Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Science ; 383(6690): eabn3263, 2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38422184

ABSTRACT

Vocal production learning ("vocal learning") is a convergently evolved trait in vertebrates. To identify brain genomic elements associated with mammalian vocal learning, we integrated genomic, anatomical, and neurophysiological data from the Egyptian fruit bat (Rousettus aegyptiacus) with analyses of the genomes of 215 placental mammals. First, we identified a set of proteins evolving more slowly in vocal learners. Then, we discovered a vocal motor cortical region in the Egyptian fruit bat, an emergent vocal learner, and leveraged that knowledge to identify active cis-regulatory elements in the motor cortex of vocal learners. Machine learning methods applied to motor cortex open chromatin revealed 50 enhancers robustly associated with vocal learning whose activity tended to be lower in vocal learners. Our research implicates convergent losses of motor cortex regulatory elements in mammalian vocal learning evolution.


Subject(s)
Enhancer Elements, Genetic , Eutheria , Evolution, Molecular , Gene Expression Regulation , Motor Cortex , Motor Neurons , Proteins , Vocalization, Animal , Animals , Chiroptera/genetics , Chiroptera/physiology , Vocalization, Animal/physiology , Motor Cortex/cytology , Motor Cortex/physiology , Chromatin/metabolism , Motor Neurons/physiology , Larynx/physiology , Epigenesis, Genetic , Genome , Proteins/genetics , Proteins/metabolism , Amino Acid Sequence , Eutheria/genetics , Eutheria/physiology , Machine Learning
2.
Neuron ; 112(1): 56-72.e4, 2024 Jan 03.
Article in English | MEDLINE | ID: mdl-37909037

ABSTRACT

A comprehensive understanding of neuronal diversity and connectivity is essential for understanding the anatomical and cellular mechanisms that underlie functional contributions. With the advent of single-cell analysis, growing information regarding molecular profiles leads to the identification of more heterogeneous cell types. Therefore, the need for additional orthogonal recombinase systems is increasingly apparent, as heterogeneous tissues can be further partitioned into increasing numbers of specific cell types defined by multiple features. Critically, new recombinase systems should work together with pre-existing systems without cross-reactivity in vivo. Here, we introduce novel site-specific recombinase systems based on ΦC31 bacteriophage recombinase for labeling multiple cell types simultaneously and a novel viral strategy for versatile and robust intersectional expression of any transgene. Together, our system will help researchers specifically target different cell types with multiple features in the same animal.


Subject(s)
Integrases , Recombinases , Animals , Recombinases/genetics , Integrases/genetics , Genetic Vectors , Neurons/metabolism , Transgenes
3.
Nat Neurosci ; 26(12): 2182-2191, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37957318

ABSTRACT

The meta-reinforcement learning (meta-RL) framework, which involves RL over multiple timescales, has been successful in training deep RL models that generalize to new environments. It has been hypothesized that the prefrontal cortex may mediate meta-RL in the brain, but the evidence is scarce. Here we show that the orbitofrontal cortex (OFC) mediates meta-RL. We trained mice and deep RL models on a probabilistic reversal learning task across sessions during which they improved their trial-by-trial RL policy through meta-learning. Ca2+/calmodulin-dependent protein kinase II-dependent synaptic plasticity in OFC was necessary for this meta-learning but not for the within-session trial-by-trial RL in experts. After meta-learning, OFC activity robustly encoded value signals, and OFC inactivation impaired the RL behaviors. Longitudinal tracking of OFC activity revealed that meta-learning gradually shapes population value coding to guide the ongoing behavioral policy. Our results indicate that two distinct RL algorithms with distinct neural mechanisms and timescales coexist in OFC to support adaptive decision-making.


Subject(s)
Reinforcement, Psychology , Reward , Mice , Animals , Prefrontal Cortex/physiology , Reversal Learning/physiology
4.
Nat Neurosci ; 26(1): 79-91, 2023 01.
Article in English | MEDLINE | ID: mdl-36510113

ABSTRACT

Early-life trauma (ELT) is a risk factor for binge eating and obesity later in life, yet the neural circuits that underlie this association have not been addressed. Here, we show in mice that downregulation of the leptin receptor (Lepr) in the lateral hypothalamus (LH) and its effect on neural activity is crucial in causing ELT-induced binge-like eating and obesity upon high-fat diet exposure. We also found that the increased activity of Lepr-expressing LH (LHLepr) neurons encodes sustained binge-like eating in ELT mice. Inhibition of LHLepr neurons projecting to the ventrolateral periaqueductal gray normalizes these behavioral features of ELT mice. Furthermore, activation of proenkephalin-expressing ventrolateral periaqueductal gray neurons, which receive inhibitory inputs from LHLepr neurons, rescues ELT-induced maladaptive eating habits. Our results identify a circuit pathway that mediates ELT-induced maladaptive eating and may lead to the identification of novel therapeutic targets for binge eating and obesity.


Subject(s)
Hypothalamic Area, Lateral , Leptin , Mice , Animals , Leptin/metabolism , Hypothalamic Area, Lateral/metabolism , Feeding Behavior , Obesity/metabolism , Periaqueductal Gray , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Eating
5.
Nat Neurosci ; 25(6): 726-737, 2022 06.
Article in English | MEDLINE | ID: mdl-35654957

ABSTRACT

Learning induces the formation of new excitatory synapses in the form of dendritic spines, but their functional properties remain unknown. Here, using longitudinal in vivo two-photon imaging and correlated electron microscopy of dendritic spines in the motor cortex of mice during motor learning, we describe a framework for the formation, survival and resulting function of new, learning-related spines. Specifically, our data indicate that the formation of new spines during learning is guided by the potentiation of functionally clustered preexisting spines exhibiting task-related activity during earlier sessions of learning. We present evidence that this clustered potentiation induces the local outgrowth of multiple filopodia from the nearby dendrite, locally sampling the adjacent neuropil for potential axonal partners, likely via targeting preexisting presynaptic boutons. Successful connections are then selected for survival based on co-activity with nearby task-related spines, ensuring that the new spine preserves functional clustering. The resulting locally coherent activity of new spines signals the learned movement. Furthermore, we found that a majority of new spines synapse with axons previously unrepresented in these dendritic domains. Thus, learning involves the binding of new information streams into functional synaptic clusters to subserve learned behaviors.


Subject(s)
Learning , Synapses , Animals , Axons , Dendritic Spines , Mice , Neuropil , Presynaptic Terminals , Synapses/metabolism
6.
Nature ; 598(7879): 188-194, 2021 10.
Article in English | MEDLINE | ID: mdl-34616074

ABSTRACT

The cortico-basal ganglia-thalamo-cortical loop is one of the fundamental network motifs in the brain. Revealing its structural and functional organization is critical to understanding cognition, sensorimotor behaviour, and the natural history of many neurological and neuropsychiatric disorders. Classically, this network is conceptualized to contain three information channels: motor, limbic and associative1-4. Yet this three-channel view cannot explain the myriad functions of the basal ganglia. We previously subdivided the dorsal striatum into 29 functional domains on the basis of the topography of inputs from the entire cortex5. Here we map the multi-synaptic output pathways of these striatal domains through the globus pallidus external part (GPe), substantia nigra reticular part (SNr), thalamic nuclei and cortex. Accordingly, we identify 14 SNr and 36 GPe domains and a direct cortico-SNr projection. The striatonigral direct pathway displays a greater convergence of striatal inputs than the more parallel striatopallidal indirect pathway, although direct and indirect pathways originating from the same striatal domain ultimately converge onto the same postsynaptic SNr neurons. Following the SNr outputs, we delineate six domains in the parafascicular and ventromedial thalamic nuclei. Subsequently, we identify six parallel cortico-basal ganglia-thalamic subnetworks that sequentially transduce specific subsets of cortical information through every elemental node of the cortico-basal ganglia-thalamic loop. Thalamic domains relay this output back to the originating corticostriatal neurons of each subnetwork in a bona fide closed loop.


Subject(s)
Basal Ganglia/cytology , Cerebral Cortex/cytology , Neural Pathways , Neurons/cytology , Thalamus/cytology , Animals , Basal Ganglia/anatomy & histology , Cerebral Cortex/anatomy & histology , Male , Mice , Mice, Inbred C57BL , Thalamus/anatomy & histology
7.
Nat Neurosci ; 24(10): 1402-1413, 2021 10.
Article in English | MEDLINE | ID: mdl-34373644

ABSTRACT

Pain decreases the activity of many ventral tegmental area (VTA) dopamine (DA) neurons, yet the underlying neural circuitry connecting nociception and the DA system is not understood. Here we show that a subpopulation of lateral parabrachial (LPB) neurons is critical for relaying nociceptive signals from the spinal cord to the substantia nigra pars reticulata (SNR). SNR-projecting LPB neurons are activated by noxious stimuli and silencing them blocks pain responses in two different models of pain. LPB-targeted and nociception-recipient SNR neurons regulate VTA DA activity directly through feed-forward inhibition and indirectly by inhibiting a distinct subpopulation of VTA-projecting LPB neurons thereby reducing excitatory drive onto VTA DA neurons. Correspondingly, ablation of SNR-projecting LPB neurons is sufficient to reduce pain-mediated inhibition of DA release in vivo. The identification of a neural circuit conveying nociceptive input to DA neurons is critical to our understanding of how pain influences learning and behavior.


Subject(s)
Dopaminergic Neurons , Mesencephalon/physiopathology , Neural Pathways/physiopathology , Pain/physiopathology , Parabrachial Nucleus/physiopathology , Spinal Cord/physiopathology , Animals , Behavior, Animal , Brain Mapping , Male , Mice , Mice, Inbred C57BL , Neurons , Nociception , Optogenetics , Pain/psychology , Pain Management , Substantia Nigra/physiopathology , Ventral Tegmental Area/physiopathology
8.
eNeuro ; 8(4)2021.
Article in English | MEDLINE | ID: mdl-34348983

ABSTRACT

Animals precisely coordinate their left and right limbs for various adaptive purposes. While the left and right limbs are clearly controlled by different cortical hemispheres, the neural mechanisms that determine the action sequence between them remains elusive. Here, we have established a novel head-fixed bimanual-press (biPress) sequence task in which mice sequentially press left and right pedals with their forelimbs in a predetermined order. Using this motor task, we found that the motor cortical neurons responsible for the first press (1P) also generate independent motor signals for the second press (2P) by the opposite forelimb during the movement transitions between forelimbs. Projection-specific calcium imaging and optogenetic manipulation revealed these motor signals are transferred from one motor cortical hemisphere to the other via corticocortical projections. Together, our results suggest the motor cortices coordinate sequential bimanual movements through corticocortical pathways.


Subject(s)
Functional Laterality , Motor Cortex , Animals , Forelimb , Mice , Movement , Psychomotor Performance
9.
Neuron ; 109(13): 2165-2182.e10, 2021 07 07.
Article in English | MEDLINE | ID: mdl-34048697

ABSTRACT

Drugs of abuse induce persistent remodeling of reward circuit function, a process thought to underlie the emergence of drug craving and relapse to drug use. However, how circuit-specific, drug-induced molecular and cellular plasticity can have distributed effects on the mesolimbic dopamine reward system to facilitate relapse to drug use is not fully elucidated. Here, we demonstrate that dopamine receptor D3 (DRD3)-dependent plasticity in the ventral pallidum (VP) drives potentiation of dopamine release in the nucleus accumbens during relapse to cocaine seeking after abstinence. We show that two distinct VP DRD3+ neuronal populations projecting to either the lateral habenula (LHb) or the ventral tegmental area (VTA) display different patterns of activity during drug seeking following abstinence from cocaine self-administration and that selective suppression of elevated activity or DRD3 signaling in the LHb-projecting population reduces drug seeking. Together, our results uncover how circuit-specific DRD3-mediated plasticity contributes to the process of drug relapse.


Subject(s)
Basal Forebrain/physiology , Cocaine/administration & dosage , Dopamine/physiology , Drug-Seeking Behavior/physiology , Habenula/physiology , Neurons/physiology , Nucleus Accumbens/physiology , Receptors, Dopamine D3/physiology , Animals , Female , Male , Mice, Inbred C57BL , Mice, Transgenic , Reward , Ventral Tegmental Area/physiology
10.
Nature ; 593(7857): 108-113, 2021 05.
Article in English | MEDLINE | ID: mdl-33790464

ABSTRACT

Innate vocal sounds such as laughing, screaming or crying convey one's feelings to others. In many species, including humans, scaling the amplitude and duration of vocalizations is essential for effective social communication1-3. In mice, female scent triggers male mice to emit innate courtship ultrasonic vocalizations (USVs)4,5. However, whether mice flexibly scale their vocalizations and how neural circuits are structured to generate flexibility remain largely unknown. Here we identify mouse neurons from the lateral preoptic area (LPOA) that express oestrogen receptor 1 (LPOAESR1 neurons) and, when activated, elicit the complete repertoire of USV syllables emitted during natural courtship. Neural anatomy and functional data reveal a two-step, di-synaptic circuit motif in which primary long-range inhibitory LPOAESR1 neurons relieve a clamp of local periaqueductal grey (PAG) inhibition, enabling excitatory PAG USV-gating neurons to trigger vocalizations. We find that social context shapes a wide range of USV amplitudes and bout durations. This variability is absent when PAG neurons are stimulated directly; PAG-evoked vocalizations are time-locked to neural activity and stereotypically loud. By contrast, increasing the activity of LPOAESR1 neurons scales the amplitude of vocalizations, and delaying the recovery of the inhibition clamp prolongs USV bouts. Thus, the LPOA disinhibition motif contributes to flexible loudness and the duration and persistence of bouts, which are key aspects of effective vocal social communication.


Subject(s)
Hypothalamus/physiology , Vocalization, Animal/physiology , Animals , Courtship , Estrogen Receptor alpha/metabolism , Female , Hypothalamus/cytology , Male , Mice , Mice, Inbred BALB C , Neurons/physiology , Periaqueductal Gray/cytology , Periaqueductal Gray/physiology , Preoptic Area/cytology , Preoptic Area/physiology , Synapses/metabolism , Time Factors , Ultrasonic Waves
11.
Neuron ; 109(10): 1721-1738.e4, 2021 05 19.
Article in English | MEDLINE | ID: mdl-33823137

ABSTRACT

Basal ganglia play a central role in regulating behavior, but the organization of their outputs to other brain areas is incompletely understood. We investigate the largest output nucleus, the substantia nigra pars reticulata (SNr), and delineate the organization and physiology of its projection populations in mice. Using genetically targeted viral tracing and whole-brain anatomical analysis, we identify over 40 SNr targets that encompass a roughly 50-fold range of axonal densities. Retrograde tracing from the volumetrically largest targets indicates that the SNr contains segregated subpopulations that differentially project to functionally distinct brain stem regions. These subpopulations are electrophysiologically specialized and topographically organized and collateralize to common diencephalon targets, including the motor and intralaminar thalamus as well as the pedunculopontine nucleus and the midbrain reticular formation. These findings establish that SNr signaling is organized as dense, parallel outputs to specific brain stem targets concurrent with extensive collateral branches that encompass the majority of SNr axonal boutons.


Subject(s)
Basal Ganglia/cytology , Brain Stem/cytology , Diencephalon/cytology , Neurons/physiology , Animals , Basal Ganglia/physiology , Brain Stem/physiology , Diencephalon/physiology , Evoked Potentials , Mice , Mice, Inbred C57BL , Neural Pathways/cytology , Neural Pathways/physiology
12.
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
13.
Nat Neurosci ; 24(4): 504-515, 2021 04.
Article in English | MEDLINE | ID: mdl-33723433

ABSTRACT

The basal ganglia regulate a wide range of behaviors, including motor control and cognitive functions, and are profoundly affected in Parkinson's disease (PD). However, the functional organization of different basal ganglia nuclei has not been fully elucidated at the circuit level. In this study, we investigated the functional roles of distinct parvalbumin-expressing neuronal populations in the external globus pallidus (GPe-PV) and their contributions to different PD-related behaviors. We demonstrate that substantia nigra pars reticulata (SNr)-projecting GPe-PV neurons and parafascicular thalamus (PF)-projecting GPe-PV neurons are associated with locomotion and reversal learning, respectively. In a mouse model of PD, we found that selective manipulation of the SNr-projecting GPe-PV neurons alleviated locomotor deficit, whereas manipulation of the PF-projecting GPe-PV neurons rescued the impaired reversal learning. Our findings establish the behavioral importance of two distinct GPe-PV neuronal populations and, thereby, provide a new framework for understanding the circuit basis of different behavioral deficits in the Parkinsonian state.


Subject(s)
Globus Pallidus/physiopathology , Neural Pathways/physiopathology , Neurons/physiology , Parkinsonian Disorders/physiopathology , Animals , Female , Gait Disorders, Neurologic/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Parvalbumins , Reversal Learning/physiology
14.
Neuron ; 107(6): 992-994, 2020 09 23.
Article in English | MEDLINE | ID: mdl-32971000

ABSTRACT

Opioids are commonly used as analgesics for severe pain, but their addictive potential has sparked a misuse epidemic. In this issue of Neuron, Keyes et al. (2020) examine the contributions of distinct paraventricular thalamus (PVT) outputs to contextual opioid memories. They identify a PVT→NAc→LH circuit essential for recall of opioid experiences.


Subject(s)
Analgesics, Opioid , Opiate Alkaloids , Neural Pathways , Neurons , Thalamus
15.
J Neurosci ; 40(4): 743-768, 2020 01 22.
Article in English | MEDLINE | ID: mdl-31811030

ABSTRACT

Within the basal ganglia circuit, the external globus pallidus (GPe) is critically involved in motor control. Aside from Foxp2+ neurons and ChAT+ neurons that have been established as unique neuron types, there is little consensus on the classification of GPe neurons. Properties of the remaining neuron types are poorly defined. In this study, we leverage new mouse lines, viral tools, and molecular markers to better define GPe neuron subtypes. We found that Sox6 represents a novel, defining marker for GPe neuron subtypes. Lhx6+ neurons that lack the expression of Sox6 were devoid of both parvalbumin and Npas1. This result confirms previous assertions of the existence of a unique Lhx6+ population. Neurons that arise from the Dbx1+ lineage were similarly abundant in the GPe and displayed a heterogeneous makeup. Importantly, tracing experiments revealed that Npas1+-Nkx2.1+ neurons represent the principal noncholinergic, cortically-projecting neurons. In other words, they form the pallido-cortical arm of the cortico-pallido-cortical loop. Our data further show that pyramidal-tract neurons in the cortex collateralized within the GPe, forming a closed-loop system between the two brain structures. Overall, our findings reconcile some of the discrepancies that arose from differences in techniques or the reliance on preexisting tools. Although spatial distribution and electrophysiological properties of GPe neurons reaffirm the diversification of GPe subtypes, statistical analyses strongly support the notion that these neuron subtypes can be categorized under the two principal neuron classes: PV+ neurons and Npas1+ neurons.SIGNIFICANCE STATEMENT The poor understanding of the neuronal composition in the external globus pallidus (GPe) undermines our ability to interrogate its precise behavioral and disease involvements. In this study, 12 different genetic crosses were used, hundreds of neurons were electrophysiologically characterized, and >100,000 neurons were histologically- and/or anatomically-profiled. Our current study further establishes the segregation of GPe neuron classes and illustrates the complexity of GPe neurons in adult mice. Our results support the idea that Npas1+-Nkx2.1+ neurons are a distinct GPe neuron subclass. By providing a detailed analysis of the organization of the cortico-pallidal-cortical projection, our findings establish the cellular and circuit substrates that can be important for motor function and dysfunction.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cerebral Cortex/metabolism , Globus Pallidus/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Thyroid Nuclear Factor 1/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Neural Pathways/metabolism , Thyroid Nuclear Factor 1/genetics
16.
Neuron ; 104(5): 899-915.e8, 2019 12 04.
Article in English | MEDLINE | ID: mdl-31672263

ABSTRACT

Chronic stress (CS) is a major risk factor for the development of depression. Here, we demonstrate that CS-induced hyperactivity in ventral tegmental area (VTA)-projecting lateral habenula (LHb) neurons is associated with increased passive coping (PC), but not anxiety or anhedonia. LHb→VTA neurons in mice with increased PC show increased burst and tonic firing as well as synaptic adaptations in excitatory inputs from the entopeduncular nucleus (EP). In vivo manipulations of EP→LHb or LHb→VTA neurons selectively alter PC and effort-related motivation. Conversely, dorsal raphe (DR)-projecting LHb neurons do not show CS-induced hyperactivity and are targeted indirectly by the EP. Using single-cell transcriptomics, we reveal a set of genes that can collectively serve as biomarkers to identify mice with increased PC and differentiate LHb→VTA from LHb→DR neurons. Together, we provide a set of biological markers at the level of genes, synapses, cells, and circuits that define a distinctive CS-induced behavioral phenotype.


Subject(s)
Habenula/physiopathology , Motivation/physiology , Neurons , Psychological Distress , Animals , Behavior, Animal , Depression/etiology , Depression/physiopathology , Male , Mice , Mice, Inbred C57BL , Phenotype
17.
Neuron ; 104(6): 1126-1140.e6, 2019 12 18.
Article in English | MEDLINE | ID: mdl-31706697

ABSTRACT

The posterior parietal cortex (PPC) performs many functions, including decision making and movement control. It remains unknown which input and output pathways of PPC support different functions. We addressed this issue in mice, focusing on PPC neurons projecting to the dorsal striatum (PPC-STR) and the posterior secondary motor cortex (PPC-pM2). Projection-specific, retrograde labeling showed that PPC-STR and PPC-pM2 represent largely distinct subpopulations, with PPC-STR receiving stronger inputs from association areas and PPC-pM2 receiving stronger sensorimotor inputs. Two-photon calcium imaging during decision making revealed that the PPC-STR population encodes history-dependent choice bias more strongly than PPC-pM2 or general PPC populations. Furthermore, optogenetic inactivation of PPC-STR neurons or their terminals in STR decreased history-dependent bias, while inactivation of PPC-pM2 neurons altered movement kinematics. Therefore, PPC biases action selection through its STR projection while controlling movements through PPC-pM2 neurons. PPC may support multiple functions through parallel subpopulations, each with distinct input-output connectivity.


Subject(s)
Decision Making/physiology , Movement/physiology , Neural Pathways/physiology , Parietal Lobe/physiology , Animals , Corpus Striatum/physiology , Female , Male , Mice , Mice, Inbred C57BL , Motor Cortex/physiology
18.
Nat Biotechnol ; 2018 Dec 17.
Article in English | MEDLINE | ID: mdl-30556815

ABSTRACT

Understanding complex biological systems requires the system-wide characterization of both molecular and cellular features. Existing methods for spatial mapping of biomolecules in intact tissues suffer from information loss caused by degradation and tissue damage. We report a tissue transformation strategy named stabilization under harsh conditions via intramolecular epoxide linkages to prevent degradation (SHIELD), which uses a flexible polyepoxide to form controlled intra- and intermolecular cross-link with biomolecules. SHIELD preserves protein fluorescence and antigenicity, transcripts and tissue architecture under a wide range of harsh conditions. We applied SHIELD to interrogate system-level wiring, synaptic architecture, and molecular features of virally labeled neurons and their targets in mouse at single-cell resolution. We also demonstrated rapid three-dimensional phenotyping of core needle biopsies and human brain cells. SHIELD enables rapid, multiscale, integrated molecular phenotyping of both animal and clinical tissues.

19.
Nat Commun ; 9(1): 3701, 2018 09 12.
Article in English | MEDLINE | ID: mdl-30209250

ABSTRACT

Specialized cells in the medial entorhinal cortex (mEC), such as speed cells, head direction (HD) cells, and grid cells, are thought to support spatial navigation. To determine whether these computations are dependent on local circuits, we record neuronal activity in mEC layers II and III and optogenetically perturb locally projecting layer II pyramidal cells. We find that sharply tuned HD cells are only weakly responsive while speed, broadly tuned HD cells, and grid cells show pronounced transient excitatory and inhibitory responses. During the brief period of feedback inhibition, there is a reduction in specifically grid accuracy, which is corrected as firing rates return to baseline. These results suggest that sharp HD cells are embedded in a separate mEC sub-network from broad HD cells, speed cells, and grid cells. Furthermore, grid tuning is not only dependent on local processing but also rapidly updated by HD, speed, or other afferent inputs to mEC.


Subject(s)
Entorhinal Cortex/metabolism , Grid Cells/metabolism , Action Potentials/physiology , Animals , Female , Male , Pyramidal Cells/metabolism , Spatial Navigation/physiology
20.
Nat Neurosci ; 21(9): 1229-1238, 2018 09.
Article in English | MEDLINE | ID: mdl-30104734

ABSTRACT

Voluntary urination ensures that waste is eliminated when safe and socially appropriate, even without a pressing urge. Uncontrolled urination, or incontinence, is a common problem with few treatment options. Normal urine release requires a small region in the brainstem known as Barrington's nucleus (Bar), but specific neurons that relax the urethral sphincter and enable urine flow are unknown. Here we identify a small subset of Bar neurons that control the urethral sphincter in mice. These excitatory neurons express estrogen receptor 1 (BarESR1), project to sphincter-relaxing interneurons in the spinal cord and are active during natural urination. Optogenetic stimulation of BarESR1 neurons rapidly initiates sphincter bursting and efficient voiding in anesthetized and behaving animals. Conversely, optogenetic and chemogenetic inhibition reveals their necessity in motivated urination behavior. The identification of these cells provides an expanded model for the control of urination and its dysfunction.


Subject(s)
Brain Stem/physiology , Neurons/physiology , Urethra/innervation , Urethra/physiology , Urination/physiology , Animals , Brain Stem/cytology , Electromyography , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/physiology , Male , Mice , Neural Pathways/physiology , Odorants , Optogenetics , Urination Disorders/genetics , Urination Disorders/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...