Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 103
Filter
1.
Cell ; 186(3): 543-559.e19, 2023 02 02.
Article in English | MEDLINE | ID: mdl-36669484

ABSTRACT

Learning has been associated with modifications of synaptic and circuit properties, but the precise changes storing information in mammals have remained largely unclear. We combined genetically targeted voltage imaging with targeted optogenetic activation and silencing of pre- and post-synaptic neurons to study the mechanisms underlying hippocampal behavioral timescale plasticity. In mice navigating a virtual-reality environment, targeted optogenetic activation of individual CA1 cells at specific places induced stable representations of these places in the targeted cells. Optical elicitation, recording, and modulation of synaptic transmission in behaving mice revealed that activity in presynaptic CA2/3 cells was required for the induction of plasticity in CA1 and, furthermore, that during induction of these place fields in single CA1 cells, synaptic input from CA2/3 onto these same cells was potentiated. These results reveal synaptic implementation of hippocampal behavioral timescale plasticity and define a methodology to resolve synaptic plasticity during learning and memory in behaving mammals.


Subject(s)
CA1 Region, Hippocampal , Hippocampus , Mice , Animals , CA1 Region, Hippocampal/physiology , Hippocampus/physiology , Neuronal Plasticity/physiology , Learning/physiology , Neurons , Synaptic Transmission/physiology , Mammals
2.
Cell ; 186(20): 4325-4344.e26, 2023 09 28.
Article in English | MEDLINE | ID: mdl-37652010

ABSTRACT

KCR channelrhodopsins (K+-selective light-gated ion channels) have received attention as potential inhibitory optogenetic tools but more broadly pose a fundamental mystery regarding how their K+ selectivity is achieved. Here, we present 2.5-2.7 Å cryo-electron microscopy structures of HcKCR1 and HcKCR2 and of a structure-guided mutant with enhanced K+ selectivity. Structural, electrophysiological, computational, spectroscopic, and biochemical analyses reveal a distinctive mechanism for K+ selectivity; rather than forming the symmetrical filter of canonical K+ channels achieving both selectivity and dehydration, instead, three extracellular-vestibule residues within each monomer form a flexible asymmetric selectivity gate, while a distinct dehydration pathway extends intracellularly. Structural comparisons reveal a retinal-binding pocket that induces retinal rotation (accounting for HcKCR1/HcKCR2 spectral differences), and design of corresponding KCR variants with increased K+ selectivity (KALI-1/KALI-2) provides key advantages for optogenetic inhibition in vitro and in vivo. Thus, discovery of a mechanism for ion-channel K+ selectivity also provides a framework for next-generation optogenetics.


Subject(s)
Channelrhodopsins , Rhinosporidium , Humans , Channelrhodopsins/chemistry , Channelrhodopsins/genetics , Channelrhodopsins/metabolism , Channelrhodopsins/ultrastructure , Cryoelectron Microscopy , Ion Channels , Potassium/metabolism , Rhinosporidium/chemistry
3.
Cell ; 185(4): 654-671.e22, 2022 02 17.
Article in English | MEDLINE | ID: mdl-35065713

ABSTRACT

Sex hormones exert a profound influence on gendered behaviors. How individual sex hormone-responsive neuronal populations regulate diverse sex-typical behaviors is unclear. We performed orthogonal, genetically targeted sequencing of four estrogen receptor 1-expressing (Esr1+) populations and identified 1,415 genes expressed differentially between sexes or estrous states. Unique subsets of these genes were distributed across all 137 transcriptomically defined Esr1+ cell types, including estrous stage-specific ones, that comprise the four populations. We used differentially expressed genes labeling single Esr1+ cell types as entry points to functionally characterize two such cell types, BNSTprTac1/Esr1 and VMHvlCckar/Esr1. We observed that these two cell types, but not the other Esr1+ cell types in these populations, are essential for sex recognition in males and mating in females, respectively. Furthermore, VMHvlCckar/Esr1 cell type projections are distinct from those of other VMHvlEsr1 cell types. Together, projection and functional specialization of dimorphic cell types enables sex hormone-responsive populations to regulate diverse social behaviors.


