Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 642
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 187(11): 2703-2716.e23, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38657602

ABSTRACT

Antigen presentation defects in tumors are prevalent mechanisms of adaptive immune evasion and resistance to cancer immunotherapy, whereas how tumors evade innate immunity is less clear. Using CRISPR screens, we discovered that IGSF8 expressed on tumors suppresses NK cell function by interacting with human KIR3DL2 and mouse Klra9 receptors on NK cells. IGSF8 is normally expressed in neuronal tissues and is not required for cell survival in vitro or in vivo. It is overexpressed and associated with low antigen presentation, low immune infiltration, and worse clinical outcomes in many tumors. An antibody that blocks IGSF8-NK receptor interaction enhances NK cell killing of malignant cells in vitro and upregulates antigen presentation, NK cell-mediated cytotoxicity, and T cell signaling in vivo. In syngeneic tumor models, anti-IGSF8 alone, or in combination with anti-PD1, inhibits tumor growth. Our results indicate that IGSF8 is an innate immune checkpoint that could be exploited as a therapeutic target.


Subject(s)
Immunity, Innate , Immunotherapy , Killer Cells, Natural , Neoplasms , Animals , Female , Humans , Mice , Antigen Presentation , Cell Line, Tumor , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Membrane Proteins/metabolism , Mice, Inbred C57BL , Neoplasms/immunology , Neoplasms/therapy
2.
Cell ; 186(18): 3845-3861.e24, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37591240

ABSTRACT

Dopaminergic projections regulate various brain functions and are implicated in many neuropsychiatric disorders. There are two anatomically and functionally distinct dopaminergic projections connecting the midbrain to striatum: nigrostriatal, which controls movement, and mesolimbic, which regulates motivation. However, how these discrete dopaminergic synaptic connections are established is unknown. Through an unbiased search, we identify that two groups of antagonistic TGF-ß family members, bone morphogenetic protein (BMP)6/BMP2 and transforming growth factor (TGF)-ß2, regulate dopaminergic synapse development of nigrostriatal and mesolimbic neurons, respectively. Projection-preferential expression of their receptors contributes to specific synapse development. Downstream, Smad1 and Smad2 are specifically activated and required for dopaminergic synapse development and function in nigrostriatal vs. mesolimbic projections. Remarkably, Smad1 mutant mice show motor defects, whereas Smad2 mutant mice show lack of motivation. These results uncover the molecular logic underlying the proper establishment of functionally segregated dopaminergic synapses and may provide strategies to treat relevant, projection-specific disease symptoms by targeting specific BMPs/TGF-ß and/or Smads.


Subject(s)
Corpus Striatum , Dopamine , Animals , Mice , Mesencephalon , Motivation , Movement , Synapses
3.
Cell ; 186(25): 5500-5516.e21, 2023 12 07.
Article in English | MEDLINE | ID: mdl-38016470

ABSTRACT

Most animals require sleep, and sleep loss induces serious pathophysiological consequences, including death. Previous experimental approaches for investigating sleep impacts in mice have been unable to persistently deprive animals of both rapid eye movement sleep (REMS) and non-rapid eye movement sleep (NREMS). Here, we report a "curling prevention by water" paradigm wherein mice remain awake 96% of the time. After 4 days of exposure, mice exhibit severe inflammation, and approximately 80% die. Sleep deprivation increases levels of prostaglandin D2 (PGD2) in the brain, and we found that elevated PGD2 efflux across the blood-brain-barrier-mediated by ATP-binding cassette subfamily C4 transporter-induces both accumulation of circulating neutrophils and a cytokine-storm-like syndrome. Experimental disruption of the PGD2/DP1 axis dramatically reduced sleep-deprivation-induced inflammation. Thus, our study reveals that sleep-related changes in PGD2 in the central nervous system drive profound pathological consequences in the peripheral immune system.


Subject(s)
Sleep Deprivation , Animals , Mice , Cytokines/metabolism , Inflammation , Prostaglandin D2 , Sleep/physiology , Sleep Deprivation/genetics , Sleep Deprivation/metabolism , Syndrome , Humans , Rats , Cell Line , Cyclonic Storms , Neutrophils/metabolism
4.
Cell ; 185(18): 3390-3407.e18, 2022 09 01.
Article in English | MEDLINE | ID: mdl-36055200

ABSTRACT

Chemical synapses between axons and dendrites mediate neuronal intercellular communication. Here, we describe a synapse between axons and primary cilia: the axo-ciliary synapse. Using enhanced focused ion beam-scanning electron microscopy on samples with optimally preserved ultrastructure, we discovered synapses between brainstem serotonergic axons and the primary cilia of hippocampal CA1 pyramidal neurons. Functionally, these cilia are enriched in a ciliary-restricted serotonin receptor, the 5-hydroxytryptamine receptor 6 (5-HTR6). Using a cilia-targeted serotonin sensor, we show that opto- and chemogenetic stimulation of serotonergic axons releases serotonin onto cilia. Ciliary 5-HTR6 stimulation activates a non-canonical Gαq/11-RhoA pathway, which modulates nuclear actin and increases histone acetylation and chromatin accessibility. Ablation of this pathway reduces chromatin accessibility in CA1 pyramidal neurons. As a signaling apparatus with proximity to the nucleus, axo-ciliary synapses short circuit neurotransmission to alter the postsynaptic neuron's epigenetic state.


Subject(s)
Axons/physiology , Chromatin/chemistry , Cilia , Synapses , Cell Nucleus/metabolism , Chromatin/metabolism , Cilia/metabolism , Hippocampus/cytology , Hippocampus/physiology , Serotonin/metabolism , Signal Transduction , Synapses/physiology
5.
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
6.
Cell ; 183(6): 1600-1616.e25, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33248024

ABSTRACT

Rapid phasic activity of midbrain dopamine neurons is thought to signal reward prediction errors (RPEs), resembling temporal difference errors used in machine learning. However, recent studies describing slowly increasing dopamine signals have instead proposed that they represent state values and arise independent from somatic spiking activity. Here we developed experimental paradigms using virtual reality that disambiguate RPEs from values. We examined dopamine circuit activity at various stages, including somatic spiking, calcium signals at somata and axons, and striatal dopamine concentrations. Our results demonstrate that ramping dopamine signals are consistent with RPEs rather than value, and this ramping is observed at all stages examined. Ramping dopamine signals can be driven by a dynamic stimulus that indicates a gradual approach to a reward. We provide a unified computational understanding of rapid phasic and slowly ramping dopamine signals: dopamine neurons perform a derivative-like computation over values on a moment-by-moment basis.


Subject(s)
Dopamine/metabolism , Signal Transduction , Action Potentials/physiology , Animals , Axons/metabolism , Calcium/metabolism , Calcium Signaling , Cell Body/metabolism , Cues , Dopaminergic Neurons/physiology , Fluorometry , Male , Mice, Inbred C57BL , Models, Neurological , Photic Stimulation , Reward , Sensation , Time Factors , Ventral Tegmental Area/metabolism , Virtual Reality
7.
Cell ; 178(1): 60-75.e19, 2019 06 27.
Article in English | MEDLINE | ID: mdl-31230716

ABSTRACT

Animals rely on the relative timing of events in their environment to form and update predictive associations, but the molecular and circuit mechanisms for this temporal sensitivity remain incompletely understood. Here, we show that olfactory associations in Drosophila can be written and reversed on a trial-by-trial basis depending on the temporal relationship between an odor cue and dopaminergic reinforcement. Through the synchronous recording of neural activity and behavior, we show that reversals in learned odor attraction correlate with bidirectional neural plasticity in the mushroom body, the associative olfactory center of the fly. Two dopamine receptors, DopR1 and DopR2, contribute to this temporal sensitivity by coupling to distinct second messengers and directing either synaptic depression or potentiation. Our results reveal how dopamine-receptor signaling pathways can detect the order of events to instruct opposing forms of synaptic and behavioral plasticity, allowing animals to flexibly update their associations in a dynamic environment.


Subject(s)
Association Learning/physiology , Drosophila Proteins/metabolism , Drosophila/physiology , Mushroom Bodies/physiology , Receptors, Dopamine D1/metabolism , Receptors, Dopamine/metabolism , Animals , Behavior, Animal/physiology , Conditioning, Classical/physiology , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Neuronal Plasticity , Odorants , Reward , Smell/physiology , Synaptic Potentials/physiology , Time Factors
8.
Cell ; 174(2): 481-496.e19, 2018 07 12.
Article in English | MEDLINE | ID: mdl-30007419

ABSTRACT

Dopamine (DA) is a central monoamine neurotransmitter involved in many physiological and pathological processes. A longstanding yet largely unmet goal is to measure DA changes reliably and specifically with high spatiotemporal precision, particularly in animals executing complex behaviors. Here, we report the development of genetically encoded GPCR-activation-based-DA (GRABDA) sensors that enable these measurements. In response to extracellular DA, GRABDA sensors exhibit large fluorescence increases (ΔF/F0 ∼90%) with subcellular resolution, subsecond kinetics, nanomolar to submicromolar affinities, and excellent molecular specificity. GRABDA sensors can resolve a single-electrical-stimulus-evoked DA release in mouse brain slices and detect endogenous DA release in living flies, fish, and mice. In freely behaving mice, GRABDA sensors readily report optogenetically elicited nigrostriatal DA release and depict dynamic mesoaccumbens DA signaling during Pavlovian conditioning or during sexual behaviors. Thus, GRABDA sensors enable spatiotemporally precise measurements of DA dynamics in a variety of model organisms while exhibiting complex behaviors.


Subject(s)
Dopamine/analysis , Drosophila/metabolism , Zebrafish/metabolism , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/metabolism , Behavior, Animal , Dopamine/metabolism , Female , Green Fluorescent Proteins/genetics , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Neurons/cytology , Neurons/metabolism , Optogenetics/methods , Receptors, G-Protein-Coupled/genetics , TRPV Cation Channels/genetics , Zebrafish Proteins/genetics
9.
Annu Rev Neurosci ; 45: 273-294, 2022 07 08.
Article in English | MEDLINE | ID: mdl-35316611

ABSTRACT

Recent advances in fluorescence imaging permit large-scale recording of neural activity and dynamics of neurochemical release with unprecedented resolution in behaving animals. Calcium imaging with highly optimized genetically encoded indicators provides a mesoscopic view of neural activity from genetically defined populations at cellular and subcellular resolutions. Rigorously improved voltage sensors and microscopy allow for robust spike imaging of populational neurons in various brain regions. In addition, recent protein engineering efforts in the past few years have led to the development of sensors for neurotransmitters and neuromodulators. Here, we discuss the development and applications of these genetically encoded fluorescent indicators in reporting neural activity in response to various behaviors in different biological systems as well as in drug discovery. We also report a simple model to guide sensor selection and optimization.


Subject(s)
Neurons , Receptors, Drug , Animals , Brain/metabolism , Neurons/physiology , Neurotransmitter Agents/metabolism , Optical Imaging , Receptors, Drug/metabolism
10.
Cell ; 161(2): 418-418.e1, 2015 Apr 09.
Article in English | MEDLINE | ID: mdl-25860614

ABSTRACT

The fundamental unit of DNA replication, the replicon, is governed by a cis-acting replicator sequence and a trans-activating initiator factor. Despite the increased size and complexity of eukaryotic genomes, eukaryotic DNA replication continues to be guided by the fundamental principles and concepts established in the replicon model.


