Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
1.
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
2.
Cell ; 166(3): 716-728, 2016 Jul 28.
Article in English | MEDLINE | ID: mdl-27426949

ABSTRACT

Fear behaviors are regulated by adaptive mechanisms that dampen their expression in the absence of danger. By studying circuits and the molecular mechanisms underlying this adaptive response, we show that cholinergic neurons of the medial habenula reduce fear memory expression through GABAB presynaptic excitation. Ablating these neurons or inactivating their GABAB receptors impairs fear extinction in mice, whereas activating the neurons or their axonal GABAB receptors reduces conditioned fear. Although considered exclusively inhibitory, here, GABAB mediates excitation by amplifying presynaptic Ca(2+) entry through Cav2.3 channels and potentiating co-release of glutamate, acetylcholine, and neurokinin B to excite interpeduncular neurons. Activating the receptors for these neurotransmitters or enhancing neurotransmission with a phosphodiesterase inhibitor reduces fear responses of both wild-type and GABAB mutant mice. We identify the role of an extra-amygdalar circuit and presynaptic GABAB receptors in fear control, suggesting that boosting neurotransmission in this pathway might ameliorate some fear disorders.


Subject(s)
Cholinergic Neurons/metabolism , Fear/physiology , Habenula/physiology , Memory/physiology , Receptors, GABA-B/metabolism , Animals , Calcium/metabolism , Calcium Channels/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Neural Pathways , Neurotransmitter Agents/metabolism , Synaptic Transmission
3.
Nature ; 629(8013): 893-900, 2024 May.
Article in English | MEDLINE | ID: mdl-38632402

ABSTRACT

The blood-brain barrier (BBB) protects the central nervous system from infections or harmful substances1; its impairment can lead to or exacerbate various diseases of the central nervous system2-4. However, the mechanisms of BBB disruption during infection and inflammatory conditions5,6 remain poorly defined. Here we find that activation of the pore-forming protein GSDMD by the cytosolic lipopolysaccharide (LPS) sensor caspase-11 (refs. 7-9), but not by TLR4-induced cytokines, mediates BBB breakdown in response to circulating LPS or during LPS-induced sepsis. Mice deficient in the LBP-CD14 LPS transfer and internalization pathway10-12 resist BBB disruption. Single-cell RNA-sequencing analysis reveals that brain endothelial cells (bECs), which express high levels of GSDMD, have a prominent response to circulating LPS. LPS acting on bECs primes Casp11 and Cd14 expression and induces GSDMD-mediated plasma membrane permeabilization and pyroptosis in vitro and in mice. Electron microscopy shows that this features ultrastructural changes in the disrupted BBB, including pyroptotic endothelia, abnormal appearance of tight junctions and vasculature detachment from the basement membrane. Comprehensive mouse genetic analyses, combined with a bEC-targeting adeno-associated virus system, establish that GSDMD activation in bECs underlies BBB disruption by LPS. Delivery of active GSDMD into bECs bypasses LPS stimulation and opens the BBB. In CASP4-humanized mice, Gram-negative Klebsiella pneumoniae infection disrupts the BBB; this is blocked by expression of a GSDMD-neutralizing nanobody in bECs. Our findings outline a mechanism for inflammatory BBB breakdown, and suggest potential therapies for diseases of the central nervous system associated with BBB impairment.


Subject(s)
Blood-Brain Barrier , Brain , Endothelial Cells , Gasdermins , Inflammation , Animals , Female , Humans , Male , Mice , Basement Membrane/metabolism , Basement Membrane/ultrastructure , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Blood-Brain Barrier/ultrastructure , Blood-Brain Barrier/virology , Brain/metabolism , Brain/pathology , Brain/ultrastructure , Caspases, Initiator/metabolism , Dependovirus , Endothelial Cells/metabolism , Endothelial Cells/ultrastructure , Gasdermins/antagonists & inhibitors , Gasdermins/metabolism , Inflammation/pathology , Inflammation/metabolism , Klebsiella pneumoniae/physiology , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/blood , Lipopolysaccharides/pharmacology , Mice, Inbred C57BL , Pyroptosis , Sepsis/metabolism , Sepsis/pathology , Sepsis/microbiology , Single-Cell Analysis , Tight Junctions/metabolism , Tight Junctions/ultrastructure
4.
Annu Rev Neurosci ; 43: 141-162, 2020 07 08.
Article in English | MEDLINE | ID: mdl-32640931

