopamine receptor type-two medium spiny neurons, indicating that aberrantly formed microglia due to an adverse prenatal environment affect the long-term microglia reactivity and proper striatal circuit development.


Subject(s)
Inflammation , Microglia , Mothers , Neural Pathways , Prenatal Exposure Delayed Effects , Animals , Chromatin/genetics , Chromatin/metabolism , Female , Inflammation/immunology , Inflammation/pathology , Mice , Microglia/immunology , Microglia/pathology , Neostriatum/cytology , Neural Pathways/pathology , Neurons/pathology , Pregnancy , Prenatal Exposure Delayed Effects/genetics , Prenatal Exposure Delayed Effects/immunology , RNA-Seq , Receptors, Dopamine/metabolism , Single-Cell Analysis , Transcription Factors/metabolism
4.
Nat Neurosci ; 24(10): 1452-1464, 2021 10.
Article in English | MEDLINE | ID: mdl-34413512

ABSTRACT

Cortical circuits process both sensory and motor information in animals performing perceptual tasks. However, it is still unclear how sensory inputs are transformed into motor signals in the cortex to initiate goal-directed actions. In this study, we found that a visual-to-motor inhibitory circuit in the anterior cingulate cortex (ACC) triggers precise action in mice performing visual Go/No-go tasks. Three distinct features of ACC neurons-visual amplitudes of sensory neurons, suppression times of motor neurons and network activity from other neurons-predicted response times of the mice. Moreover, optogenetic activation of visual inputs in the ACC, which drives fast-spiking sensory neurons, prompted task-relevant actions in mice by suppressing ACC motor neurons and disinhibiting downstream striatal neurons. Notably, when mice terminated actions in response to stop signals, both motor neuron and network activity increased. Collectively, our data demonstrate that visual inputs to the frontal cortex trigger gated feedforward inhibition to initiate goal-directed actions.


Subject(s)
Feedback, Psychological , Frontal Lobe/physiology , Goals , Inhibition, Psychological , Animals , Gyrus Cinguli/physiology , Mice , Mice, Inbred C57BL , Motor Neurons/physiology , Neostriatum/cytology , Neostriatum/physiology , Nerve Net/physiology , Optogenetics , Psychomotor Performance/physiology , Reaction Time , Visual Perception/physiology
5.
J Neurosci ; 41(18): 3966-3987, 2021 05 05.
Article in English | MEDLINE | ID: mdl-33731445

ABSTRACT

The classic basal ganglia circuit model asserts a complete segregation of the two striatal output pathways. Empirical data argue that, in addition to indirect-pathway striatal projection neurons (iSPNs), direct-pathway striatal projection neurons (dSPNs) innervate the external globus pallidus (GPe). However, the functions of the latter were not known. In this study, we interrogated the organization principles of striatopallidal projections and their roles in full-body movement in mice (both males and females). In contrast to the canonical motor-promoting response of dSPNs in the dorsomedial striatum (DMSdSPNs), optogenetic stimulation of dSPNs in the dorsolateral striatum (DLSdSPNs) suppressed locomotion. Circuit analyses revealed that dSPNs selectively target Npas1+ neurons in the GPe. In a chronic 6-hydroxydopamine lesion model of Parkinson's disease, the dSPN-Npas1+ projection was dramatically strengthened. As DLSdSPN-Npas1+ projection suppresses movement, the enhancement of this projection represents a circuit mechanism for the hypokinetic symptoms of Parkinson's disease that has not been previously considered. In sum, our results suggest that dSPN input to the GPe is a critical circuit component that is involved in the regulation of movement in both healthy and parkinsonian states.SIGNIFICANCE STATEMENT In the classic basal ganglia model, the striatum is described as a divergent structure: it controls motor and adaptive functions through two segregated, opposing output streams. However, the experimental results that show the projection from direct-pathway neurons to the external pallidum have been largely ignored. Here, we showed that this striatopallidal subpathway targets a select subset of neurons in the external pallidum and is motor-suppressing. We found that this subpathway undergoes changes in a Parkinson's disease model. In particular, our results suggest that the increase in strength of this subpathway contributes to the slowness or reduced movements observed in Parkinson's disease.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Globus Pallidus/physiology , Neostriatum/physiology , Nerve Tissue Proteins/physiology , Neurons/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Female , Globus Pallidus/cytology , Locomotion/physiology , Male , Mice , Mice, Inbred C57BL , Movement/physiology , Neostriatum/cytology , Nerve Tissue Proteins/genetics , Neural Pathways/cytology , Neural Pathways/physiology , Optogenetics , Oxidopamine , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/physiopathology , Rabbits
6.
STAR Protoc ; 2(1): 100230, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33364620

ABSTRACT

We developed an adeno-associated virus (AAV) vector-based technique to label mouse neostriatal neurons comprising direct and indirect pathways with different fluorescent proteins and analyze their axonal projections. The AAV vector expresses GFP or RFP in the presence or absence of Cre recombinase and should be useful for labeling two cell populations exclusively dependent on its expression. Here, we describe the AAV vector design, stereotaxic injection of the AAV vector, and a highly sensitive immunoperoxidase method for axon visualization. For complete details on the use and execution of this protocol, please refer to Okamoto et al. (2020).


Subject(s)
Dependovirus , Genetic Vectors , Neostriatum/metabolism , Neural Pathways/metabolism , Neurons/metabolism , Transduction, Genetic , Animals , Integrases/biosynthesis , Integrases/genetics , Mice , Neostriatum/cytology , Neural Pathways/cytology , Neurons/cytology
7.
J Neurosci ; 41(1): 73-88, 2021 01 06.
Article in English | MEDLINE | ID: mdl-33177068

ABSTRACT

The capacity for sensory systems to encode relevant information that is invariant to many stimulus changes is central to normal, real-world, cognitive function. This invariance is thought to be reflected in the complex spatiotemporal activity patterns of neural populations, but our understanding of population-level representational invariance remains coarse. Applied topology is a promising tool to discover invariant structure in large datasets. Here, we use topological techniques to characterize and compare the spatiotemporal pattern of coactive spiking within populations of simultaneously recorded neurons in the secondary auditory region caudal medial neostriatum of European starlings (Sturnus vulgaris). We show that the pattern of population spike train coactivity carries stimulus-specific structure that is not reducible to that of individual neurons. We then introduce a topology-based similarity measure for population coactivity that is sensitive to invariant stimulus structure and show that this measure captures invariant neural representations tied to the learned relationships between natural vocalizations. This demonstrates one mechanism whereby emergent stimulus properties can be encoded in population activity, and shows the potential of applied topology for understanding invariant representations in neural populations.SIGNIFICANCE STATEMENT Information in neural populations is carried by the temporal patterns of spikes. We applied novel mathematical tools from the field of algebraic topology to quantify the structure of these temporal patterns. We found that, in a secondary auditory region of a songbird, these patterns reflected invariant information about a learned stimulus relationship. These results demonstrate that topology provides a novel approach for characterizing neural responses that is sensitive to invariant relationships that are critical for the perception of natural stimuli.


Subject(s)
Auditory Cortex/physiology , Electrophysiological Phenomena , Songbirds/physiology , Starlings/physiology , Acoustic Stimulation , Algorithms , Animals , Auditory Pathways/cytology , Auditory Pathways/physiology , Conditioning, Operant , Evoked Potentials, Auditory/physiology , Female , Male , Models, Neurological , Neostriatum/cytology , Neostriatum/physiology , Neurons/physiology , Vocalization, Animal/physiology
8.
Nature ; 586(7828): 262-269, 2020 10.
Article in English | MEDLINE | ID: mdl-32999462

ABSTRACT

Primates and rodents, which descended from a common ancestor around 90 million years ago1, exhibit profound differences in behaviour and cognitive capacity; the cellular basis for these differences is unknown. Here we use single-nucleus RNA sequencing to profile RNA expression in 188,776 individual interneurons across homologous brain regions from three primates (human, macaque and marmoset), a rodent (mouse) and a weasel (ferret). Homologous interneuron types-which were readily identified by their RNA-expression patterns-varied in abundance and RNA expression among ferrets, mice and primates, but varied less among primates. Only a modest fraction of the genes identified as 'markers' of specific interneuron subtypes in any one species had this property in another species. In the primate neocortex, dozens of genes showed spatial expression gradients among interneurons of the same type, which suggests that regional variation in cortical contexts shapes the RNA expression patterns of adult neocortical interneurons. We found that an interneuron type that was previously associated with the mouse hippocampus-the 'ivy cell', which has neurogliaform characteristics-has become abundant across the neocortex of humans, macaques and marmosets but not mice or ferrets. We also found a notable subcortical innovation: an abundant striatal interneuron type in primates that had no molecularly homologous counterpart in mice or ferrets. These interneurons expressed a unique combination of genes that encode transcription factors, receptors and neuropeptides and constituted around 30% of striatal interneurons in marmosets and humans.