Subject(s)
DNA Replication , Eukaryota/metabolism , Replication Origin , Animals , Bacteria/metabolism , DNA/metabolism , Epigenesis, Genetic , Histone Code , Humans
11.
Nature ; 621(7979): 543-549, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37558873

ABSTRACT

External rewards such as food and money are potent modifiers of behaviour1,2. Pioneering studies established that these salient sensory stimuli briefly interrupt the tonic discharge of neurons that produce the neuromodulators dopamine (DA) and acetylcholine (ACh): midbrain DA neurons (DANs) fire a burst of action potentials that broadly elevates DA in the striatum3,4 at the same time that striatal cholinergic interneurons (CINs) produce a characteristic pause in firing5,6. These phasic responses are thought to create unique, temporally limited conditions that motivate action and promote learning7-11. However, the dynamics of DA and ACh outside explicitly rewarded situations remain poorly understood. Here we show that extracellular DA and ACh levels fluctuate spontaneously and periodically at a frequency of approximately 2 Hz in the dorsal striatum of mice and maintain the same temporal relationship relative to one another as that evoked by reward. We show that this neuromodulatory coordination does not arise from direct interactions between DA and ACh within the striatum. Instead, we provide evidence that periodic fluctuations in striatal DA are inherited from midbrain DANs, while striatal ACh transients are driven by glutamatergic inputs, which act to locally synchronize the spiking of CINs. Together, our findings show that striatal neuromodulatory dynamics are autonomously organized by distributed extra-striatal afferents. The dominance of intrinsic rhythms in DA and ACh offers new insights for explaining how reward-associated neural dynamics emerge and how the brain motivates action and promotes learning from within.


Subject(s)
Acetylcholine , Corpus Striatum , Dopamine , Animals , Mice , Acetylcholine/metabolism , Action Potentials , Corpus Striatum/cytology , Corpus Striatum/metabolism , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Glutamine/metabolism , Interneurons/metabolism , Motivation , Neostriatum/cytology , Neostriatum/metabolism , Reward , Afferent Pathways
12.
Nature ; 606(7915): 732-738, 2022 06.
Article in English | MEDLINE | ID: mdl-35650441

ABSTRACT

Noradrenaline released from the locus coeruleus (LC) is a ubiquitous neuromodulator1-4 that has been linked to multiple functions including arousal5-8, action and sensory gain9-11, and learning12-16. Whether and how activation of noradrenaline-expressing neurons in the LC (LC-NA) facilitates different components of specific behaviours is unknown. Here we show that LC-NA activity displays distinct spatiotemporal dynamics to enable two functions during learned behaviour: facilitating task execution and encoding reinforcement to improve performance accuracy. To examine these functions, we used a behavioural task in mice with graded auditory stimulus detection and task performance. Optogenetic inactivation of the LC demonstrated that LC-NA activity was causal for both task execution and optimization. Targeted recordings of LC-NA neurons using photo-tagging, two-photon micro-endoscopy and two-photon output monitoring showed that transient LC-NA activation preceded behavioural execution and followed reinforcement. These two components of phasic activity were heterogeneously represented in LC-NA cortical outputs, such that the behavioural response signal was higher in the motor cortex and facilitated task execution, whereas the negative reinforcement signal was widely distributed among cortical regions and improved response sensitivity on the subsequent trial. Modular targeting of LC outputs thus enables diverse functions, whereby some noradrenaline signals are segregated among targets, whereas others are broadly distributed.


Subject(s)
Behavior, Animal , Learning , Locus Coeruleus , Norepinephrine , Animals , Learning/physiology , Locus Coeruleus/physiology , Mice , Neurons , Norepinephrine/metabolism , Optogenetics
13.
Nature ; 608(7923): 586-592, 2022 08.
Article in English | MEDLINE | ID: mdl-35859170

ABSTRACT

The ability to associate temporally segregated information and assign positive or negative valence to environmental cues is paramount for survival. Studies have shown that different projections from the basolateral amygdala (BLA) are potentiated following reward or punishment learning1-7. However, we do not yet understand how valence-specific information is routed to the BLA neurons with the appropriate downstream projections, nor do we understand how to reconcile the sub-second timescales of synaptic plasticity8-11 with the longer timescales separating the predictive cues from their outcomes. Here we demonstrate that neurotensin (NT)-expressing neurons in the paraventricular nucleus of the thalamus (PVT) projecting to the BLA (PVT-BLA:NT) mediate valence assignment by exerting NT concentration-dependent modulation in BLA during associative learning. We found that optogenetic activation of the PVT-BLA:NT projection promotes reward learning, whereas PVT-BLA projection-specific knockout of the NT gene (Nts) augments punishment learning. Using genetically encoded calcium and NT sensors, we further revealed that both calcium dynamics within the PVT-BLA:NT projection and NT concentrations in the BLA are enhanced after reward learning and reduced after punishment learning. Finally, we showed that CRISPR-mediated knockout of the Nts gene in the PVT-BLA pathway blunts BLA neural dynamics and attenuates the preference for active behavioural strategies to reward and punishment predictive cues. In sum, we have identified NT as a neuropeptide that signals valence in the BLA, and showed that NT is a critical neuromodulator that orchestrates positive and negative valence assignment in amygdala neurons by extending valence-specific plasticity to behaviourally relevant timescales.


Subject(s)
Basolateral Nuclear Complex , Learning , Neural Pathways , Neurotensin , Punishment , Reward , Basolateral Nuclear Complex/cytology , Basolateral Nuclear Complex/physiology , Calcium/metabolism , Cues , Neuronal Plasticity , Neurotensin/metabolism , Optogenetics , Thalamic Nuclei/cytology , Thalamic Nuclei/physiology
14.
Am J Hum Genet ; 111(3): 473-486, 2024 03 07.
Article in English | MEDLINE | ID: mdl-38354736

ABSTRACT

Disease-associated variants identified from genome-wide association studies (GWASs) frequently map to non-coding areas of the genome such as introns and intergenic regions. An exclusive reliance on gene-agnostic methods of genomic investigation could limit the identification of relevant genes associated with polygenic diseases such as Alzheimer disease (AD). To overcome such potential restriction, we developed a gene-constrained analytical method that considers only moderate- and high-risk variants that affect gene coding sequences. We report here the application of this approach to publicly available datasets containing 181,388 individuals without and with AD and the resulting identification of 660 genes potentially linked to the higher AD prevalence among Africans/African Americans. By integration with transcriptome analysis of 23 brain regions from 2,728 AD case-control samples, we concentrated on nine genes that potentially enhance the risk of AD: AACS, GNB5, GNS, HIPK3, MED13, SHC2, SLC22A5, VPS35, and ZNF398. GNB5, the fifth member of the heterotrimeric G protein beta family encoding Gß5, is primarily expressed in neurons and is essential for normal neuronal development in mouse brain. Homozygous or compound heterozygous loss of function of GNB5 in humans has previously been associated with a syndrome of developmental delay, cognitive impairment, and cardiac arrhythmia. In validation experiments, we confirmed that Gnb5 heterozygosity enhanced the formation of both amyloid plaques and neurofibrillary tangles in the brains of AD model mice. These results suggest that gene-constrained analysis can complement the power of GWASs in the identification of AD-associated genes and may be more broadly applicable to other polygenic diseases.


Subject(s)
Alzheimer Disease , GTP-Binding Protein beta Subunits , Mice , Humans , Animals , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Genome-Wide Association Study , Neurofibrillary Tangles/metabolism , Phenotype , Genomics , Amyloid beta-Peptides/genetics , Brain/metabolism , Solute Carrier Family 22 Member 5/genetics , Solute Carrier Family 22 Member 5/metabolism , GTP-Binding Protein beta Subunits/genetics , GTP-Binding Protein beta Subunits/metabolism
15.
Nat Methods ; 21(4): 692-702, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38443508

ABSTRACT

The serotonergic system plays important roles in both physiological and pathological processes, and is a therapeutic target for many psychiatric disorders. Although several genetically encoded GFP-based serotonin (5-HT) sensors were recently developed, their sensitivities and spectral profiles are relatively limited. To overcome these limitations, we optimized green fluorescent G-protein-coupled receptor (GPCR)-activation-based 5-HT (GRAB5-HT) sensors and developed a red fluorescent GRAB5-HT sensor. These sensors exhibit excellent cell surface trafficking and high specificity, sensitivity and spatiotemporal resolution, making them suitable for monitoring 5-HT dynamics in vivo. Besides recording subcortical 5-HT release in freely moving mice, we observed both uniform and gradient 5-HT release in the mouse dorsal cortex with mesoscopic imaging. Finally, we performed dual-color imaging and observed seizure-induced waves of 5-HT release throughout the cortex following calcium and endocannabinoid waves. In summary, these 5-HT sensors can offer valuable insights regarding the serotonergic system in both health and disease.


Subject(s)
Receptors, G-Protein-Coupled , Serotonin , Humans , Mice , Animals , Serotonin/metabolism , Receptors, G-Protein-Coupled/metabolism , Cerebral Cortex/metabolism
16.
Nat Methods ; 21(4): 680-691, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38036855

ABSTRACT

Dopamine (DA) plays multiple roles in a wide range of physiological and pathological processes via a large network of dopaminergic projections. To dissect the spatiotemporal dynamics of DA release in both dense and sparsely innervated brain regions, we developed a series of green and red fluorescent G-protein-coupled receptor activation-based DA (GRABDA) sensors using a variety of DA receptor subtypes. These sensors have high sensitivity, selectivity and signal-to-noise ratio with subsecond response kinetics and the ability to detect a wide range of DA concentrations. We then used these sensors in mice to measure both optogenetically evoked and behaviorally relevant DA release while measuring neurochemical signaling in the nucleus accumbens, amygdala and cortex. Using these sensors, we also detected spatially resolved heterogeneous cortical DA release in mice performing various behaviors. These next-generation GRABDA sensors provide a robust set of tools for imaging dopaminergic activity under a variety of physiological and pathological conditions.


Subject(s)
Dopamine , Nucleus Accumbens , Mice , Animals , Nucleus Accumbens/physiology , Receptors, Dopamine , Brain , Receptors, G-Protein-Coupled
17.
Nat Rev Neurosci ; 23(5): 257-274, 2022 05.
Article in English | MEDLINE | ID: mdl-35361961

ABSTRACT

Neurotransmitters and neuromodulators have a wide range of key roles throughout the nervous system. However, their dynamics in both health and disease have been challenging to assess, owing to the lack of in vivo tools to track them with high spatiotemporal resolution. Thus, developing a platform that enables minimally invasive, large-scale and long-term monitoring of neurotransmitters and neuromodulators with high sensitivity, high molecular specificity and high spatiotemporal resolution has been essential. Here, we review the methods available for monitoring the dynamics of neurotransmitters and neuromodulators. Following a brief summary of non-genetically encoded methods, we focus on recent developments in genetically encoded fluorescent indicators, highlighting how these novel indicators have facilitated advances in our understanding of the functional roles of neurotransmitters and neuromodulators in the nervous system. These studies present a promising outlook for the future development and use of tools to monitor neurotransmitters and neuromodulators.


Subject(s)
Neurotransmitter Agents , Humans
18.
Proc Natl Acad Sci U S A ; 120(21): e2301330120, 2023 05 23.
Article in English | MEDLINE | ID: mdl-37186824

ABSTRACT

The hypothalamic suprachiasmatic nucleus (SCN) is the master mammalian circadian clock. Its cell-autonomous timing mechanism, a transcriptional/translational feedback loop (TTFL), drives daily peaks of neuronal electrical activity, which in turn control circadian behavior. Intercellular signals, mediated by neuropeptides, synchronize and amplify TTFL and electrical rhythms across the circuit. SCN neurons are GABAergic, but the role of GABA in circuit-level timekeeping is unclear. How can a GABAergic circuit sustain circadian cycles of electrical activity, when such increased neuronal firing should become inhibitory to the network? To explore this paradox, we show that SCN slices expressing the GABA sensor iGABASnFR demonstrate a circadian oscillation of extracellular GABA ([GABA]e) that, counterintuitively, runs in antiphase to neuronal activity, with a prolonged peak in circadian night and a pronounced trough in circadian day. Resolving this unexpected relationship, we found that [GABA]e is regulated by GABA transporters (GATs), with uptake peaking during circadian day, hence the daytime trough and nighttime peak. This uptake is mediated by the astrocytically expressed transporter GAT3 (Slc6a11), expression of which is circadian-regulated, being elevated in daytime. Clearance of [GABA]e in circadian day facilitates neuronal firing and is necessary for circadian release of the neuropeptide vasoactive intestinal peptide, a critical regulator of TTFL and circuit-level rhythmicity. Finally, we show that genetic complementation of the astrocytic TTFL alone, in otherwise clockless SCN, is sufficient to drive [GABA]e rhythms and control network timekeeping. Thus, astrocytic clocks maintain the SCN circadian clockwork by temporally controlling GABAergic inhibition of SCN neurons.


Subject(s)
Circadian Clocks , Circadian Rhythm , Animals , Circadian Rhythm/genetics , Circadian Clocks/genetics , GABA Plasma Membrane Transport Proteins/metabolism , Suprachiasmatic Nucleus/metabolism , gamma-Aminobutyric Acid/metabolism , Mammals/metabolism
19.
Proc Natl Acad Sci U S A ; 120(14): e2216231120, 2023 04 04.
Article in English | MEDLINE | ID: mdl-36976764

ABSTRACT

Histamine is a conserved neuromodulator in mammalian brains and critically involved in many physiological functions. Understanding the precise structure of the histaminergic network is the cornerstone in elucidating its function. Herein, using histidine decarboxylase (HDC)-CreERT2 mice and genetic labeling strategies, we reconstructed a whole-brain three dimensional (3D) structure of histaminergic neurons and their outputs at 0.32 × 0.32 × 2 µm3 pixel resolution with a cutting-edge fluorescence microoptical sectioning tomography system. We quantified the fluorescence density of all brain areas and found that histaminergic fiber density varied significantly among brain regions. The density of histaminergic fiber was positively correlated with the amount of histamine release induced by optogenetic stimulation or physiological aversive stimulation. Lastly, we reconstructed a fine morphological structure of 60 histaminergic neurons via sparse labeling and uncovered the largely heterogeneous projection pattern of individual histaminergic neurons. Collectively, this study reveals an unprecedented whole-brain quantitative analysis of histaminergic projections at the mesoscopic level, providing a foundation for future functional histaminergic study.


Subject(s)
Brain , Histamine , Mice , Animals , Brain/metabolism , Neurons/metabolism , Brain Mapping , Histidine Decarboxylase/genetics , Histidine Decarboxylase/metabolism , Mammals/metabolism
20.
Proc Natl Acad Sci U S A ; 120(14): e2212387120, 2023 04 04.
Article in English | MEDLINE | ID: mdl-36996110

ABSTRACT

The purinergic signaling molecule adenosine (Ado) modulates many physiological and pathological functions in the brain. However, the exact source of extracellular Ado remains controversial. Here, utilizing a newly optimized genetically encoded GPCR-Activation-Based Ado fluorescent sensor (GRABAdo), we discovered that the neuronal activity-induced extracellular Ado elevation is due to direct Ado release from somatodendritic compartments of neurons, rather than from the axonal terminals, in the hippocampus. Pharmacological and genetic manipulations reveal that the Ado release depends on equilibrative nucleoside transporters but not the conventional vesicular release mechanisms. Compared with the fast-vesicular glutamate release, the Ado release is slow (~40 s) and requires calcium influx through L-type calcium channels. Thus, this study reveals an activity-dependent second-to-minute local Ado release from the somatodendritic compartments of neurons, potentially serving modulatory functions as a retrograde signal.


Subject(s)
Adenosine , Neurons , Adenosine/pharmacology , Nucleoside Transport Proteins/genetics , Signal Transduction/physiology , Guanine Nucleotide Exchange Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL