Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.057
Filter
1.
Sci Adv ; 10(18): eadm7039, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38701209

ABSTRACT

Long-range glutamatergic inputs originating from the cortex and thalamus are indispensable for striatal development, providing the foundation for motor and cognitive functions. Despite their significance, transcriptional regulation governing these inputs remains largely unknown. We investigated the role of a transcription factor encoded by a high-risk autism-associated gene, FOXP1, in sculpting glutamatergic inputs onto spiny projection neurons (SPNs) within the striatum. We find a neuron subtype-specific role of FOXP1 in strengthening and maturing glutamatergic inputs onto dopamine receptor 2-expressing SPNs (D2 SPNs). We also find that FOXP1 promotes synaptically driven excitability in these neurons. Using single-nuclei RNA sequencing, we identify candidate genes that mediate these cell-autonomous processes through postnatal FOXP1 function at the post-synapse. Last, we demonstrate that postnatal FOXP1 reinstatement rescues electrophysiological deficits, cell type-specific gene expression changes, and behavioral phenotypes. Together, this study enhances our understanding of striatal circuit development and provides proof of concept for a therapeutic approach for FOXP1 syndrome and other neurodevelopmental disorders.


Subject(s)
Corpus Striatum , Forkhead Transcription Factors , Neurons , Receptors, Dopamine D2 , Repressor Proteins , Animals , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Corpus Striatum/metabolism , Corpus Striatum/cytology , Mice , Neurons/metabolism , Repressor Proteins/metabolism , Repressor Proteins/genetics , Phenotype , Synapses/metabolism , Synapses/physiology , Male
2.
Nature ; 627(8003): 358-366, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38418885

ABSTRACT

Astrocytes are heterogeneous glial cells of the central nervous system1-3. However, the physiological relevance of astrocyte diversity for neural circuits and behaviour remains unclear. Here we show that a specific population of astrocytes in the central striatum expresses µ-crystallin (encoded by Crym in mice and CRYM in humans) that is associated with several human diseases, including neuropsychiatric disorders4-7. In adult mice, reducing the levels of µ-crystallin in striatal astrocytes through CRISPR-Cas9-mediated knockout of Crym resulted in perseverative behaviours, increased fast synaptic excitation in medium spiny neurons and dysfunctional excitatory-inhibitory synaptic balance. Increased perseveration stemmed from the loss of astrocyte-gated control of neurotransmitter release from presynaptic terminals of orbitofrontal cortex-striatum projections. We found that perseveration could be remedied using presynaptic inhibitory chemogenetics8, and that this treatment also corrected the synaptic deficits. Together, our findings reveal converging molecular, synaptic, circuit and behavioural mechanisms by which a molecularly defined and allocated population of striatal astrocytes gates perseveration phenotypes that accompany neuropsychiatric disorders9-12. Our data show that Crym-positive striatal astrocytes have key biological functions within the central nervous system, and uncover astrocyte-neuron interaction mechanisms that could be targeted in treatments for perseveration.


Subject(s)
Astrocytes , Corpus Striatum , Rumination, Cognitive , mu-Crystallins , Animals , Humans , Mice , Astrocytes/metabolism , Corpus Striatum/cytology , Corpus Striatum/physiology , Gene Editing , Gene Knockout Techniques , mu-Crystallins/deficiency , mu-Crystallins/genetics , mu-Crystallins/metabolism , Rumination, Cognitive/physiology , Synaptic Transmission , CRISPR-Cas Systems , Medium Spiny Neurons/metabolism , Synapses/metabolism , Prefrontal Cortex/cytology , Prefrontal Cortex/metabolism , Presynaptic Terminals/metabolism , Neural Inhibition
3.
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
4.
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
5.
Neuropharmacology ; 232: 109527, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37011784

