Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 849
1.
Nat Neurosci ; 27(5): 927-939, 2024 May.
Article En | MEDLINE | ID: mdl-38570661

An essential feature of neurons is their ability to centrally integrate information from their dendrites. The activity of astrocytes, in contrast, has been described as mostly uncoordinated across cellular compartments without clear central integration. Here we report conditional integration of calcium signals in astrocytic distal processes at their soma. In the hippocampus of adult mice of both sexes, we found that global astrocytic activity, as recorded with population calcium imaging, reflected past neuronal and behavioral events on a timescale of seconds. Salient past events, indicated by pupil dilations, facilitated the propagation of calcium signals from distal processes to the soma. Centripetal propagation to the soma was reproduced by optogenetic activation of the locus coeruleus, a key regulator of arousal, and reduced by pharmacological inhibition of α1-adrenergic receptors. Together, our results suggest that astrocytes are computational units of the brain that slowly and conditionally integrate calcium signals upon behaviorally relevant events.


Astrocytes , Calcium Signaling , Hippocampus , Locus Coeruleus , Animals , Locus Coeruleus/physiology , Locus Coeruleus/cytology , Astrocytes/physiology , Mice , Hippocampus/physiology , Hippocampus/cytology , Male , Calcium Signaling/physiology , Female , Optogenetics , Mice, Transgenic , Neurons/physiology , Mice, Inbred C57BL , Calcium/metabolism
2.
Commun Biol ; 6(1): 414, 2023 04 14.
Article En | MEDLINE | ID: mdl-37059729

Light exposure can profoundly affect neurological functions and behaviors. Here, we show that short-term exposure to moderate (400 lux) white light during Y-maze test promoted spatial memory retrieval and induced only mild anxiety in mice. This beneficial effect involves the activation of a circuit including neurons in the central amygdala (CeA), locus coeruleus (LC), and dentate gyrus (DG). Specifically, moderate light activated corticotropin-releasing hormone (CRH) positive (+) CeA neurons and induced the release of corticotropin-releasing factor (CRF) from their axon terminals ending in the LC. CRF then activated tyrosine hydroxylase-expressing LC neurons, which send projections to DG and release norepinephrine (NE). NE activated ß-adrenergic receptors on CaMKIIα-expressing DG neurons, ultimately promoting spatial memory retrieval. Our study thus demonstrated a specific light scheme that can promote spatial memory without excessive stress, and unraveled the underlying CeA-LC-DG circuit and associated neurochemical mechanisms.


Amygdala , Light , Spatial Memory , Amygdala/cytology , Amygdala/metabolism , Animals , Mice , Anxiety , Dentate Gyrus/cytology , Dentate Gyrus/metabolism , Neurons , Locus Coeruleus/cytology , Locus Coeruleus/metabolism , Corticotropin-Releasing Hormone/metabolism , Norepinephrine/metabolism , Neural Pathways , Maze Learning , Mice, Inbred C57BL
3.
J Neurosci ; 43(13): 2338-2348, 2023 03 29.
Article En | MEDLINE | ID: mdl-36849414

Photoaffinity ligands are best known as tools used to identify the specific binding sites of drugs to their molecular targets. However, photoaffinity ligands have the potential to further define critical neuroanatomic targets of drug action. In the brains of WT male mice, we demonstrate the feasibility of using photoaffinity ligands in vivo to prolong anesthesia via targeted yet spatially restricted photoadduction of azi-m-propofol (aziPm), a photoreactive analog of the general anesthetic propofol. Systemic administration of aziPm with bilateral near-ultraviolet photoadduction in the rostral pons, at the border of the parabrachial nucleus and locus coeruleus, produced a 20-fold increase in the duration of sedative and hypnotic effects compared with control mice without UV illumination. Photoadduction that missed the parabrachial-coerulean complex also failed to extend the sedative or hypnotic actions of aziPm and was indistinguishable from nonadducted controls. Paralleling the prolonged behavioral and EEG consequences of on target in vivo photoadduction, we conducted electrophysiologic recordings in rostral pontine brain slices. Using neurons within the locus coeruleus to further highlight the cellular consequences of irreversible aziPm binding, we demonstrate transient slowing of spontaneous action potentials with a brief bath application of aziPm that becomes irreversible on photoadduction. Together, these findings suggest that photochemistry-based strategies are a viable new approach for probing CNS physiology and pathophysiology.SIGNIFICANCE STATEMENT Photoaffinity ligands are drugs capable of light-induced irreversible binding, which have unexploited potential to identify the neuroanatomic sites of drug action. We systemically administer a centrally acting anesthetic photoaffinity ligand in mice, conduct localized photoillumination within the brain to covalently adduct the drug at its in vivo sites of action, and successfully enrich irreversible drug binding within a restricted 250 µm radius. When photoadduction encompassed the pontine parabrachial-coerulean complex, anesthetic sedation and hypnosis was prolonged 20-fold, thus illustrating the power of in vivo photochemistry to help unravel neuronal mechanisms of drug action.


Anesthetics, Intravenous , Brain , Hypnosis , Hypnotics and Sedatives , Ligands , Photoaffinity Labels , Propofol , Animals , Male , Mice , Adrenergic Neurons/drug effects , Anesthesia, Intravenous , Brain/cytology , Brain/drug effects , Brain/metabolism , Brain/radiation effects , Electrocorticography , Electroencephalography , Hypnosis/methods , Hypnotics and Sedatives/administration & dosage , Hypnotics and Sedatives/chemistry , Hypnotics and Sedatives/pharmacology , Hypnotics and Sedatives/radiation effects , Locus Coeruleus/cytology , Locus Coeruleus/drug effects , Locus Coeruleus/metabolism , Locus Coeruleus/radiation effects , Mice, Inbred C57BL , Parabrachial Nucleus/drug effects , Parabrachial Nucleus/metabolism , Parabrachial Nucleus/radiation effects , Photoaffinity Labels/chemistry , Photoaffinity Labels/radiation effects , Propofol/administration & dosage , Propofol/analogs & derivatives , Propofol/pharmacology , Propofol/radiation effects , Time Factors , Ultraviolet Rays , Anesthetics, Intravenous/administration & dosage , Anesthetics, Intravenous/chemistry , Anesthetics, Intravenous/pharmacology , Anesthetics, Intravenous/radiation effects
4.
Nature ; 613(7943): 317-323, 2023 01.
Article En | MEDLINE | ID: mdl-36544024

Cochlear implants (CIs) are neuroprosthetic devices that can provide hearing to deaf people1. Despite the benefits offered by CIs, the time taken for hearing to be restored and perceptual accuracy after long-term CI use remain highly variable2,3. CI use is believed to require neuroplasticity in the central auditory system, and differential engagement of neuroplastic mechanisms might contribute to the variability in outcomes4-7. Despite extensive studies on how CIs activate the auditory system4,8-12, the understanding of CI-related neuroplasticity remains limited. One potent factor enabling plasticity is the neuromodulator noradrenaline from the brainstem locus coeruleus (LC). Here we examine behavioural responses and neural activity in LC and auditory cortex of deafened rats fitted with multi-channel CIs. The rats were trained on a reward-based auditory task, and showed considerable individual differences of learning rates and maximum performance. LC photometry predicted when CI subjects began responding to sounds and longer-term perceptual accuracy. Optogenetic LC stimulation produced faster learning and higher long-term accuracy. Auditory cortical responses to CI stimulation reflected behavioural performance, with enhanced responses to rewarded stimuli and decreased distinction between unrewarded stimuli. Adequate engagement of central neuromodulatory systems is thus a potential clinically relevant target for optimizing neuroprosthetic device use.


Cochlear Implants , Deafness , Locus Coeruleus , Animals , Rats , Cochlear Implantation , Deafness/physiopathology , Deafness/therapy , Hearing/physiology , Learning/physiology , Locus Coeruleus/cytology , Locus Coeruleus/physiology , Neuronal Plasticity , Norepinephrine/metabolism , Auditory Cortex/cytology , Auditory Cortex/physiology , Auditory Cortex/physiopathology , Neurons/physiology , Reward , Optogenetics , Photometry
5.
Ann Neurol ; 91(2): 282-288, 2022 02.
Article En | MEDLINE | ID: mdl-34981555

Narcolepsy type 1 (NT1) is a chronic sleep disorder correlated with loss of hypocretin(orexin). In NT1 post-mortem brains, we observed 88% reduction in corticotropin-releasing hormone (CRH)-positive neurons in the paraventricular nucleus (PVN) and significantly less CRH-positive fibers in the median eminence, whereas CRH-neurons in the locus coeruleus and thalamus, and other PVN neuronal populations were spared: that is, vasopressin, oxytocin, tyrosine hydroxylase, and thyrotropin releasing hormone-expressing neurons. Other hypothalamic cell groups, that is, the suprachiasmatic, ventrolateral preoptic, infundibular, and supraoptic nuclei and nucleus basalis of Meynert, were unaffected. The surprising selective decrease in CRH-neurons provide novel targets for diagnostics and therapeutic interventions. ANN NEUROL 2022;91:282-288.


Corticotropin-Releasing Hormone/metabolism , Hypothalamus/metabolism , Hypothalamus/pathology , Narcolepsy/pathology , Neurons/pathology , Aged , Aged, 80 and over , Cell Count , Female , Humans , Hypothalamus/diagnostic imaging , Immunohistochemistry , Locus Coeruleus/cytology , Locus Coeruleus/diagnostic imaging , Locus Coeruleus/metabolism , Male , Median Eminence/cytology , Median Eminence/diagnostic imaging , Median Eminence/metabolism , Middle Aged , Narcolepsy/diagnostic imaging , Paraventricular Hypothalamic Nucleus/cytology , Paraventricular Hypothalamic Nucleus/diagnostic imaging , Paraventricular Hypothalamic Nucleus/metabolism
6.
J Neurosci ; 41(15): 3512-3530, 2021 04 14.
Article En | MEDLINE | ID: mdl-33536201

The cerebellum processes neural signals related to rewarding and aversive stimuli, suggesting that the cerebellum supports nonmotor functions in cognitive and emotional domains. Catecholamines are a class of neuromodulatory neurotransmitters well known for encoding such salient stimuli. Catecholaminergic modulation of classical cerebellar functions have been demonstrated. However, a role for cerebellar catecholamines in modulating cerebellar nonmotor functions is unknown. Using biochemical methods in male mice, we comprehensively mapped TH+ fibers throughout the entire cerebellum and known precerebellar nuclei. Using electrochemical (fast scan cyclic voltammetry), and viral/genetic methods to selectively delete Th in fibers innervating the lateral cerebellar nucleus (LCN), we interrogated sources and functional roles of catecholamines innervating the LCN, which is known for its role in supporting cognition. The LCN has the most TH+ fibers in cerebellum, as well as the most change in rostrocaudal expression among the cerebellar nuclei. Norepinephrine is the major catecholamine measured in LCN. Distinct catecholaminergic projections to LCN arise only from locus coeruleus, and a subset of Purkinje cells that are positive for staining of TH. LC stimulation was sufficient to produce catecholamine release in LCN. Deletion of Th in fibers innervating LCN (LCN-Th-cKO) resulted in impaired sensorimotor integration, associative fear learning, response inhibition, and working memory in LCN-Th-cKO mice. Strikingly, selective inhibition of excitatory LCN output neurons with inhibitory designer receptor exclusively activated by designer drugs led to facilitation of learning on the same working memory task impaired in LCN-Th-cKO mice. Collectively, these data demonstrate a role for LCN catecholamines in cognitive behaviors.SIGNIFICANCE STATEMENT Here, we report on interrogating sources and functional roles of catecholamines innervating the lateral nucleus of the cerebellum (LCN). We map and quantify expression of TH, the rate-limiting enzyme in catecholamine synthesis, in the entire cerebellar system, including several precerebellar nuclei. We used cyclic voltammetry and pharmacology to demonstrate sufficiency of LC stimulation to produce catecholamine release in LCN. We used advanced viral techniques to map and selectively KO catecholaminergic neurotransmission to the LCN, and characterized significant cognitive deficits related to this manipulation. Finally, we show that inhibition of excitatory LCN neurons with designer receptor exclusively activated by designer drugs, designed to mimic Gi-coupled catecholamine GPCR signaling, results in facilitation of a working memory task impaired in LCN-specific TH KO mice.


Cerebellar Nuclei/physiology , Cognition , Norepinephrine/metabolism , Animals , Cerebellar Nuclei/cytology , Cerebellar Nuclei/metabolism , Fear , Locus Coeruleus/cytology , Locus Coeruleus/metabolism , Locus Coeruleus/physiology , Male , Memory, Short-Term , Mice , Neural Pathways/cytology , Neural Pathways/metabolism , Neural Pathways/physiology , Neurons/metabolism , Neurons/physiology , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
7.
Int J Mol Sci ; 22(1)2020 Dec 22.
Article En | MEDLINE | ID: mdl-33374940

Our laboratory and others have previously shown that cannabinoid receptor type-1 (CB1r) activity is neuroprotective and a modulator of brain ageing; a genetic disruption of CB1r signaling accelerates brain ageing, whereas the pharmacological stimulation of CB1r activity had the opposite effect. In this study, we have investigated if the lack of CB1r affects noradrenergic neurons in the locus coeruleus (LC), which are vulnerable to age-related changes; their numbers are reduced in patients with neurodegenerative diseases and probably also in healthy aged individuals. Thus, we compared LC neuronal numbers between cannabinoid 1 receptor knockout (Cnr1-/-) mice and their wild-type littermates. Our results reveal that old Cnr1-/- mice have less noradrenergic neurons compared to their age-matched wild-type controls. This result was also confirmed by the analysis of the density of noradrenergic terminals which proved that Cnr1-/- mice had less compared to the wild-type controls. Additionally, we assessed pro-inflammatory glial activity in the LC. Although the density of microglia in Cnr1-/- mice was enhanced, they did not show enhanced inflammatory profile. We hypothesize that CB1r activity is necessary for the protection of noradrenergic neurons, but its anti-inflammatory effect probably only plays a minor role in it.


Adrenergic Neurons/metabolism , Aging , Locus Coeruleus/metabolism , Neurodegenerative Diseases/metabolism , Receptor, Cannabinoid, CB1/metabolism , Adrenergic Neurons/cytology , Age Factors , Animals , Humans , Locus Coeruleus/cytology , Male , Mice, Inbred C57BL , Mice, Knockout , Microglia/cytology , Microglia/metabolism , Neurodegenerative Diseases/genetics , Norepinephrine/metabolism , Receptor, Cannabinoid, CB1/genetics , Tyrosine 3-Monooxygenase/metabolism
8.
Front Neural Circuits ; 14: 53, 2020.
Article En | MEDLINE | ID: mdl-33071759

As the most important organ in our bodies, the brain plays a critical role in deciding sex-related differential features; however, the underlying neural circuitry basis remains unclear. Here, we used a cell-type-specific rabies virus-mediated monosynaptic tracing system to generate a sex differences-related whole-brain input atlas of locus coeruleus noradrenaline (LC-NE) neurons. We developed custom pipelines for brain-wide comparisons of input sources in both sexes with the registration of the whole-brain data set to the Allen Mouse Brain Reference Atlas. Among 257 distinct anatomical regions, we demonstrated the differential proportions of inputs to LC-NE neurons in male and female mice at different levels. Locus coeruleus noradrenaline neurons of two sexes showed general similarity in the input patterns, but with differentiated input proportions quantitatively from major brain regions and diverse sub-regions. For instance, inputs to male LC-NE neurons were found mainly in the cerebrum, interbrain, and cerebellum, whereas inputs to female LC-NE neurons were found in the midbrain and hindbrain. We further found that specific subsets of nuclei nested within sub-regions contributed to overall sex-related differences in the input circuitry. Furthermore, among the totaled 123 anatomical regions with proportion of inputs >0.1%, we also identified 11 sub-regions with significant statistical differences of total inputs between male and female mice, and seven of them also showed such differences in ipsilateral hemispheres. Our study not only provides a structural basis to facilitate our understanding of sex differences at a circuitry level but also provides clues for future sexually differentiated functional studies related to LC-NE neurons.


Adrenergic Neurons/cytology , Locus Coeruleus/cytology , Neural Pathways/cytology , Animals , Atlases as Topic , Brain , Female , Male , Mice , Neuroanatomical Tract-Tracing Techniques , Rabies virus , Sex Characteristics
9.
J Neurosci ; 40(43): 8367-8385, 2020 10 21.
Article En | MEDLINE | ID: mdl-32994339

The ability of animals to retrieve memories stored in response to the environment is essential for behavioral adaptation. Norepinephrine (NE)-containing neurons in the brain play a key role in the modulation of synaptic plasticity underlying various processes of memory formation. However, the role of the central NE system in memory retrieval remains unclear. Here, we developed a novel chemogenetic activation strategy exploiting insect olfactory ionotropic receptors (IRs), termed "IR-mediated neuronal activation," and used it for selective stimulation of NE neurons in the locus coeruleus (LC). Drosophila melanogaster IR84a and IR8a subunits were expressed in LC NE neurons in transgenic mice. Application of phenylacetic acid (a specific ligand for the IR84a/IR8a complex) at appropriate doses induced excitatory responses of NE neurons expressing the receptors in both slice preparations and in vivo electrophysiological conditions, resulting in a marked increase of NE release in the LC nerve terminal regions (male and female). Ligand-induced activation of LC NE neurons enhanced the retrieval process of conditioned taste aversion without affecting taste sensitivity, general arousal state, and locomotor activity. This enhancing effect on taste memory retrieval was mediated, in part, through α1- and ß-adrenergic receptors in the basolateral nucleus of the amygdala (BLA; male). Pharmacological inhibition of LC NE neurons confirmed the facilitative role of these neurons in memory retrieval via adrenergic receptors in the BLA (male). Our findings indicate that the LC NE system, through projections to the BLA, controls the retrieval process of taste associative memory.SIGNIFICANCE STATEMENT Norepinephrine (NE)-containing neurons in the brain play a key role in the modulation of synaptic plasticity underlying various processes of memory formation, but the role of the NE system in memory retrieval remains unclear. We developed a chemogenetic activation system based on insect olfactory ionotropic receptors and used it for selective stimulation of NE neurons in the locus coeruleus (LC) in transgenic mice. Ligand-induced activation of LC NE neurons enhanced the retrieval of conditioned taste aversion, which was mediated, in part, through adrenoceptors in the basolateral amygdala. Pharmacological blockade of LC activity confirmed the facilitative role of these neurons in memory retrieval. Our findings indicate that the LC-amygdala pathway plays an important role in the recall of taste associative memory.


Locus Coeruleus/drug effects , Memory/physiology , Norepinephrine/physiology , Receptors, Adrenergic/physiology , Sensory Receptor Cells/physiology , Taste/physiology , Animals , Arousal/physiology , Drosophila melanogaster , Electrophysiological Phenomena , Humans , Locus Coeruleus/cytology , Memory/drug effects , Mental Recall/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/physiology , Phenylacetates/pharmacology , Receptors, Adrenergic/drug effects , Receptors, Odorant/physiology , Sensory Receptor Cells/drug effects , Taste/drug effects , Taste/genetics
10.
Cell ; 182(6): 1589-1605.e22, 2020 09 17.
Article En | MEDLINE | ID: mdl-32841600

Hunger and thirst have distinct goals but control similar ingestive behaviors, and little is known about neural processes that are shared between these behavioral states. We identify glutamatergic neurons in the peri-locus coeruleus (periLCVGLUT2 neurons) as a polysynaptic convergence node from separate energy-sensitive and hydration-sensitive cell populations. We develop methods for stable hindbrain calcium imaging in free-moving mice, which show that periLCVGLUT2 neurons are tuned to ingestive behaviors and respond similarly to food or water consumption. PeriLCVGLUT2 neurons are scalably inhibited by palatability and homeostatic need during consumption. Inhibition of periLCVGLUT2 neurons is rewarding and increases consumption by enhancing palatability and prolonging ingestion duration. These properties comprise a double-negative feedback relationship that sustains food or water consumption without affecting food- or water-seeking. PeriLCVGLUT2 neurons are a hub between hunger and thirst that specifically controls motivation for food and water ingestion, which is a factor that contributes to hedonic overeating and obesity.


Appetite Regulation/physiology , Drinking/physiology , Eating/physiology , Locus Coeruleus/cytology , Nerve Net/physiology , Neurons/physiology , Rhombencephalon/physiology , Single-Cell Analysis/methods , Animals , Appetite/physiology , Behavior Rating Scale , Feedback , Feeding Behavior/physiology , Female , Glutamine/metabolism , Glutamine/physiology , Homeostasis/physiology , Hunger/physiology , Male , Mice , Mice, Knockout , Motivation/physiology , Neurons/drug effects , Recombinant Proteins , Reward , Rhombencephalon/cytology , Rhombencephalon/diagnostic imaging , Taste/physiology , Thirst/physiology
11.
Neurobiol Aging ; 94: 243-249, 2020 10.
Article En | MEDLINE | ID: mdl-32663717

Certain neuronal populations, including basal forebrain cholinergic neurons (BFCN) and noradrenergic neurons of the locus coeruleus (LC), are selectively vulnerable to pathology and loss early in the course of aging and Alzheimer's disease (AD). Human BFCN show substantial loss of the calcium-binding protein (CBP), calbindin-D28K (CB), during normal aging, which is associated with formation of neurofibrillary tangles and BFCN loss in AD. Here we determined if, similar to the BFCN, LC neurons contain CB or the other 2 ubiquitous CBPs parvalbumin and calretinin, and whether these proteins display an age-related loss from LC neurons. Immunostaining for CBP and tyrosine hydroxylase, a marker of catecholaminergic neurons, was used in sections from the LC of young and aged human brains. Parvalbumin and calretinin immunoreactivities were completely absent from human LC neurons. A subpopulation of LC neurons (~10%) contained CB immunoreactivity. Quantitative analysis revealed no age-related loss of CB from LC neurons. Thus, unlike the BFCN, age-related loss of CB does not figure prominently in the selective vulnerability of LC neurons to degeneration in AD.


Adrenergic Neurons/metabolism , Adrenergic Neurons/pathology , Aging/metabolism , Aging/pathology , Calbindin 1/metabolism , Calbindin 2/metabolism , Cholinergic Neurons/metabolism , Cholinergic Neurons/pathology , Locus Coeruleus/cytology , Locus Coeruleus/metabolism , Parvalbumins/metabolism , Adult , Aged , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Female , Humans , Male , Middle Aged , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Young Adult
12.
Sci Rep ; 10(1): 7869, 2020 05 12.
Article En | MEDLINE | ID: mdl-32398643

The norepinephrine-releasing neurons in the locus coeruleus (LC) are well known to regulate wakefulness/arousal. They display active firing during wakefulness and a decreased discharge rate during sleep. We have previously reported that LC neurons express large numbers of GABAB receptors (GABABRs) located at peri-/extrasynaptic sites and are subject to tonic inhibition due to the continuous activation of GABABRs by ambient GABA, which is significantly higher during sleep than during wakefulness. In this study, we further showed using western blot analysis that the activation of GABABRs with baclofen could increase the level of phosphorylated extracellular signal-regulated kinase 1 (ERK1) in LC tissue. Recordings from LC neurons in brain slices showed that the inhibition of ERK1/2 with U0126 and FR180204 accelerated the decay of whole-cell membrane current induced by prolonged baclofen application. In addition, the inhibition of ERK1/2 also increased spontaneous firing and reduced tonic inhibition of LC neurons after prolonged exposure to baclofen. These results suggest a new role of GABABRs in mediating ERK1-dependent autoregulation of the stability of GABABR-activated whole-cell current, in addition to its well-known effect on gated potassium channels, to cause a tonic current in LC neurons.


Action Potentials/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Homeostasis , Neurons/physiology , Receptors, GABA-B/metabolism , Action Potentials/drug effects , Animals , Baclofen/pharmacology , Butadienes/pharmacology , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Female , GABA Agents/pharmacology , GABA-B Receptor Agonists/pharmacology , Locus Coeruleus/cytology , Locus Coeruleus/metabolism , Male , Neurons/cytology , Neurons/metabolism , Nitriles/pharmacology , Patch-Clamp Techniques/methods , Rats, Sprague-Dawley , gamma-Aminobutyric Acid/pharmacology
13.
Nat Commun ; 11(1): 471, 2020 01 24.
Article En | MEDLINE | ID: mdl-31980655

Astrocytes may function as mediators of the impact of noradrenaline on neuronal function. Activation of glial α1-adrenergic receptors triggers rapid astrocytic Ca2+ elevation and facilitates synaptic plasticity, while activation of ß-adrenergic receptors elevates cAMP levels and modulates memory consolidation. However, the dynamics of these processes in behaving mice remain unexplored, as do the interactions between the distinct second messenger pathways. Here we simultaneously monitored astrocytic Ca2+ and cAMP and demonstrate that astrocytic second messengers are regulated in a temporally distinct manner. In behaving mice, we found that while an abrupt facial air puff triggered transient increases in noradrenaline release and large cytosolic astrocytic Ca2+ elevations, cAMP changes were not detectable. By contrast, repeated aversive stimuli that lead to prolonged periods of vigilance were accompanied by robust noradrenergic axonal activity and gradual sustained cAMP increases. Our findings suggest distinct astrocytic signaling pathways can integrate noradrenergic activity during vigilance states to mediate distinct functions supporting memory.


Arousal/physiology , Astrocytes/physiology , Norepinephrine/physiology , Second Messenger Systems/physiology , Animals , Calcium Signaling/physiology , Conditioning, Classical/physiology , Cyclic AMP/metabolism , Fear/physiology , Fluorescent Dyes , Locus Coeruleus/cytology , Locus Coeruleus/physiology , Memory/physiology , Mice , Neuronal Plasticity/physiology , Parietal Lobe/cytology , Parietal Lobe/physiology , Receptors, Adrenergic/physiology
14.
J Vis Exp ; (153)2019 11 26.
Article En | MEDLINE | ID: mdl-31840660

Current scientific literature provides evidence that trigeminal sensorimotor activity associated with chewing may affect arousal, attention, and cognitive performance. These effects may be due to widespread connections of the trigeminal system to the ascending reticular activating system (ARAS), to which noradrenergic neurons of the locus coeruleus (LC) belongs. LC neurons contain projections to the whole brain, and it is known that their discharge co-varies with pupil size. LC activation is necessary for eliciting task-related mydriasis. If chewing effects on cognitive performance are mediated by the LC, it is reasonable to expect that changes in cognitive performance are correlated to changes in task-related mydriasis. Two novel protocols are presented here to verify this hypothesis and document that chewing effects are not attributable to aspecific motor activation. In both protocols, performance and pupil size changes observed during specific tasks are recorded before, soon after, and half an hour following a 2 min period of either: a) no activity, b) rhythmic, bilateral handgrip, c) bilateral chewing of soft pellet, and d) bilateral chewing of hard pellet. The first protocol measures level of performance in spotting target numbers displayed within numeric matrices. Since pupil size recordings are recorded by an appropriate pupillometer that impedes vision to ensure constant illumination levels, task-related mydriasis is evaluated during a haptic task. Results from this protocol reveal that 1) chewing-induced changes in performance and task-related mydriasis are correlated and 2) neither performance nor mydriasis are enhanced by handgrip. In the second protocol, use of a wearable pupillometer allows measurement of pupil size changes and performance during the same task, thus allowing even stronger evidence to be obtained regarding LC involvement in the trigeminal effects on cognitive activity. Both protocols have been run in the historical office of Prof. Giuseppe Moruzzi, the discoverer of ARAS, at the University of Pisa.


Arousal/physiology , Locus Coeruleus/physiology , Pupil/physiology , Trigeminal Motor Nucleus/physiology , Cognition/physiology , Humans , Locus Coeruleus/cytology , Male , Mastication/physiology , Neurons/physiology
15.
Sci Rep ; 9(1): 5262, 2019 03 27.
Article En | MEDLINE | ID: mdl-30918302

Parkinson's disease (PD) is the second most common neurodegenerative disorder and is characterized by motor deficits such as tremor, rigidity and bradykinesia. These symptoms are directly caused by the loss of dopaminergic neurons. However, a wealth of clinical evidence indicates that the dopaminergic system is not the only system affected in PD. Postmortem studies of brains from PD patients have revealed the degeneration of noradrenergic neurons in the locus coeruleus (LC) to the same or even greater extent than that observed in the dopaminergic neurons of substantia nigra (SN) and ventral tegmental area (VTA). Moreover, studies performed on rodent models suggest that enhancement of noradrenergic transmission may attenuate the PD-like phenotype induced by MPTP administration, a neurotoxin-based PD model. The aim of this study was to investigate whether chronic treatment with either of two compounds targeting the noradrenergic system (reboxetine or atipamezole) possess the ability to reduce the progression of a PD-like phenotype in a novel mouse model of progressive dopaminergic neurodegeneration induced by the genetic inhibition of rRNA synthesis in dopaminergic neurons, mimicking a PD-like phenotype. The results showed that reboxetine improved the parkinsonian phenotype associated with delayed progression of SN/VTA dopaminergic neurodegeneration and higher dopamine content in the striatum. Moreover, the alpha1-adrenergic agonist phenylephrine enhanced survival of TH+ neurons in primary cell cultures, supporting the putative neuroprotective effects of noradrenergic stimulation. Our results provide new insights regarding the possible influence of the noradrenergic system on dopaminergic neuron survival and strongly support the hypothesis regarding the neuroprotective role of noradrenaline.


Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/metabolism , Reboxetine/therapeutic use , Animals , Cells, Cultured , Chromatography, High Pressure Liquid , Disease Models, Animal , Female , Imidazoles/therapeutic use , Immunohistochemistry , Locus Coeruleus/cytology , Male , Mice , Mice, Inbred C57BL , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Ventral Tegmental Area/cytology
16.
Nature ; 568(7750): 93-97, 2019 04.
Article En | MEDLINE | ID: mdl-30918407

Sodium is the main cation in the extracellular fluid and it regulates various physiological functions. Depletion of sodium in the body increases the hedonic value of sodium taste, which drives animals towards sodium consumption1,2. By contrast, oral sodium detection rapidly quenches sodium appetite3,4, suggesting that taste signals have a central role in sodium appetite and its satiation. Nevertheless, the neural mechanisms of chemosensory-based appetite regulation remain poorly understood. Here we identify genetically defined neural circuits in mice that control sodium intake by integrating chemosensory and internal depletion signals. We show that a subset of excitatory neurons in the pre-locus coeruleus express prodynorphin, and that these neurons are a critical neural substrate for sodium-intake behaviour. Acute stimulation of this population triggered robust ingestion of sodium even from rock salt, while evoking aversive signals. Inhibition of the same neurons reduced sodium consumption selectively. We further demonstrate that the oral detection of sodium rapidly suppresses these sodium-appetite neurons. Simultaneous in vivo optical recording and gastric infusion revealed that sodium taste-but not sodium ingestion per se-is required for the acute modulation of neurons in the pre-locus coeruleus that express prodynorphin, and for satiation of sodium appetite. Moreover, retrograde-virus tracing showed that sensory modulation is in part mediated by specific GABA (γ-aminobutyric acid)-producing neurons in the bed nucleus of the stria terminalis. This inhibitory neural population is activated by sodium ingestion, and sends rapid inhibitory signals to sodium-appetite neurons. Together, this study reveals a neural architecture that integrates chemosensory signals and the internal need to maintain sodium balance.


Appetite Regulation/drug effects , Appetite Regulation/physiology , Eating/drug effects , Neural Pathways/drug effects , Sodium/pharmacology , Taste/drug effects , Taste/physiology , Administration, Oral , Animals , Appetite Regulation/genetics , Avoidance Learning/drug effects , Avoidance Learning/physiology , Eating/genetics , Eating/physiology , Enkephalins/metabolism , Female , GABAergic Neurons/drug effects , GABAergic Neurons/metabolism , Homeostasis/drug effects , Homeostasis/genetics , Homeostasis/physiology , Locus Coeruleus/cytology , Locus Coeruleus/drug effects , Locus Coeruleus/physiology , Male , Mice , Motivation/drug effects , Neural Pathways/cytology , Neural Pathways/physiology , Neurons/drug effects , Neurons/physiology , Protein Precursors/metabolism , Satiety Response/drug effects , Satiety Response/physiology , Sodium/administration & dosage , Taste/genetics
17.
Neuron ; 102(3): 653-667.e6, 2019 05 08.
Article En | MEDLINE | ID: mdl-30879785

SIM1-expressing paraventricular hypothalamus (PVH) neurons are key regulators of energy balance. Within the PVHSIM1 population, melanocortin-4 receptor-expressing (PVHMC4R) neurons are known to regulate satiety and bodyweight, yet they account for only half of PVHSIM1 neuron-mediated regulation. Here we report that PVH prodynorphin-expressing (PVHPDYN) neurons, which notably lack MC4Rs, function independently and additively with PVHMC4R neurons to account for the totality of PVHSIM1 neuron-mediated satiety. Moreover, PVHPDYN neurons are necessary for prevention of obesity in an independent but equipotent manner to PVHMC4R neurons. While PVHPDYN and PVHMC4R neurons both project to the parabrachial complex (PB), they synaptically engage distinct efferent nodes, the pre-locus coeruleus (pLC), and central lateral parabrachial nucleus (cLPBN), respectively. PB-projecting PVHPDYN neurons, like PVHMC4R neurons, receive input from interoceptive ARCAgRP neurons, respond to caloric state, and are sufficient and necessary to control food intake. This expands the CNS satiety circuitry to include two non-overlapping PVH to hindbrain circuits.


Feeding Behavior/physiology , Neurons/cytology , Obesity/physiopathology , Paraventricular Hypothalamic Nucleus/cytology , Satiety Response/physiology , Agouti-Related Protein/metabolism , Animals , Arcuate Nucleus of Hypothalamus/cytology , Arcuate Nucleus of Hypothalamus/metabolism , Arcuate Nucleus of Hypothalamus/physiology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Energy Metabolism , Enkephalins/metabolism , Locus Coeruleus/cytology , Locus Coeruleus/metabolism , Locus Coeruleus/physiology , Mice , Neurons/metabolism , Neurons/physiology , Parabrachial Nucleus/cytology , Parabrachial Nucleus/metabolism , Parabrachial Nucleus/physiology , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/physiology , Protein Precursors/metabolism , Receptor, Melanocortin, Type 4/metabolism , Repressor Proteins/metabolism
18.
Sci Rep ; 9(1): 5084, 2019 03 25.
Article En | MEDLINE | ID: mdl-30911100

In the central nervous system of vertebrates, cell bodies of neurons are often assembled as nuclei or cellular layers that play specific roles as functional units. The purpose of this work was to selectively highlight such cell assemblies by magnetic resonance imaging using signals from water protons that are associated with intracellular paramagnetic ions, while saturating lipid-associated water protons as well as extracellular free water protons. Given the significant correlation between image signal intensity and water proton density, the high signal intensities observed for such cell assemblies must be attributed to their abundant paramagnetic-ion-associated water protons. In the hippocampal formation, the technique visualized cell assemblies that were so far not depicted in human in vivo. In the brainstem, the technique delineated noradrenergic neuron groups such as the locus coeruleus in human and mice in vivo. Their reduced magnetization-transfer ratios together with their prolonged relaxation times compared to other gray matter indicate that the source of their high signal intensity is not the presence of T1-shortening molecules, e.g., neuromelanin, but their high water content. Given the general absence of neuromelanin in noradrenergic neurons of rodents, their high signal intensity in mice in vivo further supports this view.


Body Water/metabolism , Brain/metabolism , Magnetic Resonance Imaging/methods , Animals , Brain/cytology , Brain Stem/cytology , Brain Stem/metabolism , Humans , Locus Coeruleus/cytology , Locus Coeruleus/metabolism , Melanins/metabolism , Mice
19.
Nat Commun ; 9(1): 5211, 2018 12 06.
Article En | MEDLINE | ID: mdl-30523254

Locus coeruleus (LC) neurons in the brainstem have long been associated with attention and arousal. Optogenetic stimulation of LC-NE neurons induces immediate sleep-to-wake transitions. However, LC neurons also secrete other neurotransmitters in addition to NE. To interrogate the role of NE derived from the LC in regulating wakefulness, we applied in vivo cell type-specific CRISPR/Cas9 technology to disrupt the dopamine beta hydroxylase (dbh) gene selectively in adult LC-NE neurons. Unilateral dbh gene disruption abolished immediate arousal following optogenetic stimulation of LC. Bilateral LC-specific dbh disruption significantly reduced NE concentration in LC projection areas and reduced wake length even in the presence of salient stimuli. These results suggest that NE may be crucial for the awakening effect of LC stimulation and serve as proof-of-principle that CRISPR gene editing in adult neurons can be used to interrogate gene function within genetically-defined neuronal circuitry associated with complex behaviors.


CRISPR-Cas Systems , Dopamine beta-Hydroxylase/genetics , Locus Coeruleus/metabolism , Wakefulness/genetics , Animals , Dopamine beta-Hydroxylase/metabolism , Electric Stimulation , Female , Locus Coeruleus/cytology , Locus Coeruleus/physiology , Male , Mice , Mice, Knockout , Mice, Transgenic , NIH 3T3 Cells , Neurons/metabolism , Neurons/physiology , Norepinephrine/metabolism , Sleep/genetics , Sleep/physiology , Wakefulness/physiology
20.
Proc Natl Acad Sci U S A ; 115(40): E9439-E9448, 2018 10 02.
Article En | MEDLINE | ID: mdl-30232259

Phasic activation of locus coeruleus (LC)-norepinephrine (NE) neurons is associated with focused attention and behavioral responses to salient stimuli. We used cell-type-specific optogenetics and single-unit neurophysiology to identify how LC activity influences neural encoding of sensory information. We found that phasic, but not tonic, LC-NE photoactivation generated a distinct event-related potential (ERP) across cortical regions. Salient sensory stimuli (which innately trigger phasic LC activity) produced strong excitatory cortical responses during this ERP window. Application of weaker, nonsalient stimuli produced limited responses, but these responses were elevated to salient stimulus levels when they were temporally locked with phasic LC photoactivation. These results demonstrate that phasic LC activity enhances cortical encoding of salient stimuli by facilitating long-latency signals within target regions in response to stimulus intensity/salience. The LC-driven salience signal identified here provides a measure of phasic LC activity that can be used to investigate the LC's role in attentional processing across species.


Evoked Potentials/physiology , Locus Coeruleus/physiology , Neurons/metabolism , Animals , Locus Coeruleus/cytology , Male , Neurons/cytology , Optogenetics , Rats , Rats, Long-Evans
...