Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Cell ; 186(18): 3862-3881.e28, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37572660

ABSTRACT

Male sexual behavior is innate and rewarding. Despite its centrality to reproduction, a molecularly specified neural circuit governing innate male sexual behavior and reward remains to be characterized. We have discovered a developmentally wired neural circuit necessary and sufficient for male mating. This circuit connects chemosensory input to BNSTprTac1 neurons, which innervate POATacr1 neurons that project to centers regulating motor output and reward. Epistasis studies demonstrate that BNSTprTac1 neurons are upstream of POATacr1 neurons, and BNSTprTac1-released substance P following mate recognition potentiates activation of POATacr1 neurons through Tacr1 to initiate mating. Experimental activation of POATacr1 neurons triggers mating, even in sexually satiated males, and it is rewarding, eliciting dopamine release and self-stimulation of these cells. Together, we have uncovered a neural circuit that governs the key aspects of innate male sexual behavior: motor displays, drive, and reward.


Subject(s)
Neural Pathways , Sexual Behavior, Animal , Animals , Male , Neurons/physiology , Reward , Sexual Behavior, Animal/physiology , Mice
2.
Cell ; 179(6): 1393-1408.e16, 2019 11 27.
Article in English | MEDLINE | ID: mdl-31735496

ABSTRACT

Behaviors are inextricably linked to internal state. We have identified a neural mechanism that links female sexual behavior with the estrus, the ovulatory phase of the estrous cycle. We find that progesterone-receptor (PR)-expressing neurons in the ventromedial hypothalamus (VMH) are active and required during this behavior. Activating these neurons, however, does not elicit sexual behavior in non-estrus females. We show that projections of PR+ VMH neurons to the anteroventral periventricular (AVPV) nucleus change across the 5-day mouse estrous cycle, with ∼3-fold more termini and functional connections during estrus. This cyclic increase in connectivity is found in adult females, but not males, and regulated by estrogen signaling in PR+ VMH neurons. We further show that these connections are essential for sexual behavior in receptive females. Thus, estrogen-regulated structural plasticity of behaviorally salient connections in the adult female brain links sexual behavior to the estrus phase of the estrous cycle.


Subject(s)
Nerve Net/physiology , Sexual Behavior, Animal/physiology , Animals , Estrogens/metabolism , Estrous Cycle/drug effects , Female , Gonadal Steroid Hormones/pharmacology , Hypothalamus, Anterior/physiology , Male , Mice, Inbred C57BL , Nerve Net/drug effects , Neuronal Plasticity/drug effects , Neurons/drug effects , Neurons/metabolism , Ovary/metabolism , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Receptors, Progesterone/metabolism , Sexual Behavior, Animal/drug effects , Signal Transduction/drug effects , Time Factors
5.
Cell ; 174(6): 1436-1449.e20, 2018 09 06.
Article in English | MEDLINE | ID: mdl-30146163

ABSTRACT

Synaptic vesicle and active zone proteins are required for synaptogenesis. The molecular mechanisms for coordinated synthesis of these proteins are not understood. Using forward genetic screens, we identified the conserved THO nuclear export complex (THOC) as an important regulator of presynapse development in C. elegans dopaminergic neurons. In THOC mutants, synaptic messenger RNAs are retained in the nucleus, resulting in dramatic decrease of synaptic protein expression, near complete loss of synapses, and compromised dopamine function. CRE binding protein (CREB) interacts with THOC to mark synaptic transcripts for efficient nuclear export. Deletion of Thoc5, a THOC subunit, in mouse dopaminergic neurons causes severe defects in synapse maintenance and subsequent neuronal death in the substantia nigra compacta. These cellular defects lead to abrogated dopamine release, ataxia, and animal death. Together, our results argue that nuclear export mechanisms can select specific mRNAs and be a rate-limiting step for neuronal differentiation and survival.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Dopaminergic Neurons/metabolism , Nuclear Proteins/genetics , Synapses/metabolism , Active Transport, Cell Nucleus , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Caenorhabditis elegans , Caenorhabditis elegans Proteins/metabolism , Calcium Signaling , Cell Nucleus/metabolism , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutagenesis , Mutation, Missense , Nuclear Proteins/deficiency , Nuclear Proteins/metabolism , Protein Subunits/deficiency , Protein Subunits/genetics , Protein Subunits/metabolism
6.
Nature ; 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39169185

ABSTRACT

Developmental myelination is a protracted process in the mammalian brain1. One theory for why oligodendrocytes mature so slowly posits that myelination may stabilize neuronal circuits and temper neuronal plasticity as animals age2-4. We tested this theory in the visual cortex, which has a well-defined critical period for experience-dependent neuronal plasticity5. During adolescence, visual experience modulated the rate of oligodendrocyte maturation in visual cortex. To determine whether oligodendrocyte maturation in turn regulates neuronal plasticity, we genetically blocked oligodendrocyte differentiation and myelination in adolescent mice. In adult mice lacking adolescent oligodendrogenesis, a brief period of monocular deprivation led to a significant decrease in visual cortex responses to the deprived eye, reminiscent of the plasticity normally restricted to adolescence. This enhanced functional plasticity was accompanied by a greater turnover of dendritic spines and coordinated reductions in spine size following deprivation. Furthermore, inhibitory synaptic transmission, which gates experience-dependent plasticity at the circuit level, was diminished in the absence of adolescent oligodendrogenesis. These results establish a critical role for oligodendrocytes in shaping the maturation and stabilization of cortical circuits and support the concept of developmental myelination acting as a functional brake on neuronal plasticity.

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.
Nat Methods ; 20(7): 1104-1113, 2023 07.
Article in English | MEDLINE | ID: mdl-37429962

ABSTRACT

Genetically encoded voltage indicators (GEVIs) enable optical recording of electrical signals in the brain, providing subthreshold sensitivity and temporal resolution not possible with calcium indicators. However, one- and two-photon voltage imaging over prolonged periods with the same GEVI has not yet been demonstrated. Here, we report engineering of ASAP family GEVIs to enhance photostability by inversion of the fluorescence-voltage relationship. Two of the resulting GEVIs, ASAP4b and ASAP4e, respond to 100-mV depolarizations with ≥180% fluorescence increases, compared with the 50% fluorescence decrease of the parental ASAP3. With standard microscopy equipment, ASAP4e enables single-trial detection of spikes in mice over the course of minutes. Unlike GEVIs previously used for one-photon voltage recordings, ASAP4b and ASAP4e also perform well under two-photon illumination. By imaging voltage and calcium simultaneously, we show that ASAP4b and ASAP4e can identify place cells and detect voltage spikes with better temporal resolution than commonly used calcium indicators. Thus, ASAP4b and ASAP4e extend the capabilities of voltage imaging to standard one- and two-photon microscopes while improving the duration of voltage recordings.


Subject(s)
Brain , Calcium , Animals , Mice , Lighting , Microscopy , Photons
9.
Mol Psychiatry ; 28(1): 434-447, 2023 01.
Article in English | MEDLINE | ID: mdl-36460726

ABSTRACT

Modulation of corticostriatal plasticity alters the information flow throughout basal ganglia circuits and represents a fundamental mechanism for motor learning, action selection, and reward. Synaptic plasticity in the striatal direct- and indirect-pathway spiny projection neurons (dSPNs and iSPNs) is regulated by two distinct networks of GPCR signaling cascades. While it is well-known that dopamine D2 and adenosine A2a receptors bi-directionally regulate iSPN plasticity, it remains unclear how D1 signaling modulation of synaptic plasticity is counteracted by dSPN-specific Gi signaling. Here, we show that striatal dynorphin selectively suppresses long-term potentiation (LTP) through Kappa Opioid Receptor (KOR) signaling in dSPNs. Both KOR antagonism and conditional deletion of dynorphin in dSPNs enhance LTP counterbalancing with different levels of D1 receptor activation. Behaviorally, mice lacking dynorphin in D1 neurons show comparable motor behavior and reward-based learning, but enhanced flexibility during reversal learning. These findings support a model in which D1R and KOR signaling bi-directionally modulate synaptic plasticity and behavior in the direct pathway.


Subject(s)
Corpus Striatum , Dynorphins , Mice , Animals , Dynorphins/metabolism , Corpus Striatum/metabolism , Basal Ganglia , Long-Term Potentiation , Neuronal Plasticity/physiology , Receptors, Opioid, kappa/genetics , Receptors, Dopamine D1/metabolism
10.
Nature ; 557(7704): 177-182, 2018 05.
Article in English | MEDLINE | ID: mdl-29720658

ABSTRACT

Loss of dopamine in Parkinson's disease is hypothesized to impede movement by inducing hypo- and hyperactivity in striatal spiny projection neurons (SPNs) of the direct (dSPNs) and indirect (iSPNs) pathways in the basal ganglia, respectively. The opposite imbalance might underlie hyperkinetic abnormalities, such as dyskinesia caused by treatment of Parkinson's disease with the dopamine precursor L-DOPA. Here we monitored thousands of SPNs in behaving mice, before and after dopamine depletion and during L-DOPA-induced dyskinesia. Normally, intermingled clusters of dSPNs and iSPNs coactivated before movement. Dopamine depletion unbalanced SPN activity rates and disrupted the movement-encoding iSPN clusters. Matching their clinical efficacy, L-DOPA or agonism of the D2 dopamine receptor reversed these abnormalities more effectively than agonism of the D1 dopamine receptor. The opposite pathophysiology arose in L-DOPA-induced dyskinesia, during which iSPNs showed hypoactivity and dSPNs showed unclustered hyperactivity. Therefore, both the spatiotemporal profiles and rates of SPN activity appear crucial to striatal function, and next-generation treatments for basal ganglia disorders should target both facets of striatal activity.


Subject(s)
Dopamine/metabolism , Dyskinesias/pathology , Dyskinesias/physiopathology , Neurons/metabolism , Parkinsonian Disorders/pathology , Parkinsonian Disorders/physiopathology , Animals , Calcium Signaling , Dopamine/deficiency , Dyskinesias/etiology , Dyskinesias/metabolism , Female , Levodopa/metabolism , Levodopa/pharmacology , Male , Mice , Models, Biological , Movement/drug effects , Neostriatum/metabolism , Neostriatum/pathology , Neostriatum/physiopathology , Parkinsonian Disorders/metabolism , Receptors, Dopamine D1/agonists , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/agonists , Receptors, Dopamine D2/metabolism
11.
J Neurosci ; 42(23): 4755-4765, 2022 06 08.
Article in English | MEDLINE | ID: mdl-35534227

ABSTRACT

Mutations in leucine-rich repeat kinase 2 (LRRK2) are the most common genetic cause of Parkinson's disease (PD), but the pathogenic mechanism underlying LRRK2 mutations remains unresolved. In this study, we investigate the consequence of inactivation of LRRK2 and its functional homolog LRRK1 in male and female mice up to 25 months of age using behavioral, neurochemical, neuropathological, and ultrastructural analyses. We report that LRRK1 and LRRK2 double knock-out (LRRK DKO) mice exhibit impaired motor coordination at 12 months of age before the onset of dopaminergic neuron loss in the substantia nigra (SNpc). Moreover, LRRK DKO mice develop age-dependent, progressive loss of dopaminergic terminals in the striatum. Evoked dopamine (DA) release measured by fast-scan cyclic voltammetry in the dorsal striatum is also reduced in the absence of LRRK. Furthermore, LRRK DKO mice at 20-25 months of age show substantial loss of dopaminergic neurons in the SNpc. The surviving SNpc neurons in LRRK DKO mice at 25 months of age accumulate large numbers of autophagic and autolysosomal vacuoles and are accompanied with microgliosis. Surprisingly, the cerebral cortex is unaffected, as shown by normal cortical volume and neuron number as well as unchanged number of apoptotic cells and microglia in LRRK DKO mice at 25 months. These findings show that loss of LRRK function causes impairments in motor coordination, degeneration of dopaminergic terminals, reduction of evoked DA release, and selective loss of dopaminergic neurons in the SNpc, indicating that LRRK DKO mice are unique models for better understanding dopaminergic neurodegeneration in PD.SIGNIFICANCE STATEMENT Our current study employs a genetic approach to uncover the normal function of the LRRK family in the brain during mouse life span. Our multidisciplinary analysis demonstrates a critical normal physiological role of LRRK in maintaining the integrity and function of dopaminergic terminals and neurons in the aging brain, and show that LRRK DKO mice recapitulate several key features of PD and provide unique mouse models for elucidating molecular mechanisms underlying dopaminergic neurodegeneration in PD.


Subject(s)
Motor Disorders , Parkinson Disease , Animals , Dopamine , Dopaminergic Neurons/physiology , Female , Leucine , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Male , Mice , Mice, Knockout , Motor Disorders/pathology , Parkinson Disease/genetics , Parkinson Disease/pathology
12.
J Biol Chem ; 298(4): 101674, 2022 04.
Article in English | MEDLINE | ID: mdl-35148987

ABSTRACT

Adeno-associated viruses (AAVs) targeting specific cell types are powerful tools for studying distinct cell types in the central nervous system (CNS). Cis-regulatory modules (CRMs), e.g., enhancers, are highly cell-type-specific and can be integrated into AAVs to render cell type specificity. Chromatin accessibility has been commonly used to nominate CRMs, which have then been incorporated into AAVs and tested for cell type specificity in the CNS. However, chromatin accessibility data alone cannot accurately annotate active CRMs, as many chromatin-accessible CRMs are not active and fail to drive gene expression in vivo. Using available large-scale datasets on chromatin accessibility, such as those published by the ENCODE project, here we explored strategies to increase efficiency in identifying active CRMs for AAV-based cell-type-specific labeling and manipulation. We found that prescreening of chromatin-accessible putative CRMs based on the density of cell-type-specific transcription factor binding sites (TFBSs) can significantly increase efficiency in identifying active CRMs. In addition, generation of synthetic CRMs by stitching chromatin-accessible regions flanking cell-type-specific genes can render cell type specificity in many cases. Using these straightforward strategies, we generated AAVs that can target the extensively studied interneuron and glial cell types in the retina and brain. Both strategies utilize available genomic datasets and can be employed to generate AAVs targeting specific cell types in CNS without conducting comprehensive screening and sequencing experiments, making a step forward in cell-type-specific research.


Subject(s)
Brain , Dependovirus , Retina , Staining and Labeling , Transcription Factors , Animals , Binding Sites , Brain/cytology , Brain/metabolism , Chromatin/genetics , Chromatin/metabolism , Dependovirus/genetics , Dependovirus/metabolism , Mice , Retina/cytology , Retina/metabolism , Staining and Labeling/methods , Transcription Factors/metabolism
13.
Opt Express ; 30(15): 26396-26406, 2022 Jul 18.
Article in English | MEDLINE | ID: mdl-36236832

ABSTRACT

Two-photon light-sheet fluorescence microscopy enables high-resolution imaging of neural activity in brain tissue at a high frame rate. Traditionally, light-sheet microscopy builds up a 3D stack by multiple depth scans with uniform spatial intervals, which substantially limits the volumetric imaging speed. Here, we introduce the depth random-access light-sheet microscopy, allowing rapid switching scanning depth for light-sheet imaging. With a low-cost electrically tunable lens and minimum modification of an existing two-photon light-sheet imaging instrument, we demonstrated fast random depth hopping light-sheet imaging at 100 frames per second in the live brain slice. Through depth random-access, calcium activities for an astrocyte were recorded on four user-selected detection planes at a refreshing rate of 25 Hz.


Subject(s)
Calcium , Lenses , Brain/diagnostic imaging , Microscopy, Fluorescence/methods , Photons
14.
Proc Natl Acad Sci U S A ; 114(36): E7612-E7621, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28827326

ABSTRACT

Striatal spiny projection neurons (SPNs) receive convergent excitatory synaptic inputs from the cortex and thalamus. Activation of spatially clustered and temporally synchronized excitatory inputs at the distal dendrites could trigger plateau potentials in SPNs. Such supralinear synaptic integration is crucial for dendritic computation. However, how plateau potentials interact with subsequent excitatory and inhibitory synaptic inputs remains unknown. By combining computational simulation, two-photon imaging, optogenetics, and dual-color uncaging of glutamate and GABA, we demonstrate that plateau potentials can broaden the spatiotemporal window for integrating excitatory inputs and promote spiking. The temporal window of spiking can be delicately controlled by GABAergic inhibition in a cell-type-specific manner. This subtle inhibitory control of plateau potential depends on the location and kinetics of the GABAergic inputs and is achieved by the balance between relief and reestablishment of NMDA receptor Mg2+ block. These findings represent a mechanism for controlling spatiotemporal synaptic integration in SPNs.


Subject(s)
Dendrites/physiology , Excitatory Postsynaptic Potentials/physiology , Neurons/physiology , Animals , Cerebral Cortex/metabolism , Cerebral Cortex/physiology , Dendrites/metabolism , Female , Glutamic Acid/metabolism , Male , Mice , Neural Pathways/metabolism , Neural Pathways/physiology , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism , Synapses/physiology , Synaptic Transmission/physiology , Thalamus/metabolism , Thalamus/physiology , gamma-Aminobutyric Acid/metabolism
15.
Nature ; 490(7419): 262-6, 2012 Oct 11.
Article in English | MEDLINE | ID: mdl-23034651

ABSTRACT

The substantia nigra pars compacta and ventral tegmental area contain the two largest populations of dopamine-releasing neurons in the mammalian brain. These neurons extend elaborate projections in the striatum, a large subcortical structure implicated in motor planning and reward-based learning. Phasic activation of dopaminergic neurons in response to salient or reward-predicting stimuli is thought to modulate striatal output through the release of dopamine to promote and reinforce motor action. Here we show that activation of dopamine neurons in striatal slices rapidly inhibits action potential firing in both direct- and indirect-pathway striatal projection neurons through vesicular release of the inhibitory transmitter GABA (γ-aminobutyric acid). GABA is released directly from dopaminergic axons but in a manner that is independent of the vesicular GABA transporter VGAT. Instead, GABA release requires activity of the vesicular monoamine transporter VMAT2, which is the vesicular transporter for dopamine. Furthermore, VMAT2 expression in GABAergic neurons lacking VGAT is sufficient to sustain GABA release. Thus, these findings expand the repertoire of synaptic mechanisms used by dopamine neurons to influence basal ganglia circuits, show a new substrate whose transport is dependent on VMAT2 and demonstrate that GABA can function as a bona fide co-transmitter in monoaminergic neurons.


Subject(s)
Corpus Striatum/metabolism , Dopaminergic Neurons/metabolism , GABA Plasma Membrane Transport Proteins/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Corpus Striatum/cytology , Gene Expression Profiling , Gene Expression Regulation , Gene Knock-In Techniques , Mice , Mice, Transgenic , Vesicular Monoamine Transport Proteins/metabolism
16.
Mov Disord ; 32(4): 487-497, 2017 04.
Article in English | MEDLINE | ID: mdl-28343366

ABSTRACT

In Parkinson's disease (PD), dopamine depletion causes major changes in the brain, resulting in the typical cardinal motor features of the disease. PD neuropathology has been restricted to postmortem examinations, which are limited to only a single time of PD progression. Models of PD in which dopamine tone in the brain is chemically or physically disrupted are valuable tools in understanding the mechanisms of the disease. The basal ganglia have been well studied in the context of PD, and circuit changes in response to dopamine loss have been linked to the motor dysfunctions in PD. However, the etiology of the cognitive dysfunctions that are comorbid in PD patients has remained unclear until now. In this article, we review recent studies exploring how dopamine depletion affects the motor cortex at the synaptic level. In particular, we highlight our recent findings on abnormal spine dynamics in the motor cortex of PD mouse models through in vivo time-lapse imaging and motor skill behavior assays. In combination with previous studies, a role of the motor cortex in skill learning and the impairment of this ability with the loss of dopamine are becoming more apparent. Taken together, we conclude with a discussion on the potential role for the motor cortex in PD, with the possibility of targeting the motor cortex for future PD therapeutics. © 2017 International Parkinson and Movement Disorder Society.


Subject(s)
Learning/physiology , Motor Cortex/pathology , Neuronal Plasticity/physiology , Parkinson Disease/pathology , Animals , Disease Models, Animal , Dopamine Agents/pharmacology , Dopamine Agents/therapeutic use , Humans , Motor Cortex/drug effects , Motor Skills Disorders/etiology , Neuronal Plasticity/drug effects , Parkinson Disease/complications , Parkinson Disease/drug therapy
17.
Neuron ; 112(15): 2486-2502, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39002543

ABSTRACT

One key function of the brain is to control our body's movements, allowing us to interact with the world around us. Yet, many motor behaviors are not innate but require learning through repeated practice. Among the brain's motor regions, the cortico-basal ganglia circuit is particularly crucial for acquiring and executing motor skills, and neuronal activity in these regions is directly linked to movement parameters. Cell-type-specific adaptations of activity patterns and synaptic connectivity support the learning of new motor skills. Functionally, neuronal activity sequences become structured and associated with learned movements. On the synaptic level, specific connections become potentiated during learning through mechanisms such as long-term synaptic plasticity and dendritic spine dynamics, which are thought to mediate functional circuit plasticity. These synaptic and circuit adaptations within the cortico-basal ganglia circuitry are thus critical for motor skill acquisition, and disruptions in this plasticity can contribute to movement disorders.


Subject(s)
Basal Ganglia , Learning , Motor Skills , Neuronal Plasticity , Basal Ganglia/physiology , Neuronal Plasticity/physiology , Learning/physiology , Humans , Animals , Motor Skills/physiology , Cerebral Cortex/physiology , Neural Pathways/physiology , Motor Cortex/physiology
18.
bioRxiv ; 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38915677

ABSTRACT

Motor skill learning induces long-lasting synaptic plasticity at not only the inputs, such as dendritic spines1-4, but also at the outputs to the striatum of motor cortical neurons5,6. However, very little is known about the activity and structural plasticity of corticostriatal axons during learning in the adult brain. Here, we used longitudinal in vivo two-photon imaging to monitor the activity and structure of thousands of corticostriatal axonal boutons in the dorsolateral striatum in awake mice. We found that learning a new motor skill induces dynamic regulation of axonal boutons. The activities of motor corticostriatal axonal boutons exhibited selectivity for rewarded movements (RM) and un-rewarded movements (UM). Strikingly, boutons on the same axonal branches showed diverse responses during behavior. Motor learning significantly increased the fraction of RM boutons and reduced the heterogeneity of bouton activities. Moreover, motor learning-induced profound structural dynamism in boutons. By combining structural and functional imaging, we identified that newly formed axonal boutons are more likely to exhibit selectivity for RM and are stabilized during motor learning, while UM boutons are selectively eliminated. Our results highlight a novel form of plasticity at corticostriatal axons induced by motor learning, indicating that motor corticostriatal axonal boutons undergo dynamic reorganization that facilitates the acquisition and execution of motor skills.

19.
bioRxiv ; 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38895486

ABSTRACT

The striatum is required for normal action selection, movement, and sensorimotor learning. Although action-specific striatal ensembles have been well documented, it is not well understood how these ensembles are formed and how their dynamics may evolve throughout motor learning. Here we used longitudinal 2-photon Ca2+ imaging of dorsal striatal neurons in head-fixed mice as they learned to self-generate locomotion. We observed a significant activation of both direct- and indirect-pathway spiny projection neurons (dSPNs and iSPNs, respectively) during early locomotion bouts and sessions that gradually decreased over time. For dSPNs, onset- and offset-ensembles were gradually refined from active motion-nonspecific cells. iSPN ensembles emerged from neurons initially active during opponent actions before becoming onset- or offset-specific. Our results show that as striatal ensembles are progressively refined, the number of active nonspecific striatal neurons decrease and the overall efficiency of the striatum information encoding for learned actions increases.

20.
Biosens Bioelectron ; 261: 116474, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38870827

ABSTRACT

Multichannel arrays capable of real-time sensing of neuromodulators in the brain are crucial for gaining insights into new aspects of neural communication. However, measuring neurochemicals, such as dopamine, at low concentrations over large areas has proven challenging. In this research, we demonstrate a novel approach that leverages the scalability and processing power offered by microelectrode array devices integrated with a functionalized, high-density microwire bundle, enabling electrochemical sensing at an unprecedented scale and spatial resolution. The sensors demonstrate outstanding selective molecular recognition by incorporating a selective polymeric membrane. By combining cutting-edge commercial multiplexing, digitization, and data acquisition hardware with a bio-compatible and highly sensitive neurochemical interface array, we establish a powerful platform for neurochemical analysis. This multichannel array has been successfully utilized in vitro and ex vivo systems. Notably, our results show a sensing area of 2.25 mm2 with an impressive detection limit of 820 pM for dopamine. This new approach paves the way for investigating complex neurochemical processes and holds promise for advancing our understanding of brain function and neurological disorders.


Subject(s)
Biosensing Techniques , Dopamine , Electrochemical Techniques , Limit of Detection , Microelectrodes , Biosensing Techniques/methods , Biosensing Techniques/instrumentation , Dopamine/analysis , Animals , Electrochemical Techniques/methods , Equipment Design , Brain/metabolism , Humans , Neurotransmitter Agents/analysis
SELECTION OF CITATIONS
SEARCH DETAIL