ABSTRACT

Parvalbumin-expressing dorsal striatal fast-spiking interneurons, comprising ∼1% of the total dorsal striatal neuronal population, are necessary for the expression of compulsive-like ethanol consumption mice. Fast-spiking interneurons are driven to fire by glutamatergic inputs derived primarily from the cortex. However, these neurons also receive substantial GABAergic input from two sources: the globus pallidus and the reticular nucleus of the thalamus. How ethanol modulates inhibitory input onto fast-spiking neurons is unclear and, more broadly, alcohol effects on GABAergic synaptic transmission onto GABAergic interneurons are understudied. Examining this, we found that acute bath application of ethanol (50 mM) potentiated GABAergic transmission from both the globus pallidus and the reticular nucleus of the thalamus onto fast-spiking interneurons in mouse of both sexes. This ethanol-induced potentiation required postsynaptic calcium and was not accompanied by a sustained change in presynaptic GABA release probability. Examining whether this ethanol effect persisted following chronic intermittent ethanol exposure, we found attenuated acute-ethanol potentiation of GABAergic transmission from both the globus pallidus and the reticular nucleus of the thalamus onto striatal fast-spiking interneurons. These data underscore the impact of ethanol on GABAergic signaling in the dorsal striatum and support the notion that ethanol may disinhibit the dorsolateral striatum.


Subject(s)
Corpus Striatum , Ethanol , GABAergic Neurons , Interneurons , Animals , Female , Male , Mice , Corpus Striatum/cytology , Corpus Striatum/drug effects , Ethanol/administration & dosage , Ethanol/pharmacology , GABAergic Neurons/drug effects , GABAergic Neurons/metabolism , Globus Pallidus/cytology , Globus Pallidus/drug effects , Interneurons/drug effects , Synaptic Transmission/drug effects , Thalamic Nuclei/cytology , Thalamic Nuclei/drug effects , Thalamic Nuclei/metabolism , Synapses/drug effects , Synapses/metabolism , Calcium/metabolism
6.
J Biol Chem ; 299(2): 102900, 2023 02.
Article in English | MEDLINE | ID: mdl-36640864

ABSTRACT

Extracellular dopamine (DA) levels are constrained by the presynaptic DA transporter (DAT), a major psychostimulant target. Despite its necessity for DA neurotransmission, DAT regulation in situ is poorly understood, and it is unknown whether regulated DAT trafficking impacts dopaminergic signaling and/or behaviors. Leveraging chemogenetics and conditional gene silencing, we found that activating presynaptic Gq-coupled receptors, either hM3Dq or mGlu5, drove rapid biphasic DAT membrane trafficking in ex vivo striatal slices, with region-specific differences between ventral and dorsal striata. DAT insertion required D2 DA autoreceptors and intact retromer, whereas DAT retrieval required PKC activation and Rit2. Ex vivo voltammetric studies revealed that DAT trafficking impacts DA clearance. Furthermore, dopaminergic mGlu5 silencing elevated DAT surface expression and abolished motor learning, which was rescued by inhibiting DAT with a subthreshold CE-158 dose. We discovered that presynaptic DAT trafficking is complex, multimodal, and region specific, and for the first time, we identified cell autonomous mechanisms that govern presynaptic DAT tone. Importantly, the findings are consistent with a role for regulated DAT trafficking in DA clearance and motor function.


Subject(s)
Corpus Striatum , Dopamine Plasma Membrane Transport Proteins , Dopamine , Dopamine/metabolism , Dopamine Plasma Membrane Transport Proteins/genetics , Dopamine Plasma Membrane Transport Proteins/metabolism , Receptors, Presynaptic/metabolism , Animals , Mice , Corpus Striatum/cytology , Corpus Striatum/physiology
7.
Nature ; 611(7937): 762-768, 2022 11.
Article in English | MEDLINE | ID: mdl-36352228

ABSTRACT

The canonical model of striatal function predicts that animal locomotion is associated with the opposing regulation of protein kinase A (PKA) in direct and indirect pathway striatal spiny projection neurons (SPNs) by dopamine1-7. However, the precise dynamics of PKA in dorsolateral SPNs during locomotion remain to be determined. It is also unclear whether other neuromodulators are involved. Here we show that PKA activity in both types of SPNs is essential for normal locomotion. Using two-photon fluorescence lifetime imaging8-10 of a PKA sensor10 through gradient index lenses, we measured PKA activity within individual SPNs of the mouse dorsolateral striatum during locomotion. Consistent with the canonical view, dopamine activated PKA activity in direct pathway SPNs during locomotion through the dopamine D1 receptor. However, indirect pathway SPNs exhibited a greater increase in PKA activity, which was largely abolished through the blockade of adenosine A2A receptors. In agreement with these results, fibre photometry measurements of an adenosine sensor11 revealed an acute increase in extracellular adenosine during locomotion. Functionally, antagonism of dopamine or adenosine receptors resulted in distinct changes in SPN PKA activity, neuronal activity and locomotion. Together, our results suggest that acute adenosine accumulation interplays with dopamine release to orchestrate PKA activity in SPNs and proper striatal function during animal locomotion.