ABSTRACT

The brain serotonin systems participate in numerous aspects of reward processing, although it remains elusive how exactly serotonin signals regulate neural computation and reward-related behavior. The application of optogenetics and imaging techniques during the last decade has provided many insights. Here, we review recent progress on the organization and physiology of the dorsal raphe serotonin neurons and the relationships between their activity and behavioral functions in the context of reward processing. We also discuss several interesting theories on serotonin's function and how these theories may be reconciled by the possibility that serotonin, acting in synergy with coreleased glutamate, tracks and calculates the so-called beneficialness of the current state to guide an animal's behavior in dynamic environments.


Subject(s)
Brain/physiology , Neurons/physiology , Reward , Serotonin/metabolism , Animals , Dorsal Raphe Nucleus/physiology , Humans , Learning/physiology
5.
PLoS Biol ; 21(12): e3002403, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38109308

ABSTRACT

The pituitary represents an essential hub in the hypothalamus-pituitary-adrenal (HPA) axis. Pituitary hormone-producing cells (HPCs) release several hormones to regulate fundamental bodily functions under normal and stressful conditions. It is well established that the pituitary endocrine gland modulates the immune system by releasing adrenocorticotropic hormone (ACTH) in response to neuronal activation in the hypothalamus. However, it remains unclear how systemic inflammation regulates the transcriptomic profiles of pituitary HPCs. Here, we performed single-cell RNA-sequencing (scRNA-seq) of the mouse pituitary and revealed that upon inflammation, all major pituitary HPCs respond robustly in a cell type-specific manner, with corticotropes displaying the strongest reaction. Systemic inflammation also led to the production and release of noncanonical bioactive molecules, including Nptx2 by corticotropes, to modulate immune homeostasis. Meanwhile, HPCs up-regulated the gene expression of chemokines that facilitated the communication between the HPCs and immune cells. Together, our study reveals extensive interactions between the pituitary and immune system, suggesting multifaceted roles of the pituitary in mediating the effects of inflammation on many aspects of body physiology.


Subject(s)
Corticotropin-Releasing Hormone , Pituitary Gland , Mice , Animals , Corticotropin-Releasing Hormone/genetics , Pituitary Gland/metabolism , Adrenocorticotropic Hormone/genetics , Adrenocorticotropic Hormone/metabolism , Adrenocorticotropic Hormone/pharmacology , Inflammation/genetics , Gene Expression Profiling
6.
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
7.
Nat Methods ; 19(8): 976-985, 2022 08.
Article in English | MEDLINE | ID: mdl-35879607

ABSTRACT

As the resident immune cells in the central nervous system (CNS), microglia orchestrate immune responses and dynamically sculpt neural circuits in the CNS. Microglial dysfunction and mutations of microglia-specific genes have been implicated in many diseases of the CNS. Developing effective and safe vehicles for transgene delivery into microglia will facilitate the studies of microglia biology and microglia-associated disease mechanisms. Here, we report the discovery of adeno-associated virus (AAV) variants that mediate efficient in vitro and in vivo microglial transduction via directed evolution of the AAV capsid protein. These AAV-cMG and AAV-MG variants are capable of delivering various genetic payloads into microglia with high efficiency, and enable sufficient transgene expression to support fluorescent labeling, Ca2+ and neurotransmitter imaging and genome editing in microglia in vivo. Furthermore, single-cell RNA sequencing shows that the AAV-MG variants mediate in vivo transgene delivery without inducing microglia immune activation. These AAV variants should facilitate the use of various genetically encoded sensors and effectors in the study of microglia-related biology.


Subject(s)
Dependovirus , Microglia , Dependovirus/genetics , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors/genetics , Transduction, Genetic
8.
J Neurosci ; 42(9): 1648-1665, 2022 03 02.
Article in English | MEDLINE | ID: mdl-35017224

ABSTRACT

GABAB receptors in habenula cholinergic neurons mediate strong presynaptic excitation and control aversive memory expression. K+ channel tetramerization domain (KCTD) proteins are key interacting partners of GABAB receptors; it remains unclear whether and how KCTDs contribute to GABAB excitatory signaling. Here, we show that KCTD8 and KCTD12 in these neurons facilitate the GABAB receptors expression in axonal terminals and contribute to presynaptic excitation by GABAB receptors. Genetically knocking out KCTD8/12/16 or KCTD8/12, but not other combinations of the three KCTD isoforms, substantially reduced GABAB receptors-mediated potentiation of glutamate release and presynaptic Ca2+ entry in response to axonal stimulation, whereas they had no effect on GABAB-mediated inhibition in the somata of cholinergic neurons within the habenulo-interpeduncular pathway in mice of either sex. The physiological phenotypes were associated with a significant decrease in the GABAB expression within the axonal terminals but not the somata. Overexpressing either KCTD8 or KCTD12 in the KCTD8/12/16 triple knock-out mice reversed the changes in axonal GABAB expression and presynaptic excitation. In mice lacking the KCTDs, aversion-predicting cues produced stronger neuronal activation in the interpeduncular nucleus, and the infusion of GABAB agonist in this nucleus produced a weaker effect on fear extinction. Collectively, our results reveal isoform-specific roles of KCTD proteins in enriching the axonal expression of GABAB receptors, facilitating their presynaptic signaling, and modulating aversion-related memory processes.SIGNIFICANCE STATEMENT GABAB receptors represent the principal inhibitory neurotransmitter receptor, but they mediate strong presynaptic excitation in the habenulo-interpeduncular pathway and modulate aversion memory expression. KCTD proteins are integral constituents of GABAB receptors. By analyzing the physiological, neuroanatomical, and behavioral phenotypes of multiple KCTD knock-out mouse lines, we show that KCTD8 and KCTD12 facilitate the axonal expression and hence presynaptic excitation of GABAB receptors in habenula cholinergic neurons and control cued-aversion memory formation and expression in the habenulo-interpeduncular pathway. These results expand the physiological and behavioral functions of KCTDs in modulating the brain neural circuits.


Subject(s)
Axons , Cholinergic Neurons , Habenula , Intracellular Signaling Peptides and Proteins , Receptors, GABA-B , Receptors, GABA , Animals , Axons/metabolism , Cholinergic Neurons/metabolism , Extinction, Psychological , Fear/physiology , Habenula/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Knockout , Receptors, GABA/metabolism , Receptors, GABA-B/genetics , Receptors, GABA-B/metabolism , gamma-Aminobutyric Acid/metabolism
9.
Nanotechnology ; 34(32)2023 May 24.
Article in English | MEDLINE | ID: mdl-37141885

ABSTRACT

Transition metal carbides show remarkable catalysis for MgH2, and the addition of carbon materials can attach excellent cycling stability. In this paper, Mg-doped with transition metal carbides (TiC) and graphene (G) composite (denoted as Mg-TiC-G) is designed to assess the influence of TiC and graphene on the hydrogen storage performance of MgH2. The as-prepared Mg-TiC-G samples showed favorable dehydrogenation kinetics compared to the pristine Mg system. After adding TiC and graphene, the dehydrogenation activation energy of MgH2decreases from 128.4 to 111.2 kJ mol-1. The peak desorption temperature of MgH2doped with TiC and graphene is 326.5 °C, which is 26.3 °C lower than the pure Mg. The improved dehydrogenation performance of Mg-TiC-G composites is attributed to synergistic effects between catalysis and confinement.

10.
Acta Pharmacol Sin ; 43(9): 2276-2288, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35217811

ABSTRACT

Drug addiction is characterized by relapse when addicts are re-exposed to drug-associated environmental cues, but the neural mechanisms underlying cue-induced relapse are unclear. In the present study we investigated the role of a specific dopaminergic (DA) pathway from ventral tegmental area (VTA) to nucleus accumbens core (NAcore) in mouse cue-induced relapse. Optical intracranial self-stimulation (oICSS) was established in DAT-Cre transgenic mice. We showed that optogenetic excitation of DA neurons in the VTA or their projection terminals in NAcore, NAshell or infralimbic prefrontal cortex (PFC-IL) was rewarding. Furthermore, activation of the VTA-NAcore pathway alone was sufficient and necessary to induce reinstatement of oICSS. In cocaine self-administration model, cocaine-associated cues activated VTA DA neurons as assessed by intracellular GCaMP signals. Cue-induced reinstatement of cocaine-seeking was triggered by optogenetic stimulation of the VTA-NAcore pathway, and inhibited by chemogenetic inhibition of this pathway. Together, these results demonstrate that cue-induced reinstatement of reward seeking is in part mediated by activation of the VTA-NAcore DA pathway.


Subject(s)
Cocaine , Dopamine , Animals , Cocaine/pharmacology , Cues , Drug-Seeking Behavior , Mice , Mice, Transgenic , Nucleus Accumbens/physiology , Rats , Rats, Sprague-Dawley , Recurrence , Reward , Self Administration
11.
Nat Methods ; 15(4): 275-278, 2018 04.
Article in English | MEDLINE | ID: mdl-29481551

ABSTRACT

Immunosignal hybridization chain reaction (isHCR) combines antibody-antigen interactions with hybridization chain reaction (HCR) technology, which results in amplification of immunofluorescence signals by up to two to three orders of magnitude with low background. isHCR's highly modular and easily adaptable design enables the technique to be applied broadly, and we further optimized its use in multiplexed imaging and in state-of-the-art tissue expansion and clearing techniques.


Subject(s)
Nucleic Acid Amplification Techniques/methods , Nucleic Acid Hybridization/methods , Animals , Antibodies , Antigens , Behavior, Animal , Brain/metabolism , Gene Expression Regulation , HEK293 Cells , Humans , Immunohistochemistry , Mice , Protein Binding
12.
Nat Methods ; 15(12): 1033-1036, 2018 12.
Article in English | MEDLINE | ID: mdl-30455464

ABSTRACT

We developed a dual-adeno-associated-virus expression system that enables strong and sparse labeling of individual neurons with cell-type and projection specificity. We demonstrated its utility for whole-brain reconstruction of midbrain dopamine neurons and striatum-projecting cortical neurons. We further extended the labeling method for rapid reconstruction in cleared thick brain sections and simultaneous dual-color labeling. This labeling system may facilitate the process of generating mesoscale single-neuron projectomes of mammalian brains.


Subject(s)
Brain Mapping/methods , Brain/cytology , Cerebral Cortex/cytology , Dopaminergic Neurons/cytology , Neural Pathways , Animals , Brain/metabolism , Brain/virology , Cells, Cultured , Cerebral Cortex/metabolism , Dependovirus/genetics , Dopaminergic Neurons/metabolism , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Mice , Mice, Inbred C57BL
13.
Proc Natl Acad Sci U S A ; 115(26): 6632-6637, 2018 06 26.
Article in English | MEDLINE | ID: mdl-29891705

ABSTRACT

Optical upconversion that converts infrared light into visible light is of significant interest for broad applications in biomedicine, imaging, and displays. Conventional upconversion materials rely on nonlinear light-matter interactions, exhibit incidence-dependent efficiencies, and require high-power excitation. We report an infrared-to-visible upconversion strategy based on fully integrated microscale optoelectronic devices. These thin-film, ultraminiaturized devices realize near-infrared (∼810 nm) to visible [630 nm (red) or 590 nm (yellow)] upconversion that is linearly dependent on incoherent, low-power excitation, with a quantum yield of ∼1.5%. Additional features of this upconversion design include broadband absorption, wide-emission spectral tunability, and fast dynamics. Encapsulated, freestanding devices are transferred onto heterogeneous substrates and show desirable biocompatibilities within biological fluids and tissues. These microscale devices are implanted in behaving animals, with in vitro and in vivo experiments demonstrating their utility for optogenetic neuromodulation. This approach provides a versatile route to achieve upconversion throughout the entire visible spectral range at lower power and higher efficiency than has previously been possible.


Subject(s)
Miniaturization , Optogenetics/instrumentation , Prostheses and Implants , Animals , Arsenicals , Behavior, Animal , Biocompatible Materials , Brain Mapping/instrumentation , Equipment Design , Gallium , Infrared Rays , Mice , Mice, Nude , Optogenetics/methods , Photons , Rats , Semiconductors , Somatosensory Cortex/physiology , Subcutaneous Tissue
14.
Cereb Cortex ; 29(6): 2437-2454, 2019 06 01.
Article in English | MEDLINE | ID: mdl-29800098

ABSTRACT

Systematic examination of the inputs and outputs of the nonlemniscal auditory thalamus will facilitate the functional elucidation of this complex structure in the central auditory system. In mice, comprehensive tracing studies that reveal the long-range connectivity of the nonlemniscal auditory thalamus are lacking. To this end, we used Cre-inducible anterograde and monosynaptic retrograde viruses in Calbindin-2A-dgCre-D and Calretinin-IRES-Cre mice, focusing on the differences across subdivisions of the nonlemniscal auditory thalamus. We found that, 1) the dorsal and medial parts of the auditory thalamus were predominantly connected to sensory processing centers, whereas the posterior intralaminar (PIN) and peripeduncular nucleus (PP) were additionally connected to emotion and motivation modulation centers; 2) ventral auditory cortical areas were the major source of cortical inputs for all subdivisions, and the PIN/PP received more inputs from cortical layer 5 than other subdivisions did; 3) deep layers of the superior colliculus and rostral part of the nonlemniscal inferior colliculus preferentially projected to the PIN/PP; and 4) compared with the dorsal auditory thalamus, the PIN/PP mainly innervated association cortices. In addition, new brain areas connected to the nonlemniscal auditory thalamus, mostly the PIN/PP, were identified. Our results suggested subdivision-specific function of the nonlemniscal auditory thalamus in sound processing.


Subject(s)
Auditory Pathways/anatomy & histology , Thalamus/anatomy & histology , Animals , Female , Male , Mice , Mice, Inbred C57BL
15.
Proc Natl Acad Sci U S A ; 114(49): 13012-13017, 2017 12 05.
Article in English | MEDLINE | ID: mdl-29158387

ABSTRACT

Repeated exposure to drugs of abuse can produce adaptive changes that lead to the establishment of dependence. It has been shown that allelic variation in the α5 nicotinic acetylcholine receptor (nAChR) gene CHRNA5 is associated with higher risk of tobacco dependence. In the brain, α5-containing nAChRs are expressed at very high levels in the interpeduncular nucleus (IPN). Here we identified two nonoverlapping α5 + cell populations (α5- Amigo1 and α5- Epyc ) in mouse IPN that respond differentially to nicotine. Chronic nicotine treatment altered the translational profile of more than 1,000 genes in α5- Amigo1 neurons, including neuronal nitric oxide synthase (Nos1) and somatostatin (Sst). In contrast, expression of few genes was altered in the α5- Epyc population. We show that both nitric oxide and SST suppress optically evoked neurotransmitter release from the terminals of habenular (Hb) neurons in IPN. Moreover, in vivo silencing of neurotransmitter release from the α5- Amigo1 but not from the α5- Epyc population eliminates nicotine reward, measured using place preference. This loss of nicotine reward was mimicked by shRNA-mediated knockdown of Nos1 in the IPN. These findings reveal a proaddiction adaptive response to chronic nicotine in which nitric oxide and SST are released by a specific α5+ neuronal population to provide retrograde inhibition of the Hb-IPN circuit and thereby enhance the motivational properties of nicotine.


Subject(s)
Interpeduncular Nucleus/drug effects , Nicotine/pharmacology , Nitric Oxide Synthase Type I/genetics , Receptors, Nicotinic/genetics , Somatostatin/genetics , Tobacco Use Disorder/genetics , Animals , Female , Gene Expression Profiling , Gene Expression Regulation , Habenula/drug effects , Habenula/metabolism , Habenula/pathology , Interpeduncular Nucleus/metabolism , Interpeduncular Nucleus/pathology , Male , Mice , Mice, Transgenic , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neurotransmitter Agents/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type I/antagonists & inhibitors , Nitric Oxide Synthase Type I/metabolism , Protein Biosynthesis , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Nicotinic/metabolism , Reward , Somatostatin/metabolism , Stereotaxic Techniques , Synaptic Transmission , Tobacco Use Disorder/metabolism , Tobacco Use Disorder/pathology
16.
J Neurosci ; 37(37): 8863-8875, 2017 09 13.
Article in English | MEDLINE | ID: mdl-28821671

ABSTRACT

The ability to predict reward promotes animal survival. Both dopamine neurons in the ventral tegmental area and serotonin neurons in the dorsal raphe nucleus (DRN) participate in reward processing. Although the learning effects on dopamine neurons have been extensively characterized, it remains largely unknown how the response of serotonin neurons evolves during learning. Moreover, although stress is known to strongly influence reward-related behavior, we know very little about how stress modulates neuronal reward responses. By monitoring Ca2+ signals during the entire process of Pavlovian conditioning, we here show that learning differentially shapes the response patterns of serotonin neurons and dopamine neurons in mice of either sex. Serotonin neurons gradually develop a slow ramp-up response to the reward-predicting cue, and ultimately remain responsive to the reward, whereas dopamine neurons increase their response to the cue but reduce their response to the reward. For both neuron types, the responses to the cue and the reward depend on reward value, are reversible when the reward is omitted, and are rapidly reinstated by restoring the reward. We also found that stressors including head restraint and fearful context substantially reduce the response strength of both neuron types, to both the cue and the reward. These results reveal the dynamic nature of the reward responses, support the hypothesis that DRN serotonin neurons signal the current likelihood of receiving a net benefit, and suggest that the inhibitory effect of stress on the reward responses of serotonin neurons and dopamine neurons may contribute to stress-induced anhedonia.SIGNIFICANCE STATEMENT Both serotonin neurons in the dorsal raphe and dopamine neurons in the ventral tegmental area are intimately involved in reward processing. Using long-term fiber photometry of Ca2+ signals from freely behaving mice, we here show that learning produces a ramp-up activation pattern in serotonin neurons that differs from that in dopamine neurons, indicating complementary roles for these two neuron types in reward processing. Moreover, stress treatment substantially reduces the reward responses of both serotonin neurons and dopamine neurons, suggesting a possible physiological basis for stress-induced anhedonia.


Subject(s)
Action Potentials/physiology , Dopaminergic Neurons , Dorsal Raphe Nucleus/physiopathology , Reward , Serotonergic Neurons , Stress, Psychological/physiopathology , Ventral Tegmental Area/physiopathology , Animals , Calcium Signaling , Choice Behavior , Cues , Female , Male , Mice , Nerve Net/physiopathology , Neuronal Plasticity
17.
Cereb Cortex ; 27(6): 3110-3124, 2017 06 01.
Article in English | MEDLINE | ID: mdl-27252353

ABSTRACT

Local inhibition by γ-amino butyric acid (GABA)-containing neurons is of vital importance for the operation of sensory cortices. However, the physiological response patterns of cortical GABAergic neurons are poorly understood, especially in the awake condition. Here, we utilized the recently developed optical tagging technique to specifically record GABAergic neurons in the anterior piriform cortex (aPC) in awake mice. The identified aPC GABAergic neurons were stimulated with robotic delivery of 32 distinct odorants, which covered a broad range of functional groups. We found that aPC GABAergic neurons could be divided into 4 types based on their response patterns. Type I, type II, and type III neurons displayed broad excitatory responses to test odorants with different dynamics. Type I neurons were constantly activated during odorant stimulation, whereas type II neurons were only transiently activated at the onset of odorant delivery. In addition, type III neurons displayed transient excitatory responses both at the onset and termination of odorant presentation. Interestingly, type IV neurons were broadly inhibited by most of the odorants. Taken together, aPC GABAergic neurons adopt different strategies to affect the cortical circuitry. Our results will allow for better understanding of the role of cortical GABAergic interneurons in sensory information processing.


Subject(s)
GABAergic Neurons/physiology , Olfactory Perception/physiology , Piriform Cortex/cytology , Wakefulness/physiology , Action Potentials/genetics , Action Potentials/physiology , Animals , Channelrhodopsins/genetics , Channelrhodopsins/metabolism , Glutamate Decarboxylase/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Light , Mice , Mice, Transgenic , Neural Inhibition/physiology , Odorants , Optogenetics , Principal Component Analysis , Smell/physiology , Vesicular Inhibitory Amino Acid Transport Proteins/genetics , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism
18.
J Neurosci ; 36(20): 5472-88, 2016 05 18.
Article in English | MEDLINE | ID: mdl-27194328

ABSTRACT

UNLABELLED: Compelling evidence demonstrates that the external globus pallidus (GPe) plays a key role in processing sensorimotor information. An anatomical projection from the GPe to the dorsal striatum has been described for decades. However, the cellular target and functional impact of this projection remain unknown. Using cell-specific transgenic mice, modern monosynaptic tracing techniques, and optogenetics-based mapping, we discovered that GPe neurons provide inhibitory inputs to direct and indirect pathway striatal projection neurons (SPNs). Our results indicate that the GPe input to SPNs arises primarily from Npas1-expressing neurons and is strengthened in a chronic Parkinson's disease (PD) model. Alterations of the GPe-SPN input in a PD model argue for the critical position of this connection in regulating basal ganglia motor output and PD symptomatology. Finally, chemogenetic activation of Npas1-expressing GPe neurons suppresses motor output, arguing that strengthening of the GPe-SPN connection is maladaptive and may underlie the hypokinetic symptoms in PD. SIGNIFICANCE STATEMENT: An anatomical projection from the pallidum to the striatum has been described for decades, but little is known about its connectivity pattern. The authors dissect the presynaptic and postsynaptic neurons involved in this projection, and show its cell-specific remodeling and strengthening in parkinsonian mice. Chemogenetic activation of Npas1(+) pallidal neurons that give rise to the principal pallidostriatal projection increases the time that the mice spend motionless. This argues that maladaptive strengthening of this connection underlies the paucity of volitional movements, which is a hallmark of Parkinson's disease.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Globus Pallidus/physiology , Nerve Tissue Proteins/metabolism , Neurons/physiology , Synaptic Potentials , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Globus Pallidus/cytology , Globus Pallidus/metabolism , Mice , Mice, Inbred C57BL , Motor Activity , Nerve Tissue Proteins/genetics , Neurons/metabolism , Optogenetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Parkinson Disease/physiopathology
19.
J Neurosci ; 35(6): 2717-30, 2015 Feb 11.
Article in English | MEDLINE | ID: mdl-25673861

ABSTRACT

The orbitofrontal cortex (OFC) is important for the cognitive processes of learning and decision making. Previous recordings have revealed that OFC neurons encode predictions of reward outcomes. The OFC is interconnected with the dorsal raphe nucleus (DRN), which is a major serotonin (5-HT) center of the brain. Recent studies have provided increasing evidence that the DRN encodes reward signals. However, it remains unclear how the activity of DRN neurons affects the prospective reward coding of OFC neurons. By combining single-unit recordings from the OFC and optogenetic activation of the DRN in behaving mice, we found that DRN stimulation is sufficient to organize and modulate the anticipatory responses of OFC neurons. During pavlovian conditioning tasks for mice, odorant cues were associated with the delayed delivery of natural rewards of sucrose solution or DRN stimulation. After training, OFC neurons exhibited prospective responses to the sucrose solution. More importantly, the coupling of an odorant with delayed DRN stimulation resulted in tonic excitation or inhibition of OFC neurons during the delay period. The intensity of the prospective responses was affected by the frequency and duration of DRN stimulation. Additionally, DRN stimulation bidirectionally modulated the prospective responses to natural rewards. These experiments indicate that signals from the DRN are incorporated into the brain reward system to shape the cortical prospective coding of rewards.


Subject(s)
Frontal Lobe/physiology , Raphe Nuclei/physiology , Reward , Animals , Behavior, Animal/physiology , Conditioning, Classical/physiology , Discrimination Learning/physiology , Female , Male , Mice , Neurons/physiology , Optogenetics , Smell/physiology
20.
Neurobiol Learn Mem ; 135: 40-49, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27544850

ABSTRACT

The neurotransmitter serotonin (5-hydroxytryptamine; 5-HT) affects numerous behavioral and physiological processes. Drugs that alter 5-HT signaling treat several major psychiatric disorders and may lead to widespread abuse. The dorsal raphe nucleus (DRN) in the midbrain provides a majority of 5-HT for the forebrain. The importance of 5-HT signaling propels the search for a general theoretical framework under which the diverse functions of the DRN 5-HT neurons can be interpreted and additional therapeutic solutions may be developed. However, experimental data so far support several seeming irreconcilable theories, suggesting that 5-HT neurons mediate behavioral inhibition, aversive processing, or reward signaling. Here, we review recent progresses and propose that DRN 5-HT neurons encode "beneficialness" - how beneficial the current environmental context represents for an individual. Specifically, we speculate that the activity of these neurons reflects the possible net benefit of the current context as determined by p·R-C, in which p indicates reward probability, R the reward value, and C the cost. Through the widespread projections of these neurons to the forebrain, the beneficialness signal may reconfigure neural circuits to bias perception, boost positive emotions, and switch behavioral choices. The "beneficialness" hypothesis can explain many conflicting observations, and at the same time raises new questions. We suggest additional experiments that will help elucidate the exact computational functions of the DRN 5-HT neurons.


Subject(s)
Decision Making/physiology , Dorsal Raphe Nucleus/physiology , Emotions/physiology , Learning/physiology , Reward , Serotonergic Neurons/physiology , Serotonin/physiology , Social Behavior , Animals , Dorsal Raphe Nucleus/metabolism , Humans , Serotonergic Neurons/metabolism , Serotonin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL