Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.589
Filter
1.
Cell ; 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39303715

ABSTRACT

Identifying the properties of the rapid eye movement (REM) sleep circuitry and its relation to diseases has been challenging due to the neuronal heterogeneity of the brainstem. Here, we show in mice that neurons in the pontine sublaterodorsal tegmentum (SubLDT) that express corticotropin-releasing hormone-binding protein (Crhbp+ neurons) and project to the medulla promote REM sleep. Within the medullary area receiving projections from Crhbp+ neurons, neurons expressing nitric oxide synthase 1 (Nos1+ neurons) project to the SubLDT and promote REM sleep, suggesting a positively interacting loop between the pons and the medulla operating as a core REM sleep circuit. Nos1+ neurons also project to areas that control wide forebrain activity. Ablating Crhbp+ neurons reduces sleep and impairs REM sleep atonia. In Parkinson's disease patients with REM sleep behavior disorders, CRHBP-immunoreactive neurons are largely reduced and contain pathologic α-synuclein, providing insight into the mechanisms underlying the sleep deficits characterizing this disease.

2.
Cell ; 186(3): 577-590.e16, 2023 02 02.
Article in English | MEDLINE | ID: mdl-36693373

ABSTRACT

Pleasurable touch is paramount during social behavior, including sexual encounters. However, the identity and precise role of sensory neurons that transduce sexual touch remain unknown. A population of sensory neurons labeled by developmental expression of the G protein-coupled receptor Mrgprb4 detects mechanical stimulation in mice. Here, we study the social relevance of Mrgprb4-lineage neurons and reveal that these neurons are required for sexual receptivity and sufficient to induce dopamine release in the brain. Even in social isolation, optogenetic stimulation of Mrgprb4-lineage neurons through the back skin is sufficient to induce a conditioned place preference and a striking dorsiflexion resembling the lordotic copulatory posture. In the absence of Mrgprb4-lineage neurons, female mice no longer find male mounts rewarding: sexual receptivity is supplanted by aggression and a coincident decline in dopamine release in the nucleus accumbens. Together, these findings establish that Mrgprb4-lineage neurons initiate a skin-to-brain circuit encoding the rewarding quality of social touch.


Subject(s)
Dopamine , Touch , Mice , Male , Female , Animals , Dopamine/metabolism , Nucleus Accumbens/metabolism , Sensory Receptor Cells/metabolism , Skin/metabolism , Reward , Dopaminergic Neurons/metabolism , Optogenetics , Receptors, G-Protein-Coupled/metabolism
3.
Cell ; 186(26): 5739-5750.e17, 2023 12 21.
Article in English | MEDLINE | ID: mdl-38070510

ABSTRACT

Conscious perception is greatly diminished during sleep, but the underlying circuit mechanism is poorly understood. We show that cortical ignition-a brain process shown to be associated with conscious awareness in humans and non-human primates-is strongly suppressed during non-rapid-eye-movement (NREM) sleep in mice due to reduced cholinergic modulation and rapid inhibition of cortical responses. Brain-wide functional ultrasound imaging and cell-type-specific calcium imaging combined with optogenetics showed that activity propagation from visual to frontal cortex is markedly reduced during NREM sleep due to strong inhibition of frontal pyramidal neurons. Chemogenetic activation and inactivation of basal forebrain cholinergic neurons powerfully increased and decreased visual-to-frontal activity propagation, respectively. Furthermore, although multiple subtypes of dendrite-targeting GABAergic interneurons in the frontal cortex are more active during wakefulness, soma-targeting parvalbumin-expressing interneurons are more active during sleep. Chemogenetic manipulation of parvalbumin interneurons showed that sleep/wake-dependent cortical ignition is strongly modulated by perisomatic inhibition of pyramidal neurons.


Subject(s)
Electroencephalography , Parvalbumins , Sleep , Animals , Mice , Cholinergic Neurons/physiology , Frontal Lobe/metabolism , Parvalbumins/metabolism , Sleep/physiology , Wakefulness/physiology
4.
Cell ; 186(7): 1369-1381.e17, 2023 03 30.
Article in English | MEDLINE | ID: mdl-37001501

ABSTRACT

Memories initially formed in hippocampus gradually stabilize to cortex over weeks-to-months for long-term storage. The mechanistic details of this brain re-organization remain poorly understood. We recorded bulk neural activity in circuits that link hippocampus and cortex as mice performed a memory-guided virtual-reality task over weeks. We identified a prominent and sustained neural correlate of memory in anterior thalamus, whose inhibition substantially disrupted memory consolidation. More strikingly, gain amplification enhanced consolidation of otherwise unconsolidated memories. To gain mechanistic insights, we developed a technology for simultaneous cellular-resolution imaging of hippocampus, thalamus, and cortex throughout consolidation. We found that whereas hippocampus equally encodes multiple memories, the anteromedial thalamus preferentially encodes salient memories, and gradually increases correlations with cortex to facilitate tuning and synchronization of cortical ensembles. We thus identify a thalamo-cortical circuit that gates memory consolidation and propose a mechanism suitable for the selection and stabilization of hippocampal memories into longer-term cortical storage.


Subject(s)
Memory Consolidation , Memory, Long-Term , Mice , Animals , Memory, Long-Term/physiology , Thalamus/physiology , Hippocampus/physiology , Memory Consolidation/physiology , Brain
5.
Cell ; 186(14): 3049-3061.e15, 2023 07 06.
Article in English | MEDLINE | ID: mdl-37311454

ABSTRACT

Membrane tension is thought to be a long-range integrator of cell physiology. Membrane tension has been proposed to enable cell polarity during migration through front-back coordination and long-range protrusion competition. These roles necessitate effective tension transmission across the cell. However, conflicting observations have left the field divided as to whether cell membranes support or resist tension propagation. This discrepancy likely originates from the use of exogenous forces that may not accurately mimic endogenous forces. We overcome this complication by leveraging optogenetics to directly control localized actin-based protrusions or actomyosin contractions while simultaneously monitoring the propagation of membrane tension using dual-trap optical tweezers. Surprisingly, actin-driven protrusions and actomyosin contractions both elicit rapid global membrane tension propagation, whereas forces applied to cell membranes alone do not. We present a simple unifying mechanical model in which mechanical forces that engage the actin cortex drive rapid, robust membrane tension propagation through long-range membrane flows.


Subject(s)
Actins , Actomyosin , Actins/metabolism , Actomyosin/metabolism , Actin Cytoskeleton/metabolism , Cell Membrane/metabolism , Cell Movement/physiology
6.
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
7.
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
8.
Cell ; 185(6): 1065-1081.e23, 2022 03 17.
Article in English | MEDLINE | ID: mdl-35245431

ABSTRACT

Motor behaviors are often planned long before execution but only released after specific sensory events. Planning and execution are each associated with distinct patterns of motor cortex activity. Key questions are how these dynamic activity patterns are generated and how they relate to behavior. Here, we investigate the multi-regional neural circuits that link an auditory "Go cue" and the transition from planning to execution of directional licking. Ascending glutamatergic neurons in the midbrain reticular and pedunculopontine nuclei show short latency and phasic changes in spike rate that are selective for the Go cue. This signal is transmitted via the thalamus to the motor cortex, where it triggers a rapid reorganization of motor cortex state from planning-related activity to a motor command, which in turn drives appropriate movement. Our studies show how midbrain can control cortical dynamics via the thalamus for rapid and precise motor behavior.


Subject(s)
Motor Cortex , Movement , Thalamus , Animals , Mesencephalon , Mice , Motor Cortex/physiology , Neurons/physiology , Thalamus/physiology
9.
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
10.
Annu Rev Biochem ; 90: 475-501, 2021 06 20.
Article in English | MEDLINE | ID: mdl-33781076

ABSTRACT

Optobiochemical control of protein activities allows the investigation of protein functions in living cells with high spatiotemporal resolution. Over the last two decades, numerous natural photosensory domains have been characterized and synthetic domains engineered and assembled into photoregulatory systems to control protein function with light. Here, we review the field of optobiochemistry, categorizing photosensory domains by chromophore, describing photoregulatory systems by mechanism of action, and discussing protein classes frequently investigated using optical methods. We also present examples of how spatial or temporal control of proteins in living cells has provided new insights not possible with traditional biochemical or cell biological techniques.


Subject(s)
Biochemistry/methods , Proteins/chemistry , Proteins/genetics , Proteins/metabolism , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cryptochromes/chemistry , Cryptochromes/metabolism , Flavin-Adenine Dinucleotide/chemistry , Flavin-Adenine Dinucleotide/metabolism , Light , Optogenetics/methods , Photochemical Processes , Photoreceptors, Microbial/chemistry , Photoreceptors, Microbial/metabolism , Phytochrome/chemistry , Phytochrome/metabolism , Protein Domains , Protein Engineering/methods , Vitamin B 12/metabolism
11.
Cell ; 184(2): 441-459.e25, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33333021

ABSTRACT

Barrier tissue immune responses are regulated in part by nociceptors. Nociceptor ablation alters local immune responses at peripheral sites and within draining lymph nodes (LNs). The mechanisms and significance of nociceptor-dependent modulation of LN function are unknown. Using high-resolution imaging, viral tracing, single-cell transcriptomics, and optogenetics, we identified and functionally tested a sensory neuro-immune circuit that is responsive to lymph-borne inflammatory signals. Transcriptomics profiling revealed that multiple sensory neuron subsets, predominantly peptidergic nociceptors, innervate LNs, distinct from those innervating surrounding skin. To uncover LN-resident cells that may interact with LN-innervating sensory neurons, we generated a LN single-cell transcriptomics atlas and nominated nociceptor target populations and interaction modalities. Optogenetic stimulation of LN-innervating sensory fibers triggered rapid transcriptional changes in the predicted interacting cell types, particularly endothelium, stromal cells, and innate leukocytes. Thus, a unique population of sensory neurons monitors peripheral LNs and may locally regulate gene expression.


Subject(s)
Immunomodulation , Lymph Nodes/immunology , Lymph Nodes/innervation , Sensory Receptor Cells/immunology , Action Potentials , Animals , Inflammation/pathology , Mice , Nociceptors/metabolism , Optogenetics , Peptides/metabolism , Skin/innervation , Sympathetic Nervous System/physiology , Toll-Like Receptors/agonists , Toll-Like Receptors/metabolism
12.
Cell ; 184(26): 6344-6360.e18, 2021 12 22.
Article in English | MEDLINE | ID: mdl-34890577

ABSTRACT

The anterior insular cortex (aIC) plays a critical role in cognitive and motivational control of behavior, but the underlying neural mechanism remains elusive. Here, we show that aIC neurons expressing Fezf2 (aICFezf2), which are the pyramidal tract neurons, signal motivational vigor and invigorate need-seeking behavior through projections to the brainstem nucleus tractus solitarii (NTS). aICFezf2 neurons and their postsynaptic NTS neurons acquire anticipatory activity through learning, which encodes the perceived value and the vigor of actions to pursue homeostatic needs. Correspondingly, aIC → NTS circuit activity controls vigor, effort, and striatal dopamine release but only if the action is learned and the outcome is needed. Notably, aICFezf2 neurons do not represent taste or valence. Moreover, aIC → NTS activity neither drives reinforcement nor influences total consumption. These results pinpoint specific functions of aIC → NTS circuit for selectively controlling motivational vigor and suggest that motivation is subserved, in part, by aIC's top-down regulation of dopamine signaling.


Subject(s)
Brain Stem/physiology , Insular Cortex/physiology , Motivation , Neural Pathways/physiology , Animals , Behavior, Animal , Dopamine/metabolism , Female , Learning , Male , Mice, Inbred C57BL , Neurons/physiology , Nucleus Accumbens/metabolism , Time Factors
13.
Cell ; 184(11): 2911-2926.e18, 2021 05 27.
Article in English | MEDLINE | ID: mdl-33932338

ABSTRACT

Hedgehog pathway components and select G protein-coupled receptors (GPCRs) localize to the primary cilium, an organelle specialized for signal transduction. We investigated whether cells distinguish between ciliary and extraciliary GPCR signaling. To test whether ciliary and extraciliary cyclic AMP (cAMP) convey different information, we engineered optogenetic and chemogenetic tools to control the subcellular site of cAMP generation. Generating equal amounts of ciliary and cytoplasmic cAMP in zebrafish and mammalian cells revealed that ciliary cAMP, but not cytoplasmic cAMP, inhibited Hedgehog signaling. Modeling suggested that the distinct geometries of the cilium and cell body differentially activate local effectors. The search for effectors identified a ciliary pool of protein kinase A (PKA). Blocking the function of ciliary PKA, but not extraciliary PKA, activated Hedgehog signal transduction and reversed the effects of ciliary cAMP. Therefore, cells distinguish ciliary and extraciliary cAMP using functionally and spatially distinct pools of PKA, and different subcellular pools of cAMP convey different information.


Subject(s)
Cilia/metabolism , Cyclic AMP/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Line , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytoplasm/metabolism , Hedgehog Proteins/antagonists & inhibitors , Hedgehog Proteins/metabolism , Neurons/metabolism , Optogenetics/methods , Signal Transduction/physiology , Zebrafish/metabolism
14.
Cell ; 180(4): 666-676.e13, 2020 02 20.
Article in English | MEDLINE | ID: mdl-32084339

ABSTRACT

The mystery of general anesthesia is that it specifically suppresses consciousness by disrupting feedback signaling in the brain, even when feedforward signaling and basic neuronal function are left relatively unchanged. The mechanism for such selectiveness is unknown. Here we show that three different anesthetics have the same disruptive influence on signaling along apical dendrites in cortical layer 5 pyramidal neurons in mice. We found that optogenetic depolarization of the distal apical dendrites caused robust spiking at the cell body under awake conditions that was blocked by anesthesia. Moreover, we found that blocking metabotropic glutamate and cholinergic receptors had the same effect on apical dendrite decoupling as anesthesia or inactivation of the higher-order thalamus. If feedback signaling occurs predominantly through apical dendrites, the cellular mechanism we found would explain not only how anesthesia selectively blocks this signaling but also why conscious perception depends on both cortico-cortical and thalamo-cortical connectivity.


Subject(s)
Anesthetics, General/pharmacology , Cerebral Cortex/drug effects , Pyramidal Cells/drug effects , Animals , Cerebral Cortex/cytology , Cerebral Cortex/physiology , Cholinergic Antagonists/pharmacology , Consciousness , Dendrites/drug effects , Dendrites/physiology , Excitatory Amino Acid Antagonists/pharmacology , Feedback, Physiological , Female , Male , Mice , Pyramidal Cells/physiology , Synaptic Transmission , Thalamus/cytology , Thalamus/drug effects , Thalamus/physiology
15.
Cell ; 183(7): 1913-1929.e26, 2020 12 23.
Article in English | MEDLINE | ID: mdl-33333020

ABSTRACT

Neurons in the cerebral cortex connect through descending pathways to hindbrain and spinal cord to activate muscle and generate movement. Although components of this pathway have been previously generated and studied in vitro, the assembly of this multi-synaptic circuit has not yet been achieved with human cells. Here, we derive organoids resembling the cerebral cortex or the hindbrain/spinal cord and assemble them with human skeletal muscle spheroids to generate 3D cortico-motor assembloids. Using rabies tracing, calcium imaging, and patch-clamp recordings, we show that corticofugal neurons project and connect with spinal spheroids, while spinal-derived motor neurons connect with muscle. Glutamate uncaging or optogenetic stimulation of cortical spheroids triggers robust contraction of 3D muscle, and assembloids are morphologically and functionally intact for up to 10 weeks post-fusion. Together, this system highlights the remarkable self-assembly capacity of 3D cultures to form functional circuits that could be used to understand development and disease.


Subject(s)
Cerebral Cortex/physiology , Motor Cortex/physiology , Organoids/physiology , Animals , Calcium/metabolism , Cell Differentiation , Cells, Cultured , Cervical Vertebrae , Gene Expression Regulation , Glutamates/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Mice , Muscles/physiology , Myoblasts/metabolism , Nerve Net/physiology , Optogenetics , Organoids/ultrastructure , Rhombencephalon/physiology , Spheroids, Cellular/cytology , Spinal Cord/cytology
16.
Cell ; 183(7): 2003-2019.e16, 2020 12 23.
Article in English | MEDLINE | ID: mdl-33308478

ABSTRACT

The ability to record transient cellular events in the DNA or RNA of cells would enable precise, large-scale analysis, selection, and reprogramming of heterogeneous cell populations. Here, we report a molecular technology for stable genetic tagging of cells that exhibit activity-related increases in intracellular calcium concentration (FLiCRE). We used FLiCRE to transcriptionally label activated neural ensembles in the nucleus accumbens of the mouse brain during brief stimulation of aversive inputs. Using single-cell RNA sequencing, we detected FLiCRE transcripts among the endogenous transcriptome, providing simultaneous readout of both cell-type and calcium activation history. We identified a cell type in the nucleus accumbens activated downstream of long-range excitatory projections. Taking advantage of FLiCRE's modular design, we expressed an optogenetic channel selectively in this cell type and showed that direct recruitment of this otherwise genetically inaccessible population elicits behavioral aversion. The specificity and minute resolution of FLiCRE enables molecularly informed characterization, manipulation, and reprogramming of activated cellular ensembles.


Subject(s)
Behavior, Animal , Calcium/metabolism , Corpus Striatum/metabolism , Animals , Female , HEK293 Cells , Humans , Kinetics , Male , Mice, Inbred C57BL , Neurons/metabolism , Optogenetics , Rats , Single-Cell Analysis , Transcriptome/genetics
17.
Cell ; 183(6): 1586-1599.e10, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33159859

ABSTRACT

The hippocampus is crucial for spatial navigation and episodic memory formation. Hippocampal place cells exhibit spatially selective activity within an environment and have been proposed to form the neural basis of a cognitive map of space that supports these mnemonic functions. However, the direct influence of place cell activity on spatial navigation behavior has not yet been demonstrated. Using an 'all-optical' combination of simultaneous two-photon calcium imaging and two-photon optogenetics, we identified and selectively activated place cells that encoded behaviorally relevant locations in a virtual reality environment. Targeted stimulation of a small number of place cells was sufficient to bias the behavior of animals during a spatial memory task, providing causal evidence that hippocampal place cells actively support spatial navigation and memory.


Subject(s)
Hippocampus/cytology , Place Cells/cytology , Spatial Behavior , Spatial Memory , Animals , Behavior, Animal , Male , Mice, Inbred C57BL , Neurons/metabolism , Opsins/metabolism , Optogenetics , Photons , Reward , Running , Spatial Navigation
18.
Cell ; 181(3): 574-589.e14, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32259485

ABSTRACT

Sensory neurons initiate defensive reflexes that ensure airway integrity. Dysfunction of laryngeal neurons is life-threatening, causing pulmonary aspiration, dysphagia, and choking, yet relevant sensory pathways remain poorly understood. Here, we discover rare throat-innervating neurons (∼100 neurons/mouse) that guard the airways against assault. We used genetic tools that broadly cover a vagal/glossopharyngeal sensory neuron atlas to map, ablate, and control specific afferent populations. Optogenetic activation of vagal P2RY1 neurons evokes a coordinated airway defense program-apnea, vocal fold adduction, swallowing, and expiratory reflexes. Ablation of vagal P2RY1 neurons eliminates protective responses to laryngeal water and acid challenge. Anatomical mapping revealed numerous laryngeal terminal types, with P2RY1 neurons forming corpuscular endings that appose laryngeal taste buds. Epithelial cells are primary airway sentinels that communicate with second-order P2RY1 neurons through ATP. These findings provide mechanistic insights into airway defense and a general molecular/genetic roadmap for internal organ sensation by the vagus nerve.


Subject(s)
Glossopharyngeal Nerve/physiology , Pharynx/innervation , Vagus Nerve/physiology , Afferent Pathways , Animals , Female , Gene Expression Regulation/genetics , Glossopharyngeal Nerve/metabolism , Larynx/pathology , Male , Mice , Mice, Inbred C57BL , Receptors, Purinergic P2Y1/genetics , Receptors, Purinergic P2Y1/metabolism , Sensory Receptor Cells/metabolism , Vagus Nerve/metabolism
19.
Cell ; 176(5): 1206-1221.e18, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30773317

ABSTRACT

Social behaviors, including behaviors directed toward young offspring, exhibit striking sex differences. Understanding how these sexually dimorphic behaviors are regulated at the level of circuits and transcriptomes will provide insights into neural mechanisms of sex-specific behaviors. Here, we uncover a sexually dimorphic role of the medial amygdala (MeA) in governing parental and infanticidal behaviors. Contrary to traditional views, activation of GABAergic neurons in the MeA promotes parental behavior in females, while activation of this population in males differentially promotes parental versus infanticidal behavior in an activity-level-dependent manner. Through single-cell transcriptomic analysis, we found that molecular sex differences in the MeA are specifically represented in GABAergic neurons. Collectively, these results establish crucial roles for the MeA as a key node in the neural circuitry underlying pup-directed behaviors and provide important insight into the connection between sex differences across transcriptomes, cells, and circuits in regulating sexually dimorphic behavior.


Subject(s)
Corticomedial Nuclear Complex/physiology , Sex Characteristics , Sexual Behavior, Animal/physiology , Amygdala/physiology , Animals , Behavior, Animal/physiology , Corticomedial Nuclear Complex/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Neurons/physiology , Parenting , Sex Factors , Social Behavior
20.
Cell ; 179(5): 1129-1143.e23, 2019 11 14.
Article in English | MEDLINE | ID: mdl-31730854

ABSTRACT

Energy homeostasis requires precise measurement of the quantity and quality of ingested food. The vagus nerve innervates the gut and can detect diverse interoceptive cues, but the identity of the key sensory neurons and corresponding signals that regulate food intake remains unknown. Here, we use an approach for target-specific, single-cell RNA sequencing to generate a map of the vagal cell types that innervate the gastrointestinal tract. We show that unique molecular markers identify vagal neurons with distinct innervation patterns, sensory endings, and function. Surprisingly, we find that food intake is most sensitive to stimulation of mechanoreceptors in the intestine, whereas nutrient-activated mucosal afferents have no effect. Peripheral manipulations combined with central recordings reveal that intestinal mechanoreceptors, but not other cell types, potently and durably inhibit hunger-promoting AgRP neurons in the hypothalamus. These findings identify a key role for intestinal mechanoreceptors in the regulation of feeding.


Subject(s)
Feeding Behavior/physiology , Genetic Phenomena , Sensory Receptor Cells/physiology , Vagus Nerve/physiology , Agouti-Related Protein/metabolism , Animals , Brain/physiology , Gastrointestinal Tract/innervation , Genetic Markers , Mechanoreceptors/metabolism , Mice , Vagus Nerve/anatomy & histology , Viscera/innervation
SELECTION OF CITATIONS
SEARCH DETAIL