Subject(s)
Adenosine , Corpus Striatum , Cyclic AMP-Dependent Protein Kinases , Dopamine , Locomotion , Neurons , Animals , Mice , Adenosine/metabolism , Corpus Striatum/cytology , Corpus Striatum/enzymology , Corpus Striatum/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Dopamine/metabolism , Locomotion/physiology , Neurons/enzymology , Neurons/metabolism , Receptors, Dopamine D1/metabolism , Receptor, Adenosine A2A/metabolism
8.
Nature ; 607(7919): 521-526, 2022 07.
Article in English | MEDLINE | ID: mdl-35794480

ABSTRACT

The direct and indirect pathways of the basal ganglia are classically thought to promote and suppress action, respectively1. However, the observed co-activation of striatal direct and indirect medium spiny neurons2 (dMSNs and iMSNs, respectively) has challenged this view. Here we study these circuits in mice performing an interval categorization task that requires a series of self-initiated and cued actions and, critically, a sustained period of dynamic action suppression. Although movement produced the co-activation of iMSNs and dMSNs in the sensorimotor, dorsolateral striatum (DLS), fibre photometry and photo-identified electrophysiological recordings revealed signatures of functional opponency between the two pathways during action suppression. Notably, optogenetic inhibition showed that DLS circuits were largely engaged to suppress-and not promote-action. Specifically, iMSNs on a given hemisphere were dynamically engaged to suppress tempting contralateral action. To understand how such regionally specific circuit function arose, we constructed a computational reinforcement learning model that reproduced key features of behaviour, neural activity and optogenetic inhibition. The model predicted that parallel striatal circuits outside the DLS learned the action-promoting functions, generating the temptation to act. Consistent with this, optogenetic inhibition experiments revealed that dMSNs in the associative, dorsomedial striatum, in contrast to those in the DLS, promote contralateral actions. These data highlight how opponent interactions between multiple circuit- and region-specific basal ganglia processes can lead to behavioural control, and establish a critical role for the sensorimotor indirect pathway in the proactive suppression of tempting actions.


Subject(s)
Corpus Striatum , Models, Neurological , Neural Inhibition , Neural Pathways , Neurons , Animals , Computer Simulation , Corpus Striatum/cytology , Corpus Striatum/physiology , Mice , Neural Pathways/cytology , Neural Pathways/physiology , Neurons/cytology , Neurons/physiology , Optogenetics
9.
Neuropathol Appl Neurobiol ; 48(5): e12812, 2022 08.
Article in English | MEDLINE | ID: mdl-35274336

ABSTRACT

AIMS: The striatum is mainly composed of projection neurons. It also contains interneurons, which modulate and control striatal output. The aim of the present study was to assess the percentages of projection neurons and interneuron populations in the striatum of control monkeys and of parkinsonian monkeys. METHODS: Unbiased stereology was used to estimate the volume density of every neuron population in the caudate, putamen and ventral striatum of control monkeys and of monkeys treated with MPTP, which results in striatal dopamine depletion. The various neuron population phenotypes were identified by immunohistochemistry. All analyses were performed within the same subjects using similar processing and analysis parameters, thus allowing for reliable data comparisons. RESULTS: In control monkeys, the projection neurons, which express the dopamine-and-cAMP-regulated-phosphoprotein, 32-KDa (DARPP-32), were the most abundant: ~86% of the total neurons counted. The interneurons accounted for the remaining 14%. Among the interneurons, those expressing calretinin were the most abundant (Cr+: ~57%; ~8% of the total striatal neurons counted), followed those expressing Parvalbumin (Pv+: ~18%; 2.6%), dinucleotide phosphate-diaphorase (NADPH+: ~13%; 1.8%), choline acetyltransferase (ChAT+: ~11%; 1.5%) and tyrosine hydroxylase (TH+: ~0.5%; 0.1%). No significant changes in volume densities occurred in any population following dopamine depletion, except for the TH+ interneurons, which increased in parkinsonian non-symptomatic monkeys and even more in symptomatic monkeys. CONCLUSIONS: These data are relevant for translational studies targeting specific neuron populations of the striatum. The fact that dopaminergic denervation does not cause neuron loss in any population has potential pathophysiological implications.


