Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nature ; 586(7828): 270-274, 2020 10.
Article in English | MEDLINE | ID: mdl-32999460

ABSTRACT

The ability to recognize information that is incongruous with previous experience is critical for survival. Novelty signals have therefore evolved in the mammalian brain to enhance attention, perception and memory1,2. Although the importance of regions such as the ventral tegmental area3,4 and locus coeruleus5 in broadly signalling novelty is well-established, these diffuse monoaminergic transmitters have yet to be shown to convey specific information on the type of stimuli that drive them. Whether distinct types of novelty, such as contextual and social novelty, are differently processed and routed in the brain is unknown. Here we identify the supramammillary nucleus (SuM) as a novelty hub in the hypothalamus6. The SuM region is unique in that it not only responds broadly to novel stimuli, but also segregates and selectively routes different types of information to discrete cortical targets-the dentate gyrus and CA2 fields of the hippocampus-for the modulation of mnemonic processing. Using a new transgenic mouse line, SuM-Cre, we found that SuM neurons that project to the dentate gyrus are activated by contextual novelty, whereas the SuM-CA2 circuit is preferentially activated by novel social encounters. Circuit-based manipulation showed that divergent novelty channelling in these projections modifies hippocampal contextual or social memory. This content-specific routing of novelty signals represents a previously unknown mechanism that enables the hypothalamus to flexibly modulate select components of cognition.


Subject(s)
Hippocampus/cytology , Hippocampus/physiology , Memory/physiology , Neural Pathways/physiology , Animals , CA2 Region, Hippocampal/cytology , CA2 Region, Hippocampal/physiology , Cognition , Dentate Gyrus/cytology , Dentate Gyrus/physiology , Female , Hypothalamus, Posterior/cytology , Hypothalamus, Posterior/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism , Social Interaction
2.
Development ; 140(8): 1762-73, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23533176

ABSTRACT

The hypothalamus is a key integrative center in the brain that consists of diverse cell types required for a variety of functions including homeostasis, reproduction, stress response, social and cognitive behavior. Despite our knowledge of several transcription factors crucial for hypothalamic development, it is not known how the wide diversity of neuron types in the hypothalamus is produced. In particular, almost nothing is known about the mechanisms that specify neurons in the posteriormost part of the hypothalamus, the mammillary area. Here, we investigated the specification of two distinct neuron types in the mammillary area that produce the hypothalamic hormones Vasoactive intestinal peptide (Vip) and Urotensin 1 (Uts1). We show that Vip- and Uts1-positive neurons develop in distinct domains in the mammillary area defined by the differential expression of the transcription factors Fezf2, Otp, Sim1a and Foxb1.2. Coordinated activities of these factors are crucial for the establishment of the mammillary area subdomains and the specification of Vip- and Uts1-positive neurons. In addition, Fezf2 is important for early development of the posterior hypothalamus. Thus, our study provides the first molecular anatomical map of the posterior hypothalamus in zebrafish and identifies, for the first time, molecular requirements underlying the specification of distinct posterior hypothalamic neuron types.


Subject(s)
Cell Differentiation/physiology , Forkhead Transcription Factors/metabolism , Hypothalamus, Posterior/cytology , Hypothalamus, Posterior/embryology , Nerve Tissue Proteins/metabolism , Neurogenesis/physiology , Neurons/physiology , Zebrafish/embryology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Immunohistochemistry , In Situ Hybridization , Morpholinos/genetics , Neurons/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Urotensins/metabolism , Vasoactive Intestinal Peptide/metabolism , Zebrafish Proteins/metabolism
3.
Neuroendocrinology ; 103(6): 678-92, 2016.
Article in English | MEDLINE | ID: mdl-26562443

ABSTRACT

BACKGROUND/AIMS: Photoperiod is a major environmental cue in temperate-zone birds which synchronizes breeding with the time of year that offers the optimal environment for offspring survival. Despite continued long photoperiods, these birds eventually become refractory to the stimulating photoperiod and their reproductive systems regress. In this study, we characterized the role of γ-aminobutyric acid (GABA)ergic neurotransmission in modulating the response of the premammillary nucleus (PMM) to a gonad stimulatory photoperiod and the onset of photorefractoriness. METHODS AND RESULTS: Bilateral ablation of the PMM blocked the light-induced neuroendocrine response from occurring in photosensitive turkeys. Microarray analyses revealed an increase in GABAergic activity in the PMM of photorefractory birds as opposed to photosensitive ones, and this enhanced GABAergic activity appeared to inhibit the photoperiodic signal. Additionally, GABAA and GABAB receptors were expressed by dopamine-melatonin neurons in the PMM, and the administration of the GABA receptor agonist baclofen blocked the photoperiodic reproductive neuroendocrine responses. CONCLUSIONS: Consistent with the present findings, we propose that the long-sought-after mechanism underlying photorefractoriness is linked to the inhibitory actions of GABA. We suggest that (1) GABAergic interference with photoperiodic entrainment in the PMM initiates the photorefractory state and terminates the annual breeding season in temperate-zone birds, and (2) the PMM is a site of photoreception and photorefractoriness that controls the initiation and termination of avian reproductive seasonality.


Subject(s)
Hypothalamus, Posterior/injuries , Light , Photoperiod , Reproduction/physiology , Seasons , Synaptic Transmission/physiology , gamma-Aminobutyric Acid/metabolism , Animals , Dopamine/metabolism , Female , GABA Agents/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Gene Expression Regulation/radiation effects , Glutamate Decarboxylase/metabolism , Hypothalamus, Posterior/cytology , Melatonin/metabolism , Neurons/metabolism , RNA, Messenger/metabolism , Receptors, GABA/metabolism , Synaptic Transmission/drug effects , Synaptic Transmission/radiation effects , Turkey
4.
Adv Gerontol ; 28(1): 37-41, 2015.
Article in Russian | MEDLINE | ID: mdl-26390608

ABSTRACT

Morphometric parameters of neuronal metabolic activity, such as the area of neuronal nuclei and perikarya and nuclear-cytoplasmic ratio, in the nucleus basalis of Meynert (NBM), tuberomamillary (TMN) and medial mammillary (MMN) hypothalamic nuclei of human subjects belonging to four age groups were studied. Statistically significant increase in the size of neuronal perikarya and their nuclei was found in elderly people aged 60-74 years. The surge in the metabolic activity of neurons in the NBM starts earlier than in the TMN and MMN, and becomes apparent morphologically in people of middle age (45-59 years). The age-related increase in the metabolic activity of neurons in the studied structures of the human brain participating in the regulation of memory and other cognitive functions, may represent protective, adaptive and/or compensatory mechanisms of the aging process that also prevents the development of Alzheimer's disease.


Subject(s)
Aging , Basal Nucleus of Meynert/cytology , Cell Nucleus , Cell Size , Hypothalamus, Posterior/cytology , Neurons/cytology , Adult , Aged , Aged, 80 and over , Cell Count , Female , Humans , Male , Middle Aged
5.
J Clin Invest ; 118(7): 2471-81, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18551194

ABSTRACT

Stress-induced analgesia (SIA) is a key component of the defensive behavioral "fight-or-flight" response. Although the neural substrates of SIA are incompletely understood, previous studies have implicated the hypocretin/orexin (Hcrt) and nociceptin/orphanin FQ (N/OFQ) peptidergic systems in the regulation of SIA. Using immunohistochemistry in brain tissue from wild-type mice, we identified N/OFQ-containing fibers forming synaptic contacts with Hcrt neurons at both the light and electron microscopic levels. Patch clamp recordings in GFP-tagged mouse Hcrt neurons revealed that N/OFQ hyperpolarized, decreased input resistance, and blocked the firing of action potentials in Hcrt neurons. N/OFQ postsynaptic effects were consistent with opening of a G protein-regulated inwardly rectifying K+ (GIRK) channel. N/OFQ also modulated presynaptic release of GABA and glutamate onto Hcrt neurons in mouse hypothalamic slices. Orexin/ataxin-3 mice, in which the Hcrt neurons degenerate, did not exhibit SIA, although analgesia was induced by i.c.v. administration of Hcrt-1. N/OFQ blocked SIA in wild-type mice, while coadministration of Hcrt-1 overcame N/OFQ inhibition of SIA. These results establish what is, to our knowledge, a novel interaction between the N/OFQ and Hcrt systems in which the corticotropin-releasing factor and N/OFQ systems coordinately modulate the Hcrt neurons to regulate SIA.


Subject(s)
Analgesia , Intracellular Signaling Peptides and Proteins/metabolism , Neuropeptides/metabolism , Opioid Peptides/metabolism , Stress, Physiological/physiopathology , Animals , Ataxin-3 , Behavior, Animal/drug effects , Behavior, Animal/physiology , Brain/cytology , Brain/drug effects , Brain/metabolism , Calcium/metabolism , Cytoplasm/metabolism , Electrophysiology , Female , Hypothalamus, Posterior/cytology , Hypothalamus, Posterior/metabolism , Hypothalamus, Posterior/ultrastructure , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/pharmacology , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Narcotic Antagonists , Neurons/cytology , Neurons/drug effects , Neurons/physiology , Neuropeptides/genetics , Neuropeptides/pharmacology , Nuclear Proteins/genetics , Opioid Peptides/genetics , Opioid Peptides/pharmacology , Orexins , Pain Threshold/drug effects , Pain Threshold/physiology , Presynaptic Terminals/physiology , Reaction Time/drug effects , Reaction Time/physiology , Receptors, Opioid , Tetrodotoxin/pharmacology , Transcription Factors/genetics , Nociceptin Receptor , Nociceptin
6.
Anesthesiology ; 115(1): 36-43, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21562401

ABSTRACT

BACKGROUND: The brain histaminergic system plays a critical role in maintenance of arousal. Previous studies suggest that histaminergic neurotransmission might be a potential mediator of general anesthetic actions. However, it is not clear whether histaminergic tuberomamillary nucleus (TMN) is necessarily involved in the sedative/hypnotic effects of general anesthetics. METHODS: Male Long Evans rats underwent either TMN orexin-saporin/sham lesion or implantation of intracerebroventricular cannula 2 weeks before the experiment. The behavioral endpoint of loss of righting reflex was used to assess the hypnotic property of isoflurane, propofol, pentobarbital, and ketamine in animals. Histaminergic cell loss was assessed by adenosine deaminase expression in the TMN using immunohistochemistry. RESULTS: Rats with bilateral TMN orexin-saporin lesion induced an average 72% loss of histaminergic cells compared with sham-lesion rats. TMN orexin-saporin lesion or intracerebroventricular administration of triprolidine (an H1 receptor antagonist) decreased the 50% effective concentration for loss of righting reflex value and prolonged emergence time to isoflurane anesthesia. However, TMN orexin-saporin lesion had no significant effect on the anesthetic sensitivity to propofol, pentobarbital, and ketamine. CONCLUSIONS: These findings suggest a role of the TMN histaminergic neurons in modulating isoflurane anesthesia and that the neural circuits for isoflurane-induced hypnosis may differ from those of γ-aminobutyric acid-mediated anesthetics and ketamine.


Subject(s)
Anesthesia, Inhalation , Anesthetics, Inhalation , Histamine/physiology , Hypothalamus, Posterior/physiology , Isoflurane , Neurons/physiology , Anesthetics, Dissociative/pharmacology , Anesthetics, Intravenous/pharmacology , Animals , Brain Chemistry/physiology , Histamine Antagonists , Hypothalamus, Posterior/cytology , Immunohistochemistry , Injections, Intraventricular , Ketamine/pharmacology , Male , Orexin Receptors , Pentobarbital/pharmacology , Postural Balance/drug effects , Propofol/pharmacology , Rats , Rats, Long-Evans , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, Neuropeptide/antagonists & inhibitors , Reflex/drug effects , Ribosome Inactivating Proteins, Type 1/pharmacology , Saporins
7.
Science ; 374(6574): 1492-1496, 2021 Dec 17.
Article in English | MEDLINE | ID: mdl-34914519

ABSTRACT

Locomotor speed is a basic input used to calculate one's position, but where this signal comes from is unclear. We identified neurons in the supramammillary nucleus (SuM) of the rodent hypothalamus that were highly correlated with future locomotor speed and reliably drove locomotion when activated. Robust locomotion control was specifically identified in Tac1 (substance P)­expressing (SuMTac1+) neurons, the activation of which selectively controlled the activity of speed-modulated hippocampal neurons. By contrast, Tac1-deficient (SuMTac1−) cells weakly regulated locomotion but potently controlled the spike timing of hippocampal neurons and were sufficient to entrain local network oscillations. These findings emphasize that the SuM not only regulates basic locomotor activity but also selectively shapes hippocampal neural activity in a manner that may support spatial navigation.


Subject(s)
Hippocampus/physiology , Hypothalamus, Posterior/physiology , Locomotion , Neurons/physiology , Action Potentials , Animals , Hippocampus/cytology , Hypothalamus, Posterior/cytology , Mice , Mice, Inbred C57BL , Neural Pathways/physiology , Rats , Spatial Navigation , Substance P/genetics , Theta Rhythm
8.
Nat Commun ; 12(1): 2811, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33990558

ABSTRACT

The supramammillary region (SuM) is a posterior hypothalamic structure, known to regulate hippocampal theta oscillations and arousal. However, recent studies reported that the stimulation of SuM neurons with neuroactive chemicals, including substances of abuse, is reinforcing. We conducted experiments to elucidate how SuM neurons mediate such effects. Using optogenetics, we found that the excitation of SuM glutamatergic (GLU) neurons was reinforcing in mice; this effect was relayed by their projections to septal GLU neurons. SuM neurons were active during exploration and approach behavior and diminished activity during sucrose consumption. Consistently, inhibition of SuM neurons disrupted approach responses, but not sucrose consumption. Such functions are similar to those of mesolimbic dopamine neurons. Indeed, the stimulation of SuM-to-septum GLU neurons and septum-to-ventral tegmental area (VTA) GLU neurons activated mesolimbic dopamine neurons. We propose that the supramammillo-septo-VTA pathway regulates arousal that reinforces and energizes behavioral interaction with the environment.


Subject(s)
Dopaminergic Neurons/physiology , Hypothalamus, Posterior/cytology , Hypothalamus, Posterior/physiology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Consummatory Behavior/drug effects , Consummatory Behavior/physiology , Dopamine/physiology , Female , Glutamic Acid/physiology , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Neurological , Neural Pathways/cytology , Neural Pathways/physiology , Optogenetics , Rats , Rats, Wistar , Reinforcement, Psychology , Septum of Brain/cytology , Septum of Brain/drug effects , Septum of Brain/physiology , Ventral Tegmental Area/cytology , Ventral Tegmental Area/physiology , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/administration & dosage
9.
Elife ; 92020 10 29.
Article in English | MEDLINE | ID: mdl-33119507

ABSTRACT

The ventral posterior hypothalamus (VPH) is an anatomically complex brain region implicated in arousal, reproduction, energy balance, and memory processing. However, neuronal cell type diversity within the VPH is poorly understood, an impediment to deconstructing the roles of distinct VPH circuits in physiology and behavior. To address this question, we employed a droplet-based single-cell RNA sequencing (scRNA-seq) approach to systematically classify molecularly distinct cell populations in the mouse VPH. Analysis of >16,000 single cells revealed 20 neuronal and 18 non-neuronal cell populations, defined by suites of discriminatory markers. We validated differentially expressed genes in selected neuronal populations through fluorescence in situ hybridization (FISH). Focusing on the mammillary bodies (MB), we discovered transcriptionally-distinct clusters that exhibit neuroanatomical parcellation within MB subdivisions and topographic projections to the thalamus. This single-cell transcriptomic atlas of VPH cell types provides a resource for interrogating the circuit-level mechanisms underlying the diverse functions of VPH circuits.


Subject(s)
Hypothalamus, Posterior/cytology , Animals , Female , Gene Expression Profiling , Hypothalamus, Posterior/anatomy & histology , Hypothalamus, Posterior/physiology , Male , Mice , Mice, Inbred C57BL , RNA/genetics , Sequence Analysis, RNA , Single-Cell Analysis
10.
Brain Struct Funct ; 225(9): 2643-2668, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32970253

ABSTRACT

Several studies suggest that neurons from the lateral region of the SuM (SuML) innervating the dorsal dentate gyrus (DG) display a dual GABAergic and glutamatergic transmission and are specifically activated during paradoxical (REM) sleep (PS). The objective of the present study is to characterize the anatomical, neurochemical and electrophysiological properties of the SuML-DG projection neurons and to determine how they control DG oscillations and neuronal activation during PS and other vigilance states. For this purpose, we combine structural connectivity techniques using neurotropic viral vectors (rabies virus, AAV), neurochemical anatomy (immunohistochemistry, in situ hybridization) and imaging (light, electron and confocal microscopy) with in vitro (patch clamp) and in vivo (LFP, EEG) optogenetic and electrophysiological recordings performed in transgenic VGLUT2-cre male mice. At the cellular level, we show that the SuML-DG neurons co-release GABA and glutamate on dentate granule cells and increase the activity of a subset of DG granule cells. At the network level, we show that activation of the SuML-DG pathway increases theta power and frequency during PS as well as gamma power during PS and waking in the DG. At the behavioral level, we show that the activation of this pathway does not change animal behavior during PS, induces awakening during slow wave sleep and increases motor activity during waking. These results suggest that the SuML-DG pathway is capable of supporting the increase of theta and gamma power in the DG observed during PS and plays an important modulatory role of DG network activity during this state.


Subject(s)
Dentate Gyrus/physiology , GABAergic Neurons/physiology , Gamma Rays , Glutamic Acid/physiology , Hypothalamus, Posterior/physiology , Neurons/physiology , Sleep, REM/physiology , Theta Rhythm , Animals , Dentate Gyrus/cytology , GABAergic Neurons/cytology , Hypothalamus, Posterior/cytology , Male , Membrane Potentials , Mice, Transgenic , Neurons/cytology
11.
J Neuroendocrinol ; 32(2): e12829, 2020 02.
Article in English | MEDLINE | ID: mdl-31925973

ABSTRACT

The supramammillary nucleus (SuM) has an emerging role in appetite control. We have shown that the rat SuM is activated during hunger or food anticipation, or by ghrelin administration. In the present study, we characterised the connectivity between the SuM and key appetite- and motivation-related nuclei in the rat. In adult wild-type rats, or rats expressing Cre recombinase under the control of the tyrosine hydroxylase (TH) promoter (TH-Cre rats), we used c-Fos immunohistochemistry to visualise and correlate the activation of medial SuM (SuMM) with activation in the lateral hypothalamic area (LH), the dorsomedial hypothalamus (DMH) or the ventral tegmental area (VTA) after voluntary consumption of a high-sugar, high-fat food. To determine neuroanatomical connectivity, we used retrograde and anterograde tracing methods to specifically investigate the neuronal inputs and outputs of the SuMM. After consumption of the food there were positive correlations between c-Fos expression in the SuMM and the LH, DMH and VTA (P = 0.0001, 0.01 and 0.004). Using Fluoro-Ruby as a retrograde tracer, we demonstrate the existence of inputs from the LH, DMH, VTA and ventromedial hypothalamus (VMH) to the SuMM. The SuMM showed reciprocal inputs to the LH and DMH, and we identified a TH-positive output from SuMM to DMH. We co-labelled retrogradely-labelled sections for TH in the VMH, or for TH, orexin and melanin-concentrating hormone in the LH and DMH. However, we did not observe any colocalisation of immunoreactivity with any retrogradely-labelled cells. Viral mapping in TH-Cre rats confirms the existence of a reciprocal SuMM-DMH connection and shows that TH-positive cells project from the SuMM and VTA to the lateral septal area and cingulate cortex, respectively. These data provide evidence for the connectivity of the SuMM to brain regions involved in appetite control, and form the foundation for functional and behavioural studies aiming to further characterise the brain circuitry controlling eating behaviours.


Subject(s)
Appetite/physiology , Brain/cytology , Brain/physiology , Hypothalamus, Posterior/cytology , Hypothalamus, Posterior/physiology , Motivation/physiology , Neurons/physiology , Animals , Appetite Regulation , Male , Neural Pathways/cytology , Neural Pathways/physiology , Neuroanatomical Tract-Tracing Techniques , Neurons/cytology , Proto-Oncogene Proteins c-fos/metabolism , Rats, Long-Evans , Rats, Sprague-Dawley , Tyrosine 3-Monooxygenase/metabolism , Ventral Tegmental Area/cytology , Ventral Tegmental Area/physiology
12.
Science ; 220(4603): 1309-11, 1983 Jun 17.
Article in English | MEDLINE | ID: mdl-6857253

ABSTRACT

Three groups of gamma-aminobutyric acid--containing neurons were found in the mammillary region of the posterior hypothalamus. The groups correspond to the tuberal, caudal, and postmammillary caudal magnocellular nuclei. Many cells in these nuclei were retrogradely labeled with fast blue after the injection of this fluorescent dye into the neocortex. Immunohistochemical experiments showed that these same neurons also contained the gamma-aminobutyric acid-synthesizing enzyme glutamate decarboxylase. These results provide morphological evidence for a gamma-aminobutyric acid pathway arising in magnocellular neurons of the posterior hypothalamus and innervating the neocortex.


Subject(s)
Cerebral Cortex/anatomy & histology , Hypothalamus, Posterior/anatomy & histology , Hypothalamus/anatomy & histology , gamma-Aminobutyric Acid/physiology , Animals , Cerebral Cortex/cytology , Glutamate Decarboxylase/physiology , Hypothalamus, Posterior/cytology , Microscopy, Fluorescence , Neural Pathways/anatomy & histology , Rats
13.
Anesthesiology ; 110(2): 326-32, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19194159

ABSTRACT

BACKGROUND: It is well documented that several general anesthetics, including propofol, potentiate glycine receptor function. Furthermore, glycine receptors exist throughout the central nervous system, including areas of the brain thought to be involved in sleep. However, the role of glycine receptors in anesthetic-induced hypnosis has not been determined. METHODS: Experiments were conducted in rats where the loss of righting reflex (LORR) was used as a marker of the hypnotic state. Propofol-induced LORR was examined in the presence and absence of strychnine (a glycine receptor antagonist), GABAzine (a gamma-aminobutyric acid A receptor antagonist), as well as ketamine (an antagonist of N-methyl-D-aspartic acid subtype of glutamate receptors). Furthermore, the effects of propofol on the currents elicited by glycine and gamma-aminobutyric acid were analyzed in neurons isolated from the posterior hypothalamus of rats. The effects of strychnine and GABAzine on propofol-induced currents were also evaluated. RESULTS: Strychnine and GABAzine dose-dependently reduced the percentage of rats exhibiting LORR induced by propofol. Furthermore, strychnine significantly increased the onset time and reduced the duration of LORR induced by propofol. In contrast, strychnine did not affect the LORR induced by ketamine. In addition, propofol markedly increased the currents elicited by glycine and GABA of hypothalamic neurons. Conversely, strychnine and GABAzine both profoundly attenuated the current induced by propofol. CONCLUSION: Strychnine, the glycine receptor antagonist, dose-dependently reduced propofol-induced LORR in rats and propofol-induced current of rat hypothalamic neurons. These results suggest that neuronal glycine receptors partially contribute to propofol-induced hypnosis.


Subject(s)
Anesthetics, Intravenous/pharmacology , Behavior, Animal/drug effects , Brain Chemistry/drug effects , Propofol/pharmacology , Receptors, Glycine/drug effects , Animals , Catheterization , Dose-Response Relationship, Drug , Electrophysiology , Excitatory Amino Acid Antagonists/pharmacology , Female , GABA Antagonists/pharmacology , GABA-A Receptor Antagonists , Glycine Agents/pharmacology , Hypothalamus, Posterior/cytology , Hypothalamus, Posterior/drug effects , Ketamine/pharmacology , Neurons/drug effects , Patch-Clamp Techniques , Postural Balance/drug effects , Pyridazines/pharmacology , Rats , Rats, Sprague-Dawley , Reflex/drug effects , Strychnine/pharmacology
14.
Brain Struct Funct ; 224(8): 2983-2999, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31485718

ABSTRACT

Guanylyl cyclase C (GUCY2C) is the afferent central receptor in the gut-brain endocrine axis regulated by the anorexigenic intestinal hormone uroguanylin. GUCY2C mRNA and protein are produced in the hypothalamus, a major center regulating appetite and metabolic homeostasis. Further, GUCY2C mRNA and protein are expressed in the ventral midbrain, a principal structure regulating hedonic reward from behaviors including eating. While GUCY2C is expressed in hypothalamus and midbrain, its precise neuroanatomical organization and relationship with circuits regulating satiety remain unknown. Here, we reveal that hypothalamic GUCY2C mRNA is confined to the ventral premammillary nucleus (PMV), while in midbrain it is produced by neurons in the ventral tegmental area (VTA) and substantia nigra (SN). GUCY2C in the PMV is produced by 46% of neurons expressing anorexigenic leptin receptors, while in the VTA/SN it is produced in most tyrosine hydroxylase-immunoreactive neurons. In contrast to mRNA, GUCY2C protein is widely distributed throughout the brain in canonical sites of PMV and VTA/SN axonal projections. Selective stereotaxic ablation of PMV or VTA/SN neurons eliminated GUCY2C only in their respective canonical projection sites. Conversely, specific anterograde tracer analyses of PMV or VTA/SN neurons confirmed distinct GUCY2C-immunoreactive axons projecting to those canonical locations. Together, these findings reveal two discrete neuronal circuits expressing GUCY2C originating in the PMV in the hypothalamus and in the VTA/SN in midbrain, which separately project to other sites throughout the brain. They suggest a structural basis for a role for the GUCY2C-uroguanylin gut-brain endocrine axis in regulating homeostatic and behavioral components contributing to satiety.


Subject(s)
Hypothalamus, Posterior/metabolism , Receptors, Enterotoxin/analysis , Substantia Nigra/metabolism , Ventral Tegmental Area/metabolism , Animals , Axons , Female , Hypothalamus, Posterior/cytology , Male , Mice, Inbred C57BL , Neural Pathways/cytology , RNA, Messenger/analysis , Substantia Nigra/cytology , Ventral Tegmental Area/cytology
15.
Brain Struct Funct ; 224(1): 99-110, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30284033

ABSTRACT

Neurons containing melanin-concentrating hormone (MCH) in the posterior lateral hypothalamus play an integral role in rapid eye movement sleep (REMs) regulation. As MCH neurons also contain a variety of other neuropeptides [e.g., cocaine- and amphetamine-regulated transcript (CART) and nesfatin-1] and neurotransmitters (e.g., glutamate), the specific neurotransmitter responsible for REMs regulation is not known. We hypothesized that glutamate, the primary fast-acting neurotransmitter in MCH neurons, is necessary for REMs regulation. To test this hypothesis, we deleted vesicular glutamate transporter (Vglut2; necessary for synaptic release of glutamate) specifically from MCH neurons by crossing MCH-Cre mice (expressing Cre recombinase in MCH neurons) with Vglut2flox/flox mice (expressing LoxP-modified alleles of Vglut2), and studied the amounts, architecture and diurnal variation of sleep-wake states during baseline conditions. We then activated the MCH neurons lacking glutamate neurotransmission using chemogenetic methods and tested whether these MCH neurons still promoted REMs. Our results indicate that glutamate in MCH neurons contributes to normal diurnal variability of REMs by regulating the levels of REMs during the dark period, but MCH neurons can promote REMs even in the absence of glutamate.


Subject(s)
Circadian Rhythm , Glutamic Acid/metabolism , Hypothalamic Hormones/metabolism , Hypothalamus, Posterior/metabolism , Melanins/metabolism , Neurons/metabolism , Pituitary Hormones/metabolism , Sleep, REM , Animals , Hypothalamic Hormones/genetics , Hypothalamus, Posterior/cytology , Male , Melanins/genetics , Mice, Transgenic , Photoperiod , Pituitary Hormones/genetics , Time Factors , Vesicular Glutamate Transport Protein 2/genetics , Vesicular Glutamate Transport Protein 2/metabolism , Wakefulness
16.
Neurosci Behav Physiol ; 38(9): 907-11, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18975111

ABSTRACT

The aim of the present work was to evaluate structural and metabolic changes in histaminergic neurons in hypothalamic nucleus E2 in rats in conditions of complete external drainage of bile. Studies were performed on male Wistar rats (n = 45). Controls consisted of animals subjected to sham surgery with preservation of physiological bile flow throughout the experiment. Quantitative histological and histochemical methods were used. Serial frontal cryostat sections cut from the posterior hypothalamus were used for detection of the activity of the following enzymes: monoamine oxidase B, succinate dehydrogenase, NADH dehydrogenase, NADPH dehydrogenase, glucose-6-phosphate dehydrogenase, lactate dehydrogenase, and acid phosphatase. Morphological studies of histaminergic neurons were performed on preparations stained with thionine. These studies showed that complete external drainage of bile led to transient size reductions and rounding of cell perikarya. Metabolic changes were seen within a day of bile loss and subsequently progressed. All energy metabolic pathways were suppressed and acid phosphatase activity was increased on day 5.


Subject(s)
Bile/physiology , Histamine/metabolism , Hypothalamus, Posterior/cytology , Hypothalamus, Posterior/enzymology , Neurons/cytology , Neurons/enzymology , Animals , Bile Ducts , Male , Rats , Rats, Wistar
17.
J Neurosci ; 26(4): 1088-97, 2006 Jan 25.
Article in English | MEDLINE | ID: mdl-16436594

ABSTRACT

The central histaminergic neuron system inhibits epileptic seizures, which is suggested to occur mainly through histamine 1 (H1) and histamine 3 (H3) receptors. However, the importance of histaminergic neurons in seizure-induced cell damage is poorly known. In this study, we used an organotypic coculture system and confocal microscopy to examine whether histaminergic neurons, which were verified by immunohistochemistry, have any protective effect on kainic acid (KA)-induced neuronal damage in the developing hippocampus. Fluoro-Jade B, a specific marker for degenerating neurons, indicated that, after the 12 h KA (5 microM) treatment, neuronal damage was significantly attenuated in the hippocampus cultured together with the posterior hypothalamic slice containing histaminergic neurons [HI plus HY (POST)] when compared with the hippocampus cultured alone (HI) or with the anterior hypothalamus devoid of histaminergic neurons. Moreover, alpha-fluoromethylhistidine, an inhibitor of histamine synthesis, eliminated the neuroprotective effect in KA-treated HI plus HY (POST), and extracellularly applied histamine (1 nM to 100 microM) significantly attenuated neuronal damage only at 1 nM concentration in HI. After the 6 h KA treatment, spontaneous electrical activity registered in the CA1 subregion contained significantly less burst activity in HI plus HY (POST) than in HI. Finally, in KA-treated slices, the H3 receptor antagonist thioperamide enhanced the neuroprotective effect of histaminergic neurons, whereas the H1 receptor antagonists triprolidine and mepyramine dose-dependently decreased the neuroprotection in HI plus HY (POST). Our results suggest that histaminergic neurons protect the developing hippocampus from KA-induced neuronal damage, with regulation of neuronal survival being at least partly mediated through H1 and H3 receptors.


Subject(s)
Convulsants/toxicity , Hippocampus/drug effects , Histamine/pharmacology , Kainic Acid/toxicity , Neurons/physiology , Neuroprotective Agents/pharmacology , Animals , Cell Death/drug effects , Cells, Cultured/drug effects , Cells, Cultured/physiology , Coculture Techniques , Hippocampus/cytology , Histamine/biosynthesis , Histamine/physiology , Histamine Antagonists/pharmacology , Histamine H1 Antagonists/pharmacology , Hypothalamus, Anterior/cytology , Hypothalamus, Posterior/cytology , Imidazoles/pharmacology , Methylhistidines/pharmacology , Microscopy, Confocal , Organ Culture Techniques , Piperidines/pharmacology , Pyrilamine/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Histamine H1/drug effects , Receptors, Histamine H1/physiology , Receptors, Histamine H3/drug effects , Receptors, Histamine H3/physiology , Thiourea/analogs & derivatives , Thiourea/pharmacology , Triprolidine/pharmacology
18.
Nat Commun ; 8(1): 1405, 2017 11 10.
Article in English | MEDLINE | ID: mdl-29123082

ABSTRACT

Basic and clinical observations suggest that the caudal hypothalamus comprises a key node of the ascending arousal system, but the cell types underlying this are not fully understood. Here we report that glutamate-releasing neurons of the supramammillary region (SuMvglut2) produce sustained behavioral and EEG arousal when chemogenetically activated. This effect is nearly abolished following selective genetic disruption of glutamate release from SuMvglut2 neurons. Inhibition of SuMvglut2 neurons decreases and fragments wake, also suppressing theta and gamma frequency EEG activity. SuMvglut2 neurons include a subpopulation containing both glutamate and GABA (SuMvgat/vglut2) and another also expressing nitric oxide synthase (SuMNos1/Vglut2). Activation of SuMvgat/vglut2 neurons produces minimal wake and optogenetic stimulation of SuMvgat/vglut2 terminals elicits monosynaptic release of both glutamate and GABA onto dentate granule cells. Activation of SuMNos1/Vglut2 neurons potently drives wakefulness, whereas inhibition reduces REM sleep theta activity. These results identify SuMvglut2 neurons as a key node of the wake-sleep regulatory system.


Subject(s)
Arousal/physiology , Glutamic Acid/physiology , Hypothalamus, Posterior/physiology , Neurons/physiology , Animals , Hypothalamus, Posterior/cytology , Male , Mice , Mice, Knockout , Mice, Transgenic , Nitric Oxide Synthase Type I/physiology , Sleep, REM/physiology , Theta Rhythm/physiology , Vesicular Glutamate Transport Protein 2/deficiency , Vesicular Glutamate Transport Protein 2/genetics , Vesicular Glutamate Transport Protein 2/physiology , Vesicular Inhibitory Amino Acid Transport Proteins/physiology , Wakefulness/physiology
19.
Brain Struct Funct ; 222(3): 1495-1507, 2017 04.
Article in English | MEDLINE | ID: mdl-27539452

ABSTRACT

We recently demonstrated that granule cells located in the dorsal dentate gyrus (dDG) are activated by neurons located in the lateral supramammillary nucleus (SumL) during paradoxical sleep (PS) hypersomnia. To determine whether these neurons are glutamatergic and/or GABAergic, we combined FOS immunostaining with in situ hybridization of vesicular glutamate transporter 2 (vGLUT2, a marker of glutamatergic neurons) or that of the vesicular GABA transporter (vGAT, a marker of GABAergic neurons) mRNA in rats displaying PS hypersomnia (PSR). We found that 84 and 76 % of the FOS+ SumL neurons in PSR rats expressed vGLUT2 and vGAT mRNA, respectively. Then, we examined vGLUT2 and FOS immunostaining in the dorsal and ventral DG of PSR rats with a neurochemical lesion of the Sum. In PSR-lesioned animals but not in sham animals, nearly all vGLUT2+ fibers and FOS+ neurons disappeared in the dDG, but not in the ventral DG (vDG). To identify the pathway (s) responsible (s) for the activation of the vDG during PS hypersomnia, we combined Fluorogold (FG) injection in the vDG of PSR rats with FOS staining. We found a large number of neurons FOS-FG+, specifically in the medial entorhinal cortex (ENTm). Altogether, our results suggest that SumL neurons with a unique dual glutamatergic and GABAergic phenotype are responsible for the activation of the dDG during PS hypersomnia, while vDG granule neurons are activated by ENTm cortical neurons. These results suggest differential mechanisms and functions for the activation of the dDG and the vDG granule cells during PS.


Subject(s)
Dentate Gyrus/cytology , Neurons/physiology , Sleep, REM/physiology , Animals , Cell Count , Dentate Gyrus/injuries , Electroencephalography , Electromyography , Hypothalamus, Posterior/cytology , Male , Oncogene Proteins v-fos/genetics , Oncogene Proteins v-fos/metabolism , Phosphopyruvate Hydratase/metabolism , Rats , Rats, Sprague-Dawley , Sleep Deprivation , Statistics, Nonparametric , Stilbamidines/metabolism , Vesicular Glutamate Transport Protein 2/genetics , Vesicular Glutamate Transport Protein 2/metabolism , Wakefulness
SELECTION OF CITATIONS
SEARCH DETAIL