Subject(s)
Interneurons/cytology , Primates , Animals , Callithrix , Cerebral Cortex/cytology , Female , Ferrets , Hippocampus/cytology , Humans , Interneurons/metabolism , LIM-Homeodomain Proteins/metabolism , Lysosomal Membrane Proteins/metabolism , Macaca , Male , Mice , Neostriatum/cytology , Nerve Tissue Proteins/metabolism , RNA/genetics , Species Specificity , Transcription Factors/metabolism
9.
Nature ; 582(7813): 550-556, 2020 06.
Article in English | MEDLINE | ID: mdl-32581380

ABSTRACT

Parkinson's disease is characterized by loss of dopamine neurons in the substantia nigra1. Similar to other major neurodegenerative disorders, there are no disease-modifying treatments for Parkinson's disease. While most treatment strategies aim to prevent neuronal loss or protect vulnerable neuronal circuits, a potential alternative is to replace lost neurons to reconstruct disrupted circuits2. Here we report an efficient one-step conversion of isolated mouse and human astrocytes to functional neurons by depleting the RNA-binding protein PTB (also known as PTBP1). Applying this approach to the mouse brain, we demonstrate progressive conversion of astrocytes to new neurons that innervate into and repopulate endogenous neural circuits. Astrocytes from different brain regions are converted to different neuronal subtypes. Using a chemically induced model of Parkinson's disease in mouse, we show conversion of midbrain astrocytes to dopaminergic neurons, which provide axons to reconstruct the nigrostriatal circuit. Notably, re-innervation of striatum is accompanied by restoration of dopamine levels and rescue of motor deficits. A similar reversal of disease phenotype is also accomplished by converting astrocytes to neurons using antisense oligonucleotides to transiently suppress PTB. These findings identify a potentially powerful and clinically feasible approach to treating neurodegeneration by replacing lost neurons.


Subject(s)
Astrocytes/cytology , Disease Models, Animal , Dopaminergic Neurons/cytology , Parkinson Disease/pathology , Parkinson Disease/therapy , Substantia Nigra/cytology , Substantia Nigra/physiology , Animals , Axons/physiology , Dopamine/biosynthesis , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Female , Heterogeneous-Nuclear Ribonucleoproteins/deficiency , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , In Vitro Techniques , Male , Mice , Neostriatum/cytology , Neostriatum/physiology , Neural Pathways , Neurogenesis , Parkinson Disease/metabolism , Phenotype , Polypyrimidine Tract-Binding Protein/deficiency , Polypyrimidine Tract-Binding Protein/genetics , Polypyrimidine Tract-Binding Protein/metabolism , Substantia Nigra/metabolism
10.
Nat Commun ; 11(1): 1957, 2020 04 23.
Article in English | MEDLINE | ID: mdl-32327644

ABSTRACT

Action control is a key brain function determining the survival of animals in their environment. In mammals, neurons expressing dopamine D2 receptors (D2R) in the dorsal striatum (DS) and the nucleus accumbens (Acb) jointly but differentially contribute to the fine regulation of movement. However, their region-specific molecular features are presently unknown. By combining RNAseq of striatal D2R neurons and histological analyses, we identified hundreds of novel region-specific molecular markers, which may serve as tools to target selective subpopulations. As a proof of concept, we characterized the molecular identity of a subcircuit defined by WFS1 neurons and evaluated multiple behavioral tasks after its temporally-controlled deletion of D2R. Consequently, conditional D2R knockout mice displayed a significant reduction in digging behavior and an exacerbated hyperlocomotor response to amphetamine. Thus, targeted molecular analyses reveal an unforeseen heterogeneity in D2R-expressing striatal neuronal populations, underlying specific D2R's functional features in the control of specific motor behaviors.


Subject(s)
Neostriatum/cytology , Neurons/physiology , Nucleus Accumbens/cytology , Receptors, Dopamine D2/metabolism , Amphetamine/pharmacology , Animals , Biomarkers/metabolism , Corpus Striatum/cytology , Corpus Striatum/metabolism , Corpus Striatum/physiology , Dopamine Agents/pharmacology , Membrane Proteins/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/genetics , Neostriatum/metabolism , Neostriatum/physiology , Neural Pathways , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Nucleus Accumbens/metabolism , Nucleus Accumbens/physiology , Receptors, Dopamine D2/genetics
11.
Neuron ; 106(2): 277-290.e6, 2020 04 22.
Article in English | MEDLINE | ID: mdl-32075716

ABSTRACT

Substantia nigra dopamine neurons have been implicated in the initiation and invigoration of movement, presumably through their modulation of striatal projection neuron (SPN) activity. However, the impact of native dopaminergic transmission on SPN excitability has not been directly demonstrated. Using perforated patch-clamp recording, we found that optogenetic stimulation of nigrostriatal dopamine axons rapidly and persistently elevated the excitability of D1 receptor-expressing SPNs (D1-SPNs). The evoked firing of D1-SPNs increased within hundreds of milliseconds of stimulation and remained elevated for ≥ 10 min. Consistent with the negative modulation of depolarization- and Ca2+-activated K+ currents, dopaminergic transmission accelerated subthreshold depolarization in response to current injection, reduced the latency to fire, and transiently diminished action potential afterhyperpolarization. Persistent modulation was protein kinase A dependent and associated with a reduction in action potential threshold. Together, these data demonstrate that dopaminergic transmission potently increases D1-SPN excitability with a time course that could support subsecond and sustained behavioral control.


Subject(s)
Dopamine/physiology , Neostriatum/physiology , Neurons/physiology , Neurotransmitter Agents/physiology , Receptors, Dopamine D1/physiology , Synaptic Transmission/physiology , Animals , Electrophysiological Phenomena , Female , Male , Mice , Neostriatum/cytology , Neostriatum/metabolism , Optogenetics , Patch-Clamp Techniques
12.
Nature ; 576(7787): 446-451, 2019 12.
Article in English | MEDLINE | ID: mdl-31801999

ABSTRACT

Individual neurons in many cortical regions have been found to encode specific, identifiable features of the environment or body that pertain to the function of the region1-3. However, in frontal cortex, which is involved in cognition, neural responses display baffling complexity, carrying seemingly disordered mixtures of sensory, motor and other task-related variables4-13. This complexity has led to the suggestion that representations in individual frontal neurons are randomly mixed and can only be understood at the neural population level14,15. Here we show that neural activity in rat orbitofrontal cortex (OFC) is instead highly structured: single neuron activity co-varies with individual variables in computational models that explain choice behaviour. To characterize neural responses across a large behavioural space, we trained rats on a behavioural task that combines perceptual and value-guided decisions. An unbiased, model-free clustering analysis identified distinct groups of OFC neurons, each with a particular response profile in task-variable space. Applying a simple model of choice behaviour to these categorical response profiles revealed that each profile quantitatively corresponds to a specific decision variable, such as decision confidence. Additionally, we demonstrate that a connectivity-defined cell type, orbitofrontal neurons projecting to the striatum, carries a selective and temporally sustained representation of a single decision variable: integrated value. We propose that neurons in frontal cortex, as in other cortical regions, form a sparse and overcomplete representation of features relevant to the region's function, and that they distribute this information selectively to downstream regions to support behaviour.


Subject(s)
Choice Behavior/physiology , Neurons/cytology , Neurons/physiology , Prefrontal Cortex/cytology , Animals , Anticipation, Psychological , Discrimination Learning , Logic , Male , Models, Neurological , Neostriatum/cytology , Neostriatum/physiology , Neural Pathways , Odorants/analysis , Organ Specificity , Prefrontal Cortex/anatomy & histology , Prefrontal Cortex/physiology , Psychometrics , Rats , Rats, Long-Evans , Reward
13.
Neuron ; 103(6): 1056-1072.e6, 2019 09 25.
Article in English | MEDLINE | ID: mdl-31324539

ABSTRACT

Motor and cognitive functions depend on the coordinated interactions between dopamine (DA) and acetylcholine (ACh) at striatal synapses. Increased ACh availability was assumed to accompany DA deficiency based on the outcome of pharmacological treatments and measurements in animals that were critically depleted of DA. Using Slc6a3DTR/+ diphtheria-toxin-sensitive mice, we demonstrate that a progressive and L-dopa-responsive DA deficiency reduces ACh availability and the transcription of hyperpolarization-activated cation (HCN) channels that encode the spike timing of ACh-releasing tonically active striatal interneurons (ChIs). Although the production and release of ACh and DA are reduced, the preponderance of ACh over DA contributes to the motor deficit. The increase in striatal ACh relative to DA is heightened via D1-type DA receptors that activate ChIs in response to DA release from residual axons. These results suggest that stabilizing the expression of HCN channels may improve ACh-DA reciprocity and motor function in Parkinson's disease (PD). VIDEO ABSTRACT.


Subject(s)
Acetylcholine/metabolism , Cholinergic Neurons/metabolism , Dopamine/deficiency , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Interneurons/metabolism , Neostriatum/metabolism , Parkinson Disease/metabolism , Amphetamine/pharmacology , Animals , Cholinergic Neurons/drug effects , Cholinergic Neurons/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Dopamine/metabolism , Dopamine Agents/pharmacology , Dopamine Plasma Membrane Transport Proteins/genetics , Interneurons/drug effects , Interneurons/physiology , Mice , Neostriatum/cytology , Neostriatum/drug effects , Neostriatum/physiopathology , Parkinson Disease/physiopathology , Patch-Clamp Techniques , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Transcription, Genetic
14.
J Neurosci ; 39(24): 4727-4737, 2019 06 12.
Article in English | MEDLINE | ID: mdl-30952811

ABSTRACT

The main excitatory inputs to the striatum arising from the cortex and the thalamus innervate both striatal spiny projection neurons and interneurons. These glutamatergic inputs to striatal GABAergic interneurons have been suggested to regulate the spike timing of striatal projection neurons via feedforward inhibition. Understanding how different excitatory inputs are integrated within the striatal circuitry and how they regulate striatal output is crucial for understanding basal ganglia function and related behaviors. Here, using VGLUT2 mice from both sexes, we report the existence of a glutamatergic projection from the mesencephalic locomotor region to the striatum that avoids the spiny neurons and selectively innervates interneurons. Specifically, optogenetic activation of glutamatergic axons from the pedunculopontine nucleus induced monosynaptic excitation in most recorded striatal cholinergic interneurons and GABAergic fast-spiking interneurons. Optogenetic stimulation in awake head-fixed mice consistently induced an increase in the firing rate of putative cholinergic interneurons and fast-spiking interneurons. In contrast, this stimulation did not induce excitatory responses in spiny neurons but rather disynaptic inhibitory responses ex vivo and a decrease in their firing rate in vivo, suggesting a feedforward mechanism mediating the inhibition of spiny projection neurons through the selective activation of striatal interneurons. Furthermore, unilateral stimulation of pedunculopontine nucleus glutamatergic axons in the striatum induced ipsilateral head rotations consistent with the inhibition of striatal output neurons. Our results demonstrate the existence of a unique interneuron-specific midbrain glutamatergic input to the striatum that exclusively recruits feedforward inhibition mechanisms.SIGNIFICANCE STATEMENT Glutamatergic inputs to the striatum have been shown to target both striatal projection neurons and interneurons and have been proposed to regulate spike timing of the projection neurons in part through feedforward inhibition. Here, we reveal the existence of a midbrain source of glutamatergic innervation to the striatum, originating in the pedunculopontine nucleus. Remarkably, this novel input selectively targets striatal interneurons, avoiding the projection neurons. Furthermore, we show that this selective innervation of interneurons can regulate the firing of the spiny projection neurons and inhibit the striatal output via feedforward inhibition. Together, our results describe a unique source of excitatory innervation to the striatum which selectively recruits feedforward inhibition of spiny neurons without any accompanying excitation.


Subject(s)
Interneurons/physiology , Neostriatum/cytology , Neostriatum/physiology , Neural Inhibition/physiology , Neurons/physiology , Pedunculopontine Tegmental Nucleus/cytology , Pedunculopontine Tegmental Nucleus/physiology , gamma-Aminobutyric Acid/physiology , Animals , Animals, Genetically Modified , Axons/physiology , Basal Ganglia/physiology , Female , Locomotion/physiology , Male , Mesencephalon/physiology , Mice , Nerve Net/cytology , Nerve Net/physiology , Optogenetics , Parasympathetic Nervous System/physiology , Vesicular Glutamate Transport Protein 2/genetics
15.
Eur J Neurosci ; 50(4): 2653-2662, 2019 08.
Article in English | MEDLINE | ID: mdl-30941837

ABSTRACT

The striatum mediates a broad range of cognitive and motor functions. Within the striatum, recently discovered tyrosine hydroxylase expressing interneurons (THINs) provide a source of intrastriatal synaptic connectivity that is critical for regulating striatal activity, yet the role of THIN's in behavior remains unknown. Given the important role of the striatum in reward-based behaviors, we investigated whether loss of striatal THINs would impact instrumental behavior in mice. We selectively ablated striatal THINs in TH-Cre mice using chemogenetic techniques, and then tested THIN-lesioned or control mice on three reward-based striatal-dependent instrumental tests: (a) progressive ratio test; (b) choice test following selective-satiety induced outcome devaluation; (c) outcome reinstatement test. Both striatal-THIN-lesioned and control mice acquired an instrumental response for flavored food pellets, and their behavior did not differ in the progressive ratio test, suggesting intact effort to obtain rewards. However, striatal THIN lesions markedly impaired choice performance following selective-satiety induced outcome devaluation. Unlike control mice, THIN-lesioned mice did not adjust their choice of actions following a change in outcome value. In the outcome reinstatement test THIN-lesioned and control mice showed response invigoration by outcome presentation, suggesting the incentive properties of outcomes were not disrupted by THIN lesions. Overall, we found that striatal THIN lesions selectively impaired goal-directed behavior, while preserving motoric and appetitive behaviors. These findings are the first to describe a function of striatal THINs in reward-based behavior, and further illustrate the important role for intrastriatal interneuronal connectivity in behavioral functions ascribed to the striatum more generally.


Subject(s)
Conditioning, Operant , Interneurons/pathology , Neostriatum/physiopathology , Tyrosine 3-Monooxygenase/metabolism , Animals , Appetitive Behavior , Choice Behavior , Extinction, Psychological , Goals , Interneurons/enzymology , Male , Mice , Mice, Transgenic , Motor Activity , Neostriatum/cytology , Neostriatum/enzymology , Psychomotor Performance , Reinforcement Schedule , Reward
16.
J Neurosci ; 39(15): 2965-2975, 2019 04 10.
Article in English | MEDLINE | ID: mdl-30737313

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.


Subject(s)
Compulsive Behavior/physiopathology , Compulsive Behavior/psychology , Motor Cortex/physiopathology , Neostriatum/physiopathology , Animals , Excitatory Postsynaptic Potentials , Female , Interneurons/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Motor Cortex/cytology , Neostriatum/cytology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Neural Pathways/physiology , Neurons , Optogenetics , Synapses
17.
Neuroscience ; 401: 106-116, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30668973

ABSTRACT

The striatum of the basal ganglia is pivotal for voluntary movements and is implicated in debilitating movement disorders such as Parkinsonism and dystonia. Striatum projects to downstream nuclei through direct (dSPN) and indirect (iSPN) pathway projection neurons thought to exert opposite effects on movement. In rodent models of striatal function, unilateral dopamine deprivation induces ipsiversive rotational behavior. The dSPNs of the dorsal striatum are believed to engage distinct motor programs but underlying mechanisms remain unclear. Here, we show by employing chemogenetics [Designer Receptors Exclusively Activated by Designer Drugs (DREADDs)] that unilateral inhibition of dorsomedial dSPNs is sufficient to selectively impair contraversive movement and elicit ipsiversive rotational behavior in mice. Adeno-associated virus (AAV) encoding Cre-dependent Gi-coupled DREADD was injected unilaterally into the dorsomedial striatum of Drd1-Cre mice, resulting in expression of the modified human M4 muscarinic receptor (hM4Di) in ∼20% of dorsostriatal dSPNs. Upon hM4Di activation, a striking positive linear correlation was found between turn ratio and viral expression, which corroborates a relationship between unilateral inhibition of dorsomedial dSPNs and rotational behavior. Bursts of ipsiversive rotations were interspersed with normal ambulation. However, partial unilateral inhibition of ∼20% of dorsostriatal dSPNs did not affect horizontal and vertical locomotion or forelimb use preference. Overall, our results substantiate a unique role of dSPNs in promoting response bias in rotational behavior and show this to be a highly sensitive measure of dSPN performance.


Subject(s)
Designer Drugs/pharmacology , Neostriatum/physiology , Neural Pathways/physiology , Neurons/physiology , Animals , Basal Ganglia/metabolism , Behavior, Animal , Corpus Striatum/cytology , Corpus Striatum/metabolism , Humans , Mice , Mice, Knockout , Mice, Transgenic , Motor Activity/drug effects , Neostriatum/cytology , Neostriatum/drug effects , Neural Pathways/drug effects , Neurites/metabolism , Neurons/cytology , Neurons/drug effects , Parkinsonian Disorders/metabolism , Receptor, Muscarinic M4/metabolism , Receptors, Dopamine D1/metabolism , Rotation
18.
Eur J Neurosci ; 49(5): 726-736, 2019 03.
Article in English | MEDLINE | ID: mdl-29603470

ABSTRACT

Dopamine-dependent synaptic plasticity is a candidate mechanism for reinforcement learning. A silent eligibility trace - initiated by synaptic activity and transformed into synaptic strengthening by later action of dopamine - has been hypothesized to explain the retroactive effect of dopamine in reinforcing past behaviour. We tested this hypothesis by measuring time-dependent modulation of synaptic plasticity by dopamine in adult mouse striatum, using whole-cell recordings. Presynaptic activity followed by postsynaptic action potentials (pre-post) caused spike-timing-dependent long-term depression in D1-expressing neurons, but not in D2 neurons, and not if postsynaptic activity followed presynaptic activity. Subsequent experiments focused on D1 neurons. Applying a dopamine D1 receptor agonist during induction of pre-post plasticity caused long-term potentiation. This long-term potentiation was hidden by long-term depression occurring concurrently and was unmasked when long-term depression blocked an L-type calcium channel antagonist. Long-term potentiation was blocked by a Ca2+ -permeable AMPA receptor antagonist but not by an NMDA antagonist or an L-type calcium channel antagonist. Pre-post stimulation caused transient elevation of rectification - a marker for expression of Ca2+ -permeable AMPA receptors - for 2-4-s after stimulation. To test for an eligibility trace, dopamine was uncaged at specific time points before and after pre- and postsynaptic conjunction of activity. Dopamine caused potentiation selectively at synapses that were active 2-s before dopamine release, but not at earlier or later times. Our results provide direct evidence for a silent eligibility trace in the synapses of striatal neurons. This dopamine-timing-dependent plasticity may play a central role in reinforcement learning.


Subject(s)
Dopamine/physiology , Neostriatum/physiopathology , Neuronal Plasticity/physiology , Neurons/physiology , Reinforcement, Psychology , Animals , Behavior, Animal/physiology , Calcium/metabolism , Dopamine/metabolism , Mice , Mice, Transgenic , Microscopy, Fluorescence, Multiphoton , Neostriatum/cytology , Neostriatum/metabolism , Neurons/metabolism , Patch-Clamp Techniques , Receptors, Dopamine D1 , Receptors, Dopamine D2
19.
Cell Transplant ; 28(3): 248-261, 2019 03.
Article in English | MEDLINE | ID: mdl-29807460

ABSTRACT

The aim of this work was to determine the effect of nicotine desensitization on dopamine (DA) release in the dorsal striatum and shell of the nucleus accumbens (NAc) from brain slices. In vitro fast-scan cyclic voltammetry analysis was used to evaluate dopamine release in the dorsal striatum and the NAc shell of Sprague-Dawley rats after infusion of nicotine, a nicotinic acetylcholine receptor (nAChR) antagonist mecamylamine (Mec), and an α4ß2 cholinergic receptor antagonist (DHße). DA release related to nicotine desensitization in the striatum and NAc shell was compared. In both structures, tonic release was suppressed by inhibition of the nicotine receptor (via Mec) and the α4ß2 receptor (via DHße). Paired-pulse ratio (PPR) was facilitated in both structures after nicotine and Mec infusion, and this facilitation was suppressed by increasing the stimulation interval. After variable frequency stimulation (simulating phasic burst), nicotine infusion induced significant augmentation of DA release in the striatum that was not seen in the absence of nicotine. In contrast, nicotine reduced phasic DA release in NAc, although frequency augmentation was seen both with and without nicotine. Evaluation of DA release evoked by various trains (high-frequency stimulation (HFS) 100 Hz) of high-frequency stimulation revealed significant enhancement after a train of three or more pulses in the striatum and NAc. The concentration differences between tonic and phasic release related to nicotine desensitization were more pronounced in the NAc shell. Nicotine desensitization is associated with suppression of tonic release of DA in both the striatum and NAc shell that may occur via the α4ß2 subtype of nAChR, whereas phasic frequency-dependent augmentation and HFS-related gating release is more pronounced in the striatum than in the NAc shell. Differences between phasic and tonic release associated with nicotine desensitization may underlie processing of reward signals in the NAc shell, and this may have major implications for addictive behavior.


Subject(s)
Dopamine/metabolism , Neostriatum/metabolism , Nicotine/pharmacology , Nucleus Accumbens/metabolism , Receptors, Nicotinic/metabolism , Animals , Male , Neostriatum/cytology , Nucleus Accumbens/cytology , Rats , Rats, Sprague-Dawley
20.
Toxicology ; 411: 110-121, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30391265

ABSTRACT

Exposure to herbicides can induce long-term chronic adverse effects such as respiratory diseases, malignancies and neurodegenerative diseases. Oxadiazon, a pre-emergence or early post-emergence herbicide, despite its low acute toxicity, may induce liver cancer and may exert adverse effects on reproductive and on endocrine functions. Unlike other herbicides, there are no indications on neurotoxicity associated with long-term exposure to oxadiazon. Therefore, we have analyzed in primary neuronal precursor cells isolated from human striatal primordium the effects of non-cytotoxic doses of oxadiazon on neuronal cell differentiation and migration, and on the expression and activity of the mitochondrial aldehyde dehydrogenase 2 (ALDH2) and of the acylphosphatase (ACYP). ALDH2 activity protects neurons against neurotoxicity induced by toxic aldehydes during oxidative stress and plays a role in neurodegenerative conditions such as Alzheimer's disease and Parkinson's disease. ACYP is involved in ion transport, cell differentiation, programmed cell death and cancer, and increased levels of ACYP have been revealed in fibroblasts from patients affected by Alzheimer's disease. In this study we demonstrated that non-cytotoxic doses of oxadiazon were able to inhibit neuronal striatal cell migration and FGF2- and BDNF-dependent differentiation towards neuronal phenotype, and to inhibit the expression and activity of ALDH2 and to increase the expression and activity of ACYP2. In addition, we have provided evidence that in human primary neuronal precursor striatal cells the inhibitory effects of oxadiazon on cell migration and differentiation towards neuronal phenotype were achieved through modulation of ACYP2. Taken together, our findings reveal for the first time that oxadiazon could exert neurotoxic effects by impairing differentiative capabilities of primary neuronal cells and indicate that ALDH2 and ACYP2 are relevant molecular targets for the neurotoxic effects of oxadiazon, suggesting a potential role of this herbicide in the onset of neurodegenerative diseases.


Subject(s)
Acid Anhydride Hydrolases/biosynthesis , Aldehyde Dehydrogenase, Mitochondrial/biosynthesis , Gene Expression Regulation, Enzymologic/drug effects , Herbicides/toxicity , Neostriatum/enzymology , Neural Stem Cells/enzymology , Neurotoxicity Syndromes/enzymology , Oxadiazoles/toxicity , Acid Anhydride Hydrolases/antagonists & inhibitors , Aldehyde Dehydrogenase, Mitochondrial/antagonists & inhibitors , Cell Differentiation/drug effects , Cell Line , Cell Movement/drug effects , Comet Assay , Humans , Neostriatum/cytology , Neostriatum/drug effects , Neural Stem Cells/drug effects , Neurotoxicity Syndromes/pathology , Oxidative Stress/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...