Subject(s)
Corpus Striatum , Dopamine , Interneurons , Neurons , Parkinsonian Disorders , Animals , Corpus Striatum/cytology , Corpus Striatum/pathology , Haplorhini , Interneurons/cytology , Neurons/cytology , Parkinsonian Disorders/physiopathology
10.
Proc Natl Acad Sci U S A ; 119(11): e2107339119, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35254903

ABSTRACT

SignificanceOutside the neurogenic niches, the adult brain lacks multipotent progenitor cells. In this study, we performed a series of in vivo screens and reveal that a single factor can induce resident brain astrocytes to become induced neural progenitor cells (iNPCs), which then generate neurons, astrocytes, and oligodendrocytes. Such a conclusion is supported by single-cell RNA sequencing and multiple lineage-tracing experiments. Our discovery of iNPCs is fundamentally important for regenerative medicine since neural injuries or degeneration often lead to loss/dysfunction of all three neural lineages. Our findings also provide insights into cell plasticity in the adult mammalian brain, which has largely lost the regenerative capacity.


Subject(s)
Astrocytes/cytology , Astrocytes/metabolism , Cell Differentiation , Cell Lineage , Cellular Reprogramming , Corpus Striatum/cytology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/genetics , Cell Lineage/genetics , Cellular Reprogramming/genetics , Corpus Striatum/metabolism , Fluorescent Antibody Technique , GABAergic Neurons/cytology , GABAergic Neurons/metabolism , Gene Expression , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Genes, Reporter , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis , RNA-Seq , Receptors, Notch/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
11.
J Pharmacol Sci ; 148(3): 326-330, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35177212

ABSTRACT

Extracellular signal-regulated protein kinase 5 (ERK5) has various physiological functions. However, the physiological role of ERK5 in the treatment of mice with an illicit drug such as methamphetamine (METH) remains unknown. We revealed that mice treated with METH showed hyperactivity, and increased p-ERK5 and Iba1 (a microglia marker) levels in the striatum. Additionally, these changes were inhibited by pretreatment with the ERK5 inhibitor BIX02189. The results suggest that METH-induced hyperactivity is associated with the activation of microglia via p-ERK5 in the striatum. Thus, the ERK5 pathway components in the central nervous system are potential therapeutic targets for preventing METH addiction.


Subject(s)
Aniline Compounds/pharmacology , Corpus Striatum/cytology , Hyperkinesis/chemically induced , Hyperkinesis/drug therapy , Indoles/pharmacology , Methantheline/adverse effects , Microglia/drug effects , Mitogen-Activated Protein Kinase 7/antagonists & inhibitors , Mitogen-Activated Protein Kinase 7/physiology , Aniline Compounds/therapeutic use , Animals , Calcium-Binding Proteins/metabolism , Corpus Striatum/metabolism , Indoles/therapeutic use , Mice , Microfilament Proteins/metabolism , Microglia/metabolism , Mitogen-Activated Protein Kinase 7/metabolism , Psychomotor Agitation , Substance-Related Disorders/prevention & control
12.
Neurosci Lett ; 771: 136466, 2022 02 06.
Article in English | MEDLINE | ID: mdl-35051434

ABSTRACT

Physical exercise is considered an adjuvant treatment to Parkinson's disease (PD) patients, possibly reducing inflammatory responses in the brain. Studies have stated that physical exercise protects dopaminergic neurons in PD models produced by neurotoxins. However, few studies focused on immunohistochemically reacted astrocytes and morphometric analyses of these cells in a PD mouse model submitted to physical exercise. We investigated the effects of treadmill training on striatal astrocytes of a PD mouse model combining immunohistochemistry and western-blotting for glial fibrillary acidic protein (GFAP) with morphometric analyses. Male Swiss mice were divided into 4 groups: sedentary control (SEDCONT), exercise control (EXERCONT), sedentary Parkinson (SEDPD), and exercise Parkinson (EXERPD). Stereotaxic bilateral injections of 6-hydroxydopamine into the striatum were adopted for PD groups. Striatal astrocytes showed increased GFAP in EXERPD, and we observed a higher level of GFAP in EXERPD than SEDPD. The number of primary and secondary processes was similar in striatal astrocytes of control groups and EXERPD. The astrocyte primary processes of SEDPD were larger than those of EXERPD, EXERCONT and SEDCONT. Cell body diameters and areas showed no difference between groups. We concluded that physical exercise influences striatal astrocytes in exercised parkinsonian mice.


Subject(s)
Astrocytes/metabolism , Corpus Striatum/physiopathology , Parkinson Disease/therapy , Physical Conditioning, Animal/methods , Animals , Corpus Striatum/cytology , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Male , Mice , Running
13.
Proc Natl Acad Sci U S A ; 119(1)2022 01 04.
Article in English | MEDLINE | ID: mdl-34934014

ABSTRACT

Cyclic adenosine monophosphate (cAMP) is a pivotal second messenger with an essential role in neuronal function. cAMP synthesis by adenylyl cyclases (AC) is controlled by G protein-coupled receptor (GPCR) signaling systems. However, the network of molecular players involved in the process is incompletely defined. Here, we used CRISPR/Cas9-based screening to identify that members of the potassium channel tetradimerization domain (KCTD) family are major regulators of cAMP signaling. Focusing on striatal neurons, we show that the dominant isoform KCTD5 exerts its effects through an unusual mechanism that modulates the influx of Zn2+ via the Zip14 transporter to exert unique allosteric effects on AC. We further show that KCTD5 controls the amplitude and sensitivity of stimulatory GPCR inputs to cAMP production by Gßγ-mediated AC regulation. Finally, we report that KCTD5 haploinsufficiency in mice leads to motor deficits that can be reversed by chelating Zn2+ Together, our findings uncover KCTD proteins as major regulators of neuronal cAMP signaling via diverse mechanisms.


Subject(s)
Cyclic AMP/metabolism , Potassium Channels/metabolism , Signal Transduction , Allosteric Regulation , Animals , Behavior, Animal , CRISPR-Cas Systems , Cation Transport Proteins/metabolism , Corpus Striatum/cytology , Corpus Striatum/metabolism , Cyclic AMP/biosynthesis , Humans , Mice , Neurons/metabolism , Receptors, G-Protein-Coupled/metabolism
14.
Cell Mol Neurobiol ; 42(1): 279-289, 2022 Jan.
Article in English | MEDLINE | ID: mdl-32445040

ABSTRACT

D-serine is synthesized by serine racemase (SR) and is a co-agonist at forebrain N-methyl-D-aspartate receptors (NMDARs). D-serine and SR are expressed primarily in neurons, but not in quiescent astrocytes. In this study, we examined the localization of D-serine and SR in the mouse striatum and the effects of genetically silencing SR expression in GABAergic interneurons (iSR-/-). iSR-/- mice had substantially reduced SR expression almost exclusively in striatum, but only exhibited marginal D-serine reduction. SR positive cells in the striatum showed strong co-localization with dopamine- and cyclic AMP-regulated neuronal phosphoprotein (DARPP32) in wild type mice. Transgenic fluorescent reporter mice for either the D1 or D2 dopamine receptors exhibited a 65:35 ratio for co-localization with D1and D2 receptor positive cells, respectively. These results indicate that GABAergic medium spiny neurons receiving dopaminergic inputs in striatum robustly and uniformly express SR. In behavioral tests, iSR-/- mice showed a blunted response to the hedonic and stimulant effects of cocaine, without affecting anxiety-related behaviors. Because the cocaine effects have been shown in the constitutive SR-/- mice, the restriction of the blunted response to cocaine to iSR-/- mice reinforces the conclusion that D-serine in striatal GABAergic neurons plays an important role in mediating dopaminergic stimulant effects. Results in this study suggest that SR in striatal GABAergic neurons is synthesizing D-serine, not as a glutamatergic co-transmitter, but rather as an autocrine whereby the GABAergic neurons control the excitability of their NMDARs by determining the availability of the co-agonist, D-serine.


Subject(s)
Neurons , Racemases and Epimerases , Animals , Corpus Striatum/cytology , Mice , Mice, Knockout , Neurons/enzymology , Racemases and Epimerases/metabolism , Serine/metabolism
15.
Physiol Rep ; 9(21): e15088, 2021 11.
Article in English | MEDLINE | ID: mdl-34762352

ABSTRACT

Neuronal networks cause changes in behaviorally important information processing through the vesicular release of neurotransmitters governed by the rate and timing of action potentials (APs). Herein, we provide evidence that dopamine (DA), nonquantally released from the cytoplasm, may exert similar effects in vivo. In mouse slice preparations, (+/-)-3,4-methylenedioxy-methamphetamine (MDMA, or ecstasy) and ß-phenylethylamine (ß-PEA)-induced DA release in the striatum and nucleus accumbens (NAc), two regions of the brain involved in reward-driven and social behavior and inhibited the axonal stimulation-induced release of tritiated acetylcholine ([3 H]ACh) in the striatum. The DA transporter (DAT) inhibitor (GBR-12909) prevented MDMA and ß-PEA from causing DA release. GBR-12909 could also restore some of the stimulated acetylcholine release reduced by MDMA or ß-PEA in the striatum confirming the fundamental role of DAT. In addition, hypothermia could prevent the ß-PEA-induced release in the striatum and in the NAc. Sulpiride, a D2 receptor antagonist, also prevented the inhibitory effects of MDMA or ß-PEA on stimulated ACh release, suggesting they act indirectly via binding of DA. Reflecting the neurochemical interactions in brain slices at higher system level, MDMA altered the social behavior of rats by preferentially enhancing passive social behavior. Similar to the in vitro effects, GBR-12909 treatment reversed specific elements of the MDMA-induced changes in behavior, such as passive social behavior, while left others including social play unchanged. The changes in behavior by the high level of extracellular DA-- a significant amount originating from cytoplasmic release--suggest that in addition to digital computation through synapses, the brain also uses analog communication, such as DA signaling, to mediate some elements of complex behaviors, but in a much longer time scale.


Subject(s)
Dopamine Plasma Membrane Transport Proteins/metabolism , Dopamine/metabolism , Serotonin/metabolism , Signal Transduction , Social Behavior , Animals , Corpus Striatum/cytology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dopamine D2 Receptor Antagonists/pharmacology , Male , Mice , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Nucleus Accumbens/cytology , Nucleus Accumbens/metabolism , Phenethylamines/pharmacology , Psychotropic Drugs/pharmacology , Rats , Rats, Wistar , Receptors, Dopamine D2/metabolism , Sulpiride/pharmacology
16.
Nature ; 599(7886): 645-649, 2021 11.
Article in English | MEDLINE | ID: mdl-34732888

ABSTRACT

The ability to suppress actions that lead to a negative outcome and explore alternative actions is necessary for optimal decision making. Although the basal ganglia have been implicated in these processes1-5, the circuit mechanisms underlying action selection and exploration remain unclear. Here, using a simple lateralized licking task, we show that indirect striatal projection neurons (iSPN) in the basal ganglia contribute to these processes through modulation of the superior colliculus (SC). Optogenetic activation of iSPNs suppresses contraversive licking and promotes ipsiversive licking. Activity in lateral superior colliculus (lSC), a region downstream of the basal ganglia, is necessary for task performance and predicts lick direction. Furthermore, iSPN activation suppresses ipsilateral lSC, but surprisingly excites contralateral lSC, explaining the emergence of ipsiversive licking. Optogenetic inactivation reveals inter-collicular competition whereby each hemisphere of the superior colliculus inhibits the other, thus allowing the indirect pathway to disinhibit the contralateral lSC and trigger licking. Finally, inactivating iSPNs impairs suppression of devalued but previously rewarded licking and reduces exploratory licking. Our results reveal that iSPNs engage the competitive interaction between lSC hemispheres to trigger a motor action and suggest a general circuit mechanism for exploration during action selection.


Subject(s)
Basal Ganglia/cytology , Basal Ganglia/physiology , Corpus Striatum/physiology , Exploratory Behavior/physiology , Neural Inhibition/physiology , Neural Pathways/physiology , Superior Colliculi/physiology , Animals , Behavior, Animal/physiology , Corpus Striatum/cytology , Decision Making , Female , Male , Mice , Neurons/physiology , Optogenetics , Reward , Superior Colliculi/cytology
17.
Sci Rep ; 11(1): 21395, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34725371

ABSTRACT

Recent studies have shown that temporal stability of the neuronal activity over time can be estimated by the structure of the spike-count autocorrelation of neuronal populations. This estimation, called the intrinsic timescale, has been computed for several cortical areas and can be used to propose a cortical hierarchy reflecting a scale of temporal receptive windows between areas. In this study, we performed an autocorrelation analysis on neuronal populations of three basal ganglia (BG) nuclei, including the striatum and the subthalamic nucleus (STN), the input structures of the BG, and the external globus pallidus (GPe). The analysis was performed during the baseline period of a motivational visuomotor task in which monkeys had to apply different amounts of force to receive different amounts of reward. We found that the striatum and the STN have longer intrinsic timescales than the GPe. Moreover, our results allow for the placement of these subcortical structures within the already-defined scale of cortical temporal receptive windows. Estimates of intrinsic timescales are important in adding further constraints in the development of computational models of the complex dynamics among these nuclei and throughout cortico-BG-thalamo-cortical loops.


Subject(s)
Basal Ganglia/physiology , Corpus Striatum/physiology , Nerve Net/physiology , Subthalamic Nucleus/physiology , Animals , Basal Ganglia/cytology , Cognition , Corpus Striatum/cytology , Globus Pallidus/cytology , Globus Pallidus/physiology , Macaca mulatta , Male , Nerve Net/cytology , Subthalamic Nucleus/cytology , Time Factors
18.
Cells ; 10(10)2021 10 17.
Article in English | MEDLINE | ID: mdl-34685760

ABSTRACT

Regulation of mitochondrial morphology and motility is critical for neurons, but the exact mechanisms are unclear. Here, we demonstrate that these mechanisms may involve collapsin response mediator protein 2 (CRMP2). CRMP2 is attached to neuronal mitochondria and binds to dynamin-related protein 1 (Drp1), Miro 2, and Kinesin 1 light chain (KLC1). Treating neurons with okadaic acid (OA), an inhibitor of phosphatases PP1 and PP2A, resulted in increased CRMP2 phosphorylation at Thr509/514, Ser522, and Thr555, and augmented Drp1 phosphorylation at Ser616. The CRMP2-binding small molecule (S)-lacosamide ((S)-LCM) prevented an OA-induced increase in CRMP2 phosphorylation at Thr509/514 and Ser522 but not at Thr555, and also failed to alleviate Drp1 phosphorylation. The increased CRMP2 phosphorylation correlated with decreased CRMP2 binding to Drp1, Miro 2, and KLC1. (S)-LCM rescued CRMP2 binding to Drp1 and Miro 2 but not to KLC1. In parallel with CRMP2 hyperphosphorylation, OA increased mitochondrial fission and suppressed mitochondrial traffic. (S)-LCM prevented OA-induced alterations in mitochondrial morphology and motility. Deletion of CRMP2 with a small interfering RNA (siRNA) resulted in increased mitochondrial fission and diminished mitochondrial traffic. Overall, our data suggest that the CRMP2 expression level and phosphorylation state are involved in regulating mitochondrial morphology and motility in neurons.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Mitochondria/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Animals , Cells, Cultured , Corpus Striatum/cytology , Gene Deletion , Kinesins , Mice , Mitochondrial Dynamics/drug effects , Neurons/drug effects , Okadaic Acid/pharmacology , Phosphoprotein Phosphatases/metabolism , Phosphorylation/drug effects
19.
Mol Pharmacol ; 100(6): 526-539, 2021 12.
Article in English | MEDLINE | ID: mdl-34503973

ABSTRACT

Genetically encoded biosensors can be used to track signaling events in living cells by measuring changes in fluorescence emitted by one or more fluorescent proteins. Here, we describe the use of genetically encoded biosensors based on Förster resonance energy transfer (FRET), combined with high-content microscopy, to image dynamic signaling events simultaneously in thousands of neurons in response to drug treatments. We first applied this approach to examine intercellular variation in signaling responses among cultured striatal neurons stimulated with multiple drugs. Using high-content FRET imaging and immunofluorescence, we identified neuronal subpopulations with unique responses to pharmacological manipulation and used nuclear morphology to identify medium spiny neurons within these heterogeneous striatal cultures. Focusing on protein kinase A (PKA) and extracellular signal-regulated kinase 1/2 (ERK1/2) signaling in the cytoplasm and nucleus, we noted pronounced intercellular differences among putative medium spiny neurons, in both the magnitude and kinetics of signaling responses to drug application. Importantly, a conventional "bulk" analysis that pooled all cells in culture yielded a different rank order of drug potency than that revealed by single-cell analysis. Using a single-cell analytical approach, we dissected the relative contributions of PKA and ERK1/2 signaling in striatal neurons and unexpectedly identified a novel role for ERK1/2 in promoting nuclear activation of PKA in striatal neurons. This finding adds a new dimension of signaling crosstalk between PKA and ERK1/2 with relevance to dopamine D1 receptor signaling in striatal neurons. In conclusion, high-content single-cell imaging can complement and extend traditional population-level analyses and provides a novel vantage point from which to study cellular signaling. SIGNIFICANCE STATEMENT: High-content imaging revealed substantial intercellular variation in the magnitude and pattern of intracellular signaling events driven by receptor stimulation. Since individual neurons within the same population can respond differently to a given agonist, interpreting measures of intracellular signaling derived from the averaged response of entire neuronal populations may not always reflect what happened at the single-cell level. This study uses this approach to identify a new form of cross-talk between PKA and ERK1/2 signaling in the nucleus of striatal neurons.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Fluorescence Resonance Energy Transfer/methods , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neurons/metabolism , Signal Transduction , Single-Cell Analysis/methods , Animals , Biosensing Techniques/methods , Cell Nucleus/metabolism , Cells, Cultured , Corpus Striatum/cytology , Enzyme Inhibitors/pharmacology , Female , Neurons/drug effects , Rats , Rats, Sprague-Dawley
20.
J Mol Biol ; 433(21): 167222, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34492254

ABSTRACT

Converging evidence points to the N-terminal domain comprising the first 17 amino acids of the Huntingtin protein (Nt17) as a key regulator of its aggregation, cellular properties and toxicity. In this study, we further investigated the interplay between Nt17 and the polyQ domain repeat length in regulating the aggregation and inclusion formation of exon 1 of the Huntingtin protein (Httex1). In addition, we investigated the effect of removing Nt17 or modulating its local structure on the membrane interactions, neuronal uptake, and toxicity of monomeric or fibrillar Httex1. Our results show that the polyQ and Nt17 domains synergistically modulate the aggregation propensity of Httex1 and that the Nt17 domain plays important roles in shaping the surface properties of mutant Httex1 fibrils and regulating their poly-Q-dependent growth, lateral association and neuronal uptake. Removal of Nt17 or disruption of its transient helical conformations slowed the aggregation of monomeric Httex1 in vitro, reduced inclusion formation in cells, enhanced the neuronal uptake and nuclear accumulation of monomeric Httex1 proteins, and was sufficient to prevent cell death induced by Httex1 72Q overexpression. Finally, we demonstrate that the uptake of Httex1 fibrils into primary neurons and the resulting toxicity are strongly influenced by mutations and phosphorylation events that influence the local helical propensity of Nt17. Altogether, our results demonstrate that the Nt17 domain serves as one of the key master regulators of Htt aggregation, internalization, and toxicity and represents an attractive target for inhibiting Htt aggregate formation, inclusion formation, and neuronal toxicity.


Subject(s)
Exons , Huntingtin Protein/chemistry , Mutation , Neurons/metabolism , Protein Aggregates , Animals , Cloning, Molecular , Corpus Striatum/cytology , Corpus Striatum/metabolism , Cryoelectron Microscopy , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HEK293 Cells , Humans , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Microscopy, Atomic Force , Neurons/cytology , Phosphorylation , Primary Cell Culture , Protein Conformation, alpha-Helical , Protein Engineering/methods , Protein Folding , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...