Subject(s)
Estrous Cycle/genetics , Gene Expression Regulation , Sex Characteristics , Sexual Behavior, Animal/physiology , Aggression , Animals , Aromatase/metabolism , Autistic Disorder/genetics , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Gene Expression Profiling , HEK293 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neurons/metabolism , Social Behavior
4.
Cell ; 185(19): 3568-3587.e27, 2022 09 15.
Article in English | MEDLINE | ID: mdl-36113428

ABSTRACT

Computational analysis of cellular activity has developed largely independently of modern transcriptomic cell typology, but integrating these approaches may be essential for full insight into cellular-level mechanisms underlying brain function and dysfunction. Applying this approach to the habenula (a structure with diverse, intermingled molecular, anatomical, and computational features), we identified encoding of reward-predictive cues and reward outcomes in distinct genetically defined neural populations, including TH+ cells and Tac1+ cells. Data from genetically targeted recordings were used to train an optimized nonlinear dynamical systems model and revealed activity dynamics consistent with a line attractor. High-density, cell-type-specific electrophysiological recordings and optogenetic perturbation provided supporting evidence for this model. Reverse-engineering predicted how Tac1+ cells might integrate reward history, which was complemented by in vivo experimentation. This integrated approach describes a process by which data-driven computational models of population activity can generate and frame actionable hypotheses for cell-type-specific investigation in biological systems.


Subject(s)
Habenula , Reward , Population Dynamics
5.
Cell ; 185(4): 672-689.e23, 2022 02 17.
Article in English | MEDLINE | ID: mdl-35114111

ABSTRACT

ChRmine, a recently discovered pump-like cation-conducting channelrhodopsin, exhibits puzzling properties (large photocurrents, red-shifted spectrum, and extreme light sensitivity) that have created new opportunities in optogenetics. ChRmine and its homologs function as ion channels but, by primary sequence, more closely resemble ion pump rhodopsins; mechanisms for passive channel conduction in this family have remained mysterious. Here, we present the 2.0 Å resolution cryo-EM structure of ChRmine, revealing architectural features atypical for channelrhodopsins: trimeric assembly, a short transmembrane-helix 3, a twisting extracellular-loop 1, large vestibules within the monomer, and an opening at the trimer interface. We applied this structure to design three proteins (rsChRmine and hsChRmine, conferring further red-shifted and high-speed properties, respectively, and frChRmine, combining faster and more red-shifted performance) suitable for fundamental neuroscience opportunities. These results illuminate the conduction and gating of pump-like channelrhodopsins and point the way toward further structure-guided creation of channelrhodopsins for applications across biology.


Subject(s)
Channelrhodopsins/chemistry , Channelrhodopsins/metabolism , Ion Channel Gating , Animals , Channelrhodopsins/ultrastructure , Cryoelectron Microscopy , Female , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Models, Molecular , Optogenetics , Phylogeny , Rats, Sprague-Dawley , Schiff Bases/chemistry , Sf9 Cells , Structure-Activity Relationship
6.
Cell ; 184(14): 3731-3747.e21, 2021 07 08.
Article in English | MEDLINE | ID: mdl-34214470

ABSTRACT

In motor neuroscience, state changes are hypothesized to time-lock neural assemblies coordinating complex movements, but evidence for this remains slender. We tested whether a discrete change from more autonomous to coherent spiking underlies skilled movement by imaging cerebellar Purkinje neuron complex spikes in mice making targeted forelimb-reaches. As mice learned the task, millimeter-scale spatiotemporally coherent spiking emerged ipsilateral to the reaching forelimb, and consistent neural synchronization became predictive of kinematic stereotypy. Before reach onset, spiking switched from more disordered to internally time-locked concerted spiking and silence. Optogenetic manipulations of cerebellar feedback to the inferior olive bi-directionally modulated neural synchronization and reaching direction. A simple model explained the reorganization of spiking during reaching as reflecting a discrete bifurcation in olivary network dynamics. These findings argue that to prepare learned movements, olivo-cerebellar circuits enter a self-regulated, synchronized state promoting motor coordination. State changes facilitating behavioral transitions may generalize across neural systems.


Subject(s)
Movement/physiology , Nerve Net/physiology , Action Potentials/physiology , Animals , Calcium/metabolism , Cerebellum/physiology , Cortical Synchronization , Forelimb/physiology , Interneurons/physiology , Learning , Mice, Inbred C57BL , Mice, Transgenic , Models, Neurological , Motor Activity/physiology , Olivary Nucleus/physiology , Optogenetics , Purkinje Cells/physiology , Stereotyped Behavior , Task Performance and Analysis
7.
Cell ; 183(1): 211-227.e20, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32937106

ABSTRACT

The striosome compartment within the dorsal striatum has been implicated in reinforcement learning and regulation of motivation, but how striosomal neurons contribute to these functions remains elusive. Here, we show that a genetically identified striosomal population, which expresses the Teashirt family zinc finger 1 (Tshz1) and belongs to the direct pathway, drives negative reinforcement and is essential for aversive learning in mice. Contrasting a "conventional" striosomal direct pathway, the Tshz1 neurons cause aversion, movement suppression, and negative reinforcement once activated, and they receive a distinct set of synaptic inputs. These neurons are predominantly excited by punishment rather than reward and represent the anticipation of punishment or the motivation for avoidance. Furthermore, inhibiting these neurons impairs punishment-based learning without affecting reward learning or movement. These results establish a major role of striosomal neurons in behaviors reinforced by punishment and moreover uncover functions of the direct pathway unaccounted for in classic models.


Subject(s)
Avoidance Learning/physiology , Corpus Striatum/physiology , Homeodomain Proteins/genetics , Repressor Proteins/genetics , Animals , Basal Ganglia , Female , Homeodomain Proteins/metabolism , Learning/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motivation , Neurons/physiology , Punishment , Reinforcement, Psychology , Repressor Proteins/metabolism
8.
Cell ; 183(4): 918-934.e49, 2020 11 12.
Article in English | MEDLINE | ID: mdl-33113354

ABSTRACT

Learning valence-based responses to favorable and unfavorable options requires judgments of the relative value of the options, a process necessary for species survival. We found, using engineered mice, that circuit connectivity and function of the striosome compartment of the striatum are critical for this type of learning. Calcium imaging during valence-based learning exhibited a selective correlation between learning and striosomal but not matrix signals. This striosomal activity encoded discrimination learning and was correlated with task engagement, which, in turn, could be regulated by chemogenetic excitation and inhibition. Striosomal function during discrimination learning was disturbed with aging and severely so in a mouse model of Huntington's disease. Anatomical and functional connectivity of parvalbumin-positive, putative fast-spiking interneurons (FSIs) to striatal projection neurons was enhanced in striosomes compared with matrix in mice that learned. Computational modeling of these findings suggests that FSIs can modulate the striosomal signal-to-noise ratio, crucial for discrimination and learning.


Subject(s)
Aging/pathology , Corpus Striatum/pathology , Huntington Disease/pathology , Learning , Action Potentials , Animals , Behavior, Animal , Biomarkers/metabolism , Corpus Striatum/physiopathology , Discrimination Learning , Disease Models, Animal , Huntington Disease/physiopathology , Interneurons/pathology , Mice, Transgenic , Models, Neurological , Nerve Net/physiopathology , Parvalbumins/metabolism , Photometry , Reward , Task Performance and Analysis
10.
Cell ; 177(5): 1346-1360.e24, 2019 05 16.
Article in English | MEDLINE | ID: mdl-31080068

ABSTRACT

To decipher dynamic brain information processing, current genetically encoded calcium indicators (GECIs) are limited in single action potential (AP) detection speed, combinatorial spectral compatibility, and two-photon imaging depth. To address this, here, we rationally engineered a next-generation quadricolor GECI suite, XCaMPs. Single AP detection was achieved within 3-10 ms of spike onset, enabling measurements of fast-spike trains in parvalbumin (PV)-positive interneurons in the barrel cortex in vivo and recording three distinct (two inhibitory and one excitatory) ensembles during pre-motion activity in freely moving mice. In vivo paired recording of pre- and postsynaptic firing revealed spatiotemporal constraints of dendritic inhibition in layer 1 in vivo, between axons of somatostatin (SST)-positive interneurons and apical tufts dendrites of excitatory pyramidal neurons. Finally, non-invasive, subcortical imaging using red XCaMP-R uncovered somatosensation-evoked persistent activity in hippocampal CA1 neurons. Thus, the XCaMPs offer a critical enhancement of solution space in studies of complex neuronal circuit dynamics. VIDEO ABSTRACT.


Subject(s)
Action Potentials/physiology , Axons/metabolism , Cerebral Cortex/metabolism , Hippocampus/metabolism , Interneurons/metabolism , Pyramidal Cells/metabolism , Animals , Cerebral Cortex/cytology , Female , Hippocampus/cytology , Interneurons/cytology , Mice , Mice, Transgenic , Pyramidal Cells/cytology , Rats , Rats, Sprague-Dawley
11.
Cell ; 170(5): 1013-1027.e14, 2017 Aug 24.
Article in English | MEDLINE | ID: mdl-28823561

ABSTRACT

Reward-seeking behavior is fundamental to survival, but suppression of this behavior can be essential as well, even for rewards of high value. In humans and rodents, the medial prefrontal cortex (mPFC) has been implicated in suppressing reward seeking; however, despite vital significance in health and disease, the neural circuitry through which mPFC regulates reward seeking remains incompletely understood. Here, we show that a specific subset of superficial mPFC projections to a subfield of nucleus accumbens (NAc) neurons naturally encodes the decision to initiate or suppress reward seeking when faced with risk of punishment. A highly resolved subpopulation of these top-down projecting neurons, identified by 2-photon Ca2+ imaging and activity-dependent labeling to recruit the relevant neurons, was found capable of suppressing reward seeking. This natural activity-resolved mPFC-to-NAc projection displayed unique molecular-genetic and microcircuit-level features concordant with a conserved role in the regulation of reward-seeking behavior, providing cellular and anatomical identifiers of behavioral and possible therapeutic significance.


Subject(s)
Reward , Animals , Behavior, Animal , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Pathways , Neuroimaging , Prefrontal Cortex/cytology , Prefrontal Cortex/metabolism , Punishment
12.
Cell ; 165(7): 1776-1788, 2016 Jun 16.
Article in English | MEDLINE | ID: mdl-27238022

ABSTRACT

A major challenge in understanding the cellular diversity of the brain has been linking activity during behavior with standard cellular typology. For example, it has not been possible to determine whether principal neurons in prefrontal cortex active during distinct experiences represent separable cell types, and it is not known whether these differentially active cells exert distinct causal influences on behavior. Here, we develop quantitative hydrogel-based technologies to connect activity in cells reporting on behavioral experience with measures for both brain-wide wiring and molecular phenotype. We find that positive and negative-valence experiences in prefrontal cortex are represented by cell populations that differ in their causal impact on behavior, long-range wiring, and gene expression profiles, with the major discriminant being expression of the adaptation-linked gene NPAS4. These findings illuminate cellular logic of prefrontal cortex information processing and natural adaptive behavior and may point the way to cell-type-specific understanding and treatment of disease-associated states.


Subject(s)
Behavior, Animal , Brain Mapping/methods , Prefrontal Cortex/cytology , Animals , Appetitive Behavior , Basic Helix-Loop-Helix Transcription Factors/genetics , Cocaine/administration & dosage , Electroshock , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Prefrontal Cortex/metabolism
13.
Cell ; 160(3): 516-27, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25635459

ABSTRACT

Optimally orchestrating complex behavioral states, such as the pursuit and consumption of food, is critical for an organism's survival. The lateral hypothalamus (LH) is a neuroanatomical region essential for appetitive and consummatory behaviors, but whether individual neurons within the LH differentially contribute to these interconnected processes is unknown. Here, we show that selective optogenetic stimulation of a molecularly defined subset of LH GABAergic (Vgat-expressing) neurons enhances both appetitive and consummatory behaviors, whereas genetic ablation of these neurons reduced these phenotypes. Furthermore, this targeted LH subpopulation is distinct from cells containing the feeding-related neuropeptides, melanin-concentrating hormone (MCH), and orexin (Orx). Employing in vivo calcium imaging in freely behaving mice to record activity dynamics from hundreds of cells, we identified individual LH GABAergic neurons that preferentially encode aspects of either appetitive or consummatory behaviors, but rarely both. These tightly regulated, yet highly intertwined, behavioral processes are thus dissociable at the cellular level.


Subject(s)
Appetitive Behavior , Consummatory Behavior , Hypothalamus/physiology , Animals , Hypothalamic Hormones/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Melanins/metabolism , Mice , Motivation , Neurons/metabolism , Neuropeptides/metabolism , Orexins , Pituitary Hormones/metabolism , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism , gamma-Aminobutyric Acid/metabolism
14.
Nature ; 615(7951): 292-299, 2023 03.
Article in English | MEDLINE | ID: mdl-36859543

ABSTRACT

Emotional states influence bodily physiology, as exemplified in the top-down process by which anxiety causes faster beating of the heart1-3. However, whether an increased heart rate might itself induce anxiety or fear responses is unclear3-8. Physiological theories of emotion, proposed over a century ago, have considered that in general, there could be an important and even dominant flow of information from the body to the brain9. Here, to formally test this idea, we developed a noninvasive optogenetic pacemaker for precise, cell-type-specific control of cardiac rhythms of up to 900 beats per minute in freely moving mice, enabled by a wearable micro-LED harness and the systemic viral delivery of a potent pump-like channelrhodopsin. We found that optically evoked tachycardia potently enhanced anxiety-like behaviour, but crucially only in risky contexts, indicating that both central (brain) and peripheral (body) processes may be involved in the development of emotional states. To identify potential mechanisms, we used whole-brain activity screening and electrophysiology to find brain regions that were activated by imposed cardiac rhythms. We identified the posterior insular cortex as a potential mediator of bottom-up cardiac interoceptive processing, and found that optogenetic inhibition of this brain region attenuated the anxiety-like behaviour that was induced by optical cardiac pacing. Together, these findings reveal that cells of both the body and the brain must be considered together to understand the origins of emotional or affective states. More broadly, our results define a generalizable approach for noninvasive, temporally precise functional investigations of joint organism-wide interactions among targeted cells during behaviour.


Subject(s)
Behavior, Animal , Brain , Emotions , Heart , Animals , Mice , Anxiety/physiopathology , Brain/physiology , Brain Mapping , Emotions/physiology , Heart/physiology , Behavior, Animal/physiology , Electrophysiology , Optogenetics , Insular Cortex/physiology , Heart Rate , Channelrhodopsins , Tachycardia/physiopathology , Pacemaker, Artificial
15.
Cell ; 147(3): 678-89, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-22019004

ABSTRACT

Prevailing theory suggests that long-term memories are encoded via a two-phase process requiring early involvement of the hippocampus followed by the neocortex. Contextual fear memories in rodents rely on the hippocampus immediately following training but are unaffected by hippocampal lesions or pharmacological inhibition weeks later. With fast optogenetic methods, we examine the real-time contribution of hippocampal CA1 excitatory neurons to remote memory and find that contextual fear memory recall, even weeks after training, can be reversibly abolished by temporally precise optogenetic inhibition of CA1. When this inhibition is extended to match the typical time course of pharmacological inhibition, remote hippocampus dependence converts to hippocampus independence, suggesting that long-term memory retrieval normally depends on the hippocampus but can adaptively shift to alternate structures. Further revealing the plasticity of mechanisms required for memory recall, we confirm the remote-timescale importance of the anterior cingulate cortex (ACC) and implicate CA1 in ACC recruitment for remote recall.


Subject(s)
Hippocampus/physiology , Memory, Long-Term , Animals , Fear , Gyrus Cinguli/metabolism , Hippocampus/cytology , Humans , Mice , Mice, Inbred C57BL , Neurons/cytology , Neurons/physiology
16.
Mol Psychiatry ; 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38965421

ABSTRACT

The parasubthalamic nucleus (PSTN) is activated by refeeding after food deprivation and several PSTN subpopulations have been shown to suppress feeding. However, no study to date directly addressed the role of PSTN neurons activated upon food access in the control of ensuing food consumption. Here we identify consumption latency as a sensitive behavioral indicator of PSTN activity, and show that, in hungry mice, the ensemble of refeeding-activated PSTN neurons drastically increases the latency to initiate refeeding with both familiar and a novel, familiar food, but does not control the amount of food consumed. In thirsty mice, this ensemble also delays sucrose consumption but accelerates water consumption, possibly reflecting anticipatory prandial thirst, with again no influence on the amount of fluid consumed. We next sought to identify which subpopulations of PSTN neurons might be driving these latency effects, using cell-type and pathway-specific chemogenetic manipulations. Our results suggest a prominent role of PSTN Tac1 neurons projecting to the central amygdala in the hindrance of feeding initiation. While PSTN Crh neurons also delay the latency of hungry mice to ingest familiar foods, they surprisingly promote the consumption of novel, palatable substances. Furthermore, PSTN Crh neurons projecting to the bed nucleus of the stria terminalis accelerate rehydration in thirsty mice. Our results demonstrate the key role of endogenous PSTN activity in the control of feeding and drinking initiation and delineate specific circuits mediating these effects, which may have relevance for eating disorders.

17.
Cell ; 141(1): 154-165, 2010 Apr 02.
Article in English | MEDLINE | ID: mdl-20303157

ABSTRACT

Optogenetic technologies employ light to control biological processes within targeted cells in vivo with high temporal precision. Here, we show that application of molecular trafficking principles can expand the optogenetic repertoire along several long-sought dimensions. Subcellular and transcellular trafficking strategies now permit (1) optical regulation at the far-red/infrared border and extension of optogenetic control across the entire visible spectrum, (2) increased potency of optical inhibition without increased light power requirement (nanoampere-scale chloride-mediated photocurrents that maintain the light sensitivity and reversible, step-like kinetic stability of earlier tools), and (3) generalizable strategies for targeting cells based not only on genetic identity, but also on morphology and tissue topology, to allow versatile targeting when promoters are not known or in genetically intractable organisms. Together, these results illustrate use of cell-biological principles to enable expansion of the versatile fast optogenetic technologies suitable for intact-systems biology and behavior.


Subject(s)
Genetic Techniques , Light , Animals , Cells, Cultured , Hippocampus/cytology , Hippocampus/metabolism , Humans , Neurons/metabolism , Opsonin Proteins/genetics , Opsonin Proteins/metabolism , Rats , Systems Biology/methods
18.
Nature ; 565(7741): 645-649, 2019 01.
Article in English | MEDLINE | ID: mdl-30651638

ABSTRACT

Categorically distinct basic drives (for example, for social versus feeding behaviour1-3) can exert potent influences on each other; such interactions are likely to have important adaptive consequences (such as appropriate regulation of feeding in the context of social hierarchies) and can become maladaptive (such as in clinical settings involving anorexia). It is known that neural systems regulating natural and adaptive caloric intake, and those regulating social behaviours, involve related circuitry4-7, but the causal circuit mechanisms of these drive adjudications are not clear. Here we investigate the causal role in behaviour of cellular-resolution experience-specific neuronal populations in the orbitofrontal cortex, a major reward-processing hub that contains diverse activity-specific neuronal populations that respond differentially to various aspects of caloric intake8-13 and social stimuli14,15. We coupled genetically encoded activity imaging with the development and application of methods for optogenetic control of multiple individually defined cells, to both optically monitor and manipulate the activity of many orbitofrontal cortex neurons at the single-cell level in real time during rewarding experiences (caloric consumption and social interaction). We identified distinct populations within the orbitofrontal cortex that selectively responded to either caloric rewards or social stimuli, and found that activity of individually specified naturally feeding-responsive neurons was causally linked to increased feeding behaviour; this effect was selective as, by contrast, single-cell resolution activation of naturally social-responsive neurons inhibited feeding, and activation of neurons responsive to neither feeding nor social stimuli did not alter feeding behaviour. These results reveal the presence of potent cellular-level subnetworks within the orbitofrontal cortex that can be precisely engaged to bidirectionally control feeding behaviours subject to, for example, social influences.


Subject(s)
Feeding Behavior/physiology , Neural Pathways/physiology , Neurons/cytology , Neurons/physiology , Prefrontal Cortex/cytology , Prefrontal Cortex/physiology , Social Behavior , Animals , Conditioning, Operant/physiology , Energy Intake , Male , Mice , Mice, Inbred C57BL , Optogenetics , Reward , Single-Cell Analysis
19.
Nature ; 561(7723): 343-348, 2018 09.
Article in English | MEDLINE | ID: mdl-30158696

ABSTRACT

The naturally occurring channelrhodopsin variant anion channelrhodopsin-1 (ACR1), discovered in the cryptophyte algae Guillardia theta, exhibits large light-gated anion conductance and high anion selectivity when expressed in heterologous settings, properties that support its use as an optogenetic tool to inhibit neuronal firing with light. However, molecular insight into ACR1 is lacking owing to the absence of structural information underlying light-gated anion conductance. Here we present the crystal structure of G. theta ACR1 at 2.9 Å resolution. The structure reveals unusual architectural features that span the extracellular domain, retinal-binding pocket, Schiff-base region, and anion-conduction pathway. Together with electrophysiological and spectroscopic analyses, these findings reveal the fundamental molecular basis of naturally occurring light-gated anion conductance, and provide a framework for designing the next generation of optogenetic tools.


Subject(s)
Anions/metabolism , Channelrhodopsins/chemistry , Channelrhodopsins/metabolism , Cryptophyta/chemistry , Bacteriorhodopsins/chemistry , Binding Sites , Channelrhodopsins/radiation effects , Crystallography, X-Ray , Electric Conductivity , Ion Channel Gating/radiation effects , Ion Transport/radiation effects , Models, Molecular , Optogenetics/methods , Optogenetics/trends , Retinaldehyde/metabolism , Schiff Bases/chemistry
20.
Nature ; 561(7723): 349-354, 2018 09.
Article in English | MEDLINE | ID: mdl-30158697

ABSTRACT

Both designed and natural anion-conducting channelrhodopsins (dACRs and nACRs, respectively) have been widely applied in optogenetics (enabling selective inhibition of target-cell activity during animal behaviour studies), but each class exhibits performance limitations, underscoring trade-offs in channel structure-function relationships. Therefore, molecular and structural insights into dACRs and nACRs will be critical not only for understanding the fundamental mechanisms of these light-gated anion channels, but also to create next-generation optogenetic tools. Here we report crystal structures of the dACR iC++, along with spectroscopic, electrophysiological and computational analyses that provide unexpected insights into pH dependence, substrate recognition, channel gating and ion selectivity of both dACRs and nACRs. These results enabled us to create an anion-conducting channelrhodopsin integrating the key features of large photocurrent and fast kinetics alongside exclusive anion selectivity.


Subject(s)
Anions/metabolism , Channelrhodopsins/chemistry , Channelrhodopsins/metabolism , Ion Channel Gating , Optogenetics/methods , Animals , Caenorhabditis elegans , Cells, Cultured , Channelrhodopsins/genetics , Channelrhodopsins/radiation effects , Crystallography, X-Ray , Electrophysiology , Female , HEK293 Cells , Hippocampus/cytology , Humans , Hydrogen-Ion Concentration , Ion Channel Gating/radiation effects , Ion Transport/radiation effects , Kinetics , Male , Mice , Models, Molecular , Neurons/metabolism , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL