Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
J Chem Neuroanat ; 116: 101997, 2021 10.
Article in English | MEDLINE | ID: mdl-34182088

ABSTRACT

In the last few years we assist to an unexpected deluge of genomic data on hypothalamic development and structure. Perhaps most surprisingly, the Lateral Zone has received much attention too. The new information focuses first of all on transcriptional heterogeneity. Many already known and a number of hitherto unknown lateral hypothalamic neurons have been described to an enormous degree of detail. Maybe the most surprising novel discoveries are two: First, some restricted regions of the embryonic forebrain neuroepithelium generate specific LHA neurons, either GABAergic or glutamatergic. Second, evidence is mounting that supports the existence of numerous kinds of "bilingual" lateral hypothalamic neurons, expressing (and releasing) glutamate and GABA both as well as assorted neuropeptides. This is not accepted by all, and it could be that genomic researchers need a common set of rules to interpret their data (sensitivity, significance, age of analysis). In any case, some of the new results appear to confirm hypotheses about the ability of the hypothalamus and in particular its Lateral Zone to achieve physiological flexibility on a fixed connectivity ("biochemical switching"). Furthermore, the results succinctly reviewed here are the basis for future advances, since the transcriptional databases generated can now be mined e.g. for adhesion genes, to figure out the causes of the peculiar histology of the Lateral Zone; or for ion channel genes, to clarify present and future electrophysiological data. And with the specific expression data about small subpopulations of neurons, their connections can now be specifically labeled, revealing novel relations with functional significance.


Subject(s)
GABAergic Neurons/chemistry , GABAergic Neurons/metabolism , Glutamic Acid/metabolism , Hypothalamic Area, Lateral/growth & development , Hypothalamic Area, Lateral/metabolism , Neurogenesis/physiology , Animals , Glutamic Acid/analysis , Humans , Hypothalamic Area, Lateral/chemistry , Transcription Factors/analysis , Transcription Factors/biosynthesis
2.
Neuropharmacology ; 178: 108239, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32771529

ABSTRACT

The glutamatergic lateral hypothalamus (LH) has been implicated in a variety of behaviors, such as evasion and feeding, while its role in defensive behaviors and relevant neurocircuits remains unclear. Here, we demonstrated that the glutamatergic LH is a critical structure regulating defensive behaviors. Trimethylthiazole (TMT), the odor of mice predator, significantly increased c-Fos expression in the LH. Using fiber photometry technology, we found that TMT exposure increased the activity of LH glutamatergic neurons. Selective activation of LH glutamatergic neurons with optogenetics and chemogenetics promoted a series of defense-related behaviors, including fleeing, avoidance, and hiding, while selective inhibition of LH glutamatergic neurons suppressed the avoidance provoked by TMT. Activation of both the glutamatergic LH terminals in the hypothalamic paraventricular nucleus (PVN) and the glutamatergic projection from the basolateral amygdala (BLA) to the LH elicited defensive behaviors. Finally, by combining the viral-mediated retrograde tracing with anterograde activation, we found that PVN-projecting glutamatergic neurons in the LH were activated by BLA glutamatergic inputs. Taken together, our results illustrate that the glutamatergic LH is a pivotal relay of defensive behaviors and possibly promotes these behaviors through the BLA→LH→PVN pathway.


Subject(s)
Avoidance Learning/physiology , Defense Mechanisms , Glutamic Acid/metabolism , Hypothalamic Area, Lateral/metabolism , Animals , Glutamic Acid/analysis , Hypothalamic Area, Lateral/chemistry , Male , Mice , Mice, Inbred C57BL , Optogenetics/methods
3.
Alcohol Clin Exp Res ; 40(2): 329-34, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26842251

ABSTRACT

BACKGROUND: The aim of this study was to examine possible differences in nicotinic acetylcholine receptors and responses in rats with genetic preference or avoidance for alcohol. This was done by using 2 rat lines with high alcohol preference (Alko Alcohol [AA]) or alcohol avoidance (Alko Non-Alcohol [ANA]). METHODS: Locomotor activity was measured following nicotine and histamine H3 receptor (H3R) antagonist treatment. In situ hybridization and receptor ligand binding experiments were used in drug-naïve animals to examine the expression of different α nicotinic receptor subunits. RESULTS: The AA rats were found to be more sensitive to the stimulatory effect of a low dose of nicotine than ANA rats, which were not significantly activated. Combination of histamine H3R antagonist, JNJ-39220675, and nicotine resulted to similar locomotor activation as nicotine alone. To further understand the mechanism underlying the difference in nicotine response in AA and ANA rats, we studied the expression of α5, α6, and α7 nicotinic receptor subunits in specific brain areas of AA and ANA rats. We found no differences in the expression of α5 nicotinic receptor subunits in the medial habenula and hippocampus or in α6 subunit in the ventral tegmental area and substantia nigra. However, the level of α7 nicotinic receptor subunit mRNA was significantly lower in the tuberomamillary nucleus of posterior hypothalamus of alcohol-preferring AA rats than in alcohol-avoiding ANA rats. Also the hypothalamic [125I-α-bungarotoxin binding was lower in AA rats indicating lower levels of α7 nicotinic receptors. CONCLUSIONS: The lower expression and receptor binding of α7 nicotinic receptors in the tuberomamillary nucleus of AA rats suggest a difference in the regulation of brain histamine neurons between the rat lines since the α7 nicotinic receptors are located in histaminergic neurons. Stronger nicotine-induced locomotor response, mediated partially via α7 receptors, and previously described high alcohol consumption in AA rats could be explained by the found difference in tuberomamillary α7 receptor levels.


Subject(s)
Alcohol Drinking/physiopathology , Hypothalamus/physiology , Nicotine/pharmacology , alpha7 Nicotinic Acetylcholine Receptor/physiology , Alcohol Abstinence , Animals , Azepines/pharmacology , Histamine Antagonists/pharmacology , Hypothalamic Area, Lateral/chemistry , Hypothalamic Area, Lateral/physiology , Hypothalamus/chemistry , In Situ Hybridization , Male , Motor Activity/drug effects , Nicotine/administration & dosage , Pyridines/pharmacology , Rats , alpha7 Nicotinic Acetylcholine Receptor/analysis , alpha7 Nicotinic Acetylcholine Receptor/antagonists & inhibitors , alpha7 Nicotinic Acetylcholine Receptor/metabolism
4.
Brain Res ; 1479: 31-43, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22902618

ABSTRACT

Melanin-concentrating hormone (MCH) is involved in the regulation of feeding behavior as well as in goal oriented behaviors, and MCH-containing neurons are distributed mainly in the lateral hypothalamic area (LHA). The anterior basomedial nucleus (BMA) and anterior cortical nucleus (CoA) of the amygdala form part of a circuit involved in processing olfactory, gustatory and visceral information, and the BMA-LHA and CoA-LHA pathways are suggested to be implicated in the control of feeding behavior. However, it is still unknown whether or not MCH-containing LHA neurons are under the direct influence of the BMA and CoA. Here the organization of projections from the BMA and CoA to MCH-containing LHA neurons was examined. Using a combined anterograde tracing with biotinylated dextranamine and immunohistochemistry for MCH, we first demonstrated that the distribution pattern of BMA fibers was almost similar to that of CoA fibers in the LHA, and a prominent overlapping distribution of these fibers and MCH-immunoreactive neurons existed in the ventral peripeduncular region of the LHA. We further revealed that asymmetrical synapses were made between these fibers and neurons. Using a combination of retrograde tract-tracing with cholera toxin B subunit and in situ hybridization for vesicular glutamate transporter (VGLUT) 2 mRNA, we finally showed that most of the LHA-projecting BMA and CoA neurons expressed VGLUT2 mRNA. These data suggest that the BMA and CoA of the amygdala may exert excitatory influence upon the MCH-containing LHA neurons for the regulation of feeding behavior.


Subject(s)
Amygdala/physiology , Hypothalamic Area, Lateral/physiology , Hypothalamic Hormones/physiology , Melanins/physiology , Neurons/physiology , Pituitary Hormones/physiology , Amygdala/chemistry , Amygdala/ultrastructure , Animals , Hypothalamic Area, Lateral/chemistry , Hypothalamic Area, Lateral/ultrastructure , Hypothalamic Hormones/analysis , Male , Melanins/analysis , Nerve Net/chemistry , Nerve Net/physiology , Nerve Net/ultrastructure , Neural Pathways/chemistry , Neural Pathways/physiology , Neural Pathways/ultrastructure , Neurons/chemistry , Neurons/ultrastructure , Pituitary Hormones/analysis , Rats , Rats, Wistar
5.
Endocrinology ; 151(5): 2106-16, 2010 May.
Article in English | MEDLINE | ID: mdl-20308532

ABSTRACT

It is widely accepted that leptin acts on first-order neurons in the arcuate nucleus (ARC) with information then relayed to other hypothalamic centers. However, the extent to which leptin mediates its central actions solely, or even primarily, via this route is unclear. We used a model of hypothalamo-pituitary disconnection (HPD) to determine whether leptin action on appetite-regulating systems requires the ARC. This surgical preparation eliminates the ARC. We measured effects of iv leptin to activate hypothalamic neurons (Fos labeling). In ARC-intact animals, leptin increased the percentage of Fos-positive melanocortin neurons and reduced percentages of Fos-positive neuropeptide Y neurons compared with saline-treated animals. HPD itself increased Fos labeling in the lateral hypothalamic area (LHA). Leptin influenced Fos labeling in the dorsomedial nucleus (DMH), ventromedial nucleus, and paraventricular nucleus (PVN) in HPD and normal animals, with effects on particular cell types varying. In the LHA and DMH, leptin decreased orexin cell activation in HPD and ARC-intact sheep. HPD abolished leptin-induced expression of Fos in melanin-concentrating hormone cells in the LHA and in CRH cells in the PVN. In contrast, HPD accentuated activation in oxytocin neurons. Our data from sheep with lesions encompassing the ARC do not suggest a primacy of action of leptin in this nucleus. We demonstrate that first order to second order signaling may not represent the predominant means by which leptin acts in the brain to generate integrated responses. We provide evidence that leptin exerts direct action on cells of the DMH, ventromedial nucleus, and PVN.


Subject(s)
Appetite/drug effects , Arcuate Nucleus of Hypothalamus/physiopathology , Hypothalamus/drug effects , Leptin/pharmacology , Animals , Appetite/physiology , Arcuate Nucleus of Hypothalamus/surgery , Dorsomedial Hypothalamic Nucleus/chemistry , Dorsomedial Hypothalamic Nucleus/cytology , Dorsomedial Hypothalamic Nucleus/drug effects , Female , Hypothalamic Area, Lateral/chemistry , Hypothalamic Area, Lateral/cytology , Hypothalamic Area, Lateral/drug effects , Hypothalamus/cytology , Hypothalamus/physiology , Immunohistochemistry , Injections, Intravenous , Leptin/administration & dosage , Neurons/chemistry , Neurons/cytology , Neurons/drug effects , Neuropeptides/analysis , Paraventricular Hypothalamic Nucleus/chemistry , Paraventricular Hypothalamic Nucleus/cytology , Paraventricular Hypothalamic Nucleus/drug effects , Pituitary Gland/surgery , Proto-Oncogene Proteins c-fos/analysis , Sheep , Ventromedial Hypothalamic Nucleus/chemistry , Ventromedial Hypothalamic Nucleus/cytology , Ventromedial Hypothalamic Nucleus/drug effects
6.
Peptides ; 29(10): 1732-9, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18577407

ABSTRACT

Neuropeptide Y (NPY) conjugated with a ribosomal inactivating toxin, saporin (SAP), is a toxin that targets NPY receptor-expressing cells. Injection of NPY-SAP into the rat arcuate nucleus (Arc) and basomedial hypothalamus (BMH) destroys two populations of NPY-receptor-expressing neurons important for the control of food intake and body weight, NPY and pro-opiomelanocortin (POMC) and cocaine and amphetamine related transcript (CART) neurons, and produces profound hyperphagia and obesity. Here, we investigated the contribution of lateral hypothalamus (LHA) orexigenic peptides, orexins and melanocortin concentrating hormone (MCH), to these lesion effects. We microinjected NPY-SAP into two sites on each side of the Arc, causing a loss of NPY and POMC/CART neurons that was limited to the Arc. Lesioned rats rapidly became hyperphagic and obese. However, MCH and prepro-orexin mRNA expression were not increased in the LHA in the lesioned rats, but were decreased at some levels of the LHA or were unchanged. NPY-SAP-induced obesity therefore differs from dietary obesity and from obesity associated with leptin or leptin receptor deficiency in which MCH gene expression is increased. The Arc NPY-SAP lesion produces obesity and hyperphagia that does not require overexpression of hypothalamic neuropeptides currently considered to provide major stimulatory drive for food intake: NPY, agouti gene-related protein, MCH or orexins. The source of the seemingly unregulated stimulatory drive for feeding in these animals has not been identified, but may be associated with hindbrain or endocrine mechanisms.


Subject(s)
Arcuate Nucleus of Hypothalamus/drug effects , Hyperphagia/chemically induced , Hypothalamic Area, Lateral/physiology , Intracellular Signaling Peptides and Proteins/genetics , Neuropeptide Y/chemistry , Neuropeptide Y/pharmacology , Neuropeptides/genetics , Obesity/chemically induced , Animals , Arcuate Nucleus of Hypothalamus/cytology , Arcuate Nucleus of Hypothalamus/metabolism , Body Weight/drug effects , Eating/drug effects , Female , Hypothalamic Area, Lateral/chemistry , Hypothalamic Area, Lateral/cytology , Hypothalamic Hormones/genetics , Hypothalamic Hormones/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Male , Melanins/genetics , Melanins/metabolism , Microinjections , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Neuropeptides/metabolism , Orexins , Pituitary Hormones/genetics , Pituitary Hormones/metabolism , Protein Precursors/genetics , Protein Precursors/metabolism , Rats , Rats, Sprague-Dawley , Up-Regulation
7.
J Endocrinol ; 191(1): 137-45, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17065397

ABSTRACT

In this study, we investigated the effects of acute morphine administration, chronic intermittent escalating-dose morphine administration and spontaneous withdrawal from chronic morphine on mRNA levels of mu opioid receptor (MOP-r), and the opioid peptides pro-opiomelanocortin (POMC) and preprodynorphin (ppDyn) in several key brain regions of the rat, associated with drug reward and motivated behaviors: lateral hypothalamus (lat.hyp), nucleus accumbens (NAc) core, amygdala, and caudate-putamen (CPu). There was no effect on MOP-r mRNA levels in these brain regions 30 min after either a single injection of morphine (10 mg/kg, i.p.) or chronic intermittent escalating-dose morphine (from 7.5 mg/kg per day on day 1 up to 120 mg/kg per day on day 10). Activation of the stress-responsive hypothalamic-pituitary-adrenal axis by 12 h withdrawal from chronic morphine was confirmed; both POMC mRNA levels in the anterior pituitary and plasma adrenocorticotropic hormone levels were significantly elevated. Under this withdrawal-related stress condition, there was an increase in MOP-r mRNA levels in the lat.hyp, NAc core, and CPu. Recent studies have demonstrated a novel role for the lat.hyp orexin (or hypocretin) activation in both drug-related positive rewarding, and withdrawal effects. Around 50% of lat.hyp orexin neurons express MOP-r. Therefore, we also examined the levels of lat.hyp orexin mRNA, and found them increased in morphine withdrawal, whereas there was no change in levels of the lat.hyp ppDyn mRNA, a gene coexpressed with the lat.hyp orexin. Our results show that there is an increase in MOP-r gene expression in a region-specific manner during morphine withdrawal, and support the hypothesis that increased lat.hyp orexin activity plays a role in morphine-withdrawal-related behaviors.


Subject(s)
Hypothalamic Area, Lateral/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Morphine/adverse effects , Neostriatum/chemistry , Neuropeptides/genetics , RNA, Messenger/analysis , Receptors, Opioid, mu/genetics , Adrenocorticotropic Hormone/blood , Animals , Corticosterone/blood , Gene Expression , Hypothalamic Area, Lateral/metabolism , Male , Morphine/pharmacology , Neostriatum/metabolism , Orexins , Pituitary Gland, Anterior/chemistry , Pituitary Gland, Anterior/metabolism , Rats , Rats, Inbred F344 , Receptors, Corticotropin-Releasing Hormone/genetics , Substance Withdrawal Syndrome/metabolism , Testosterone/blood
8.
Peptides ; 27(5): 1054-72, 2006 May.
Article in English | MEDLINE | ID: mdl-16487629

ABSTRACT

The neuropeptide FF (NPFF) is an octapeptide of the RFamide-related peptides (FaRPs) that was primarily isolated from the bovine brain. Its distribution in the CNS has been reported in several mammalian species, as well as in some amphibians. Therefore, in order to gain insight in the evolution on the expression pattern of this neuropeptide in vertebrates, we carried out an immunohistochemical study in the sea lamprey, Petromyzon marinus. The distribution of NPFF-like-immunoreactive (NPFF-ir) structures in the lamprey brain is, in general, comparable to that previously described in other vertebrate species. In lamprey, most of the NPFF-ir cells were found in the hypothalamus, particularly in two large populations, the bed nucleus of the tract of the postoptic commissure and the tuberomammillary area. Numerous NPFF-ir cells were also observed in the rostral rhombencephalon, including a population in the dorsal isthmic gray and the reticular formation. Additional labeled neurons were found inside the preoptic region, the parapineal vesicle, the periventricular mesencephalic tegmentum, the descending trigeminal tract, the nucleus of the solitary tract, as well as in the gray matter of the spinal cord. The NPFF-ir fibers were widely distributed in the brain and the spinal cord, being, in general, more concentrated throughout the basal plate. The presence of NPFF-ir fibers in the lamprey neurohypophysis suggests that the involvement of NPFF-like substances in the hypothalamo-hypophyseal system had emerged early during evolution.


Subject(s)
Central Nervous System/chemistry , Oligopeptides/analysis , Receptors, Catecholamine/analysis , Animals , Female , Hypothalamic Area, Lateral/chemistry , Hypothalamus/chemistry , Immunohistochemistry , Lampreys , Male , Tyrosine 3-Monooxygenase/analysis
9.
Brain Res ; 1020(1-2): 118-29, 2004 Sep 10.
Article in English | MEDLINE | ID: mdl-15312793

ABSTRACT

The posterior lateral hypothalamus (PLH) has long been considered crucial to normal wakefulness while the ventral part of the oral pontine reticular nucleus (vRPO) is involved in the generation and maintenance of rapid eye movement (REM) sleep. However, to date, there is no information on the ultrastructure or neurotransmitter content of the hypothalamo-reticular projection. In the present study, we examined the morphology and synaptic organization of PLH terminals in the vRPO using PLH injections of biotinylated dextran amine (BDA) as well as of wheat germ agglutinin conjugated to horseradish peroxidase (WGA-HRP). Since some PLH neurons are GABAergic, we used a post-embedding immunogold technique to determine whether any anterogradely labeled terminals were GABA-immunopositive. Electron microscope analyses revealed a variety of ultrastructural features in the vRPO anterogradely labeled terminals. Although most labeled terminals (over 63%) formed symmetric synapses on vRPO somata and dendrites, others made asymmetric synapses on vRPO dendrites. The relative percentages of labeled terminals observed on large, medium and small diameter dendrites were 44.3 +/- 5.5%, 35.3 +/- 3.0% and 20.4 +/- 3.1%, respectively. Finally, post-embedding immunogold technique revealed that there are GABA-immunopositive and immunonegative components to this projection, indicating that GABA is one of the transmitters used by the PLH cells that project to the vRPO. Furthermore, most, if not all, of the GABA-labeled axon terminals formed symmetric synapsis. In conclusion, our results suggest that the PLH could modulate the physiological responses of vRPO neurons through a GABAergic pathway as well as by other inhibitory and/or excitatory pathways. Activation of the descending PLH GABAergic projection may inhibit the REM sleep-inducing neurons within the vRPO and thus contribute to the suppression of REM sleep activation during wakefulness.


Subject(s)
Brain Mapping , Hypothalamic Area, Lateral/cytology , Presynaptic Terminals/ultrastructure , Reticular Formation/cytology , gamma-Aminobutyric Acid/analysis , Animals , Cats , Hypothalamic Area, Lateral/chemistry , Hypothalamic Area, Lateral/physiology , Neural Pathways/cytology , Neural Pathways/physiology , Reticular Formation/chemistry , Reticular Formation/physiology , Sleep, REM/physiology , Wakefulness/physiology , gamma-Aminobutyric Acid/physiology
10.
Brain Res ; 1004(1-2): 91-7, 2004 Apr 09.
Article in English | MEDLINE | ID: mdl-15033423

ABSTRACT

Sleep deprivation increases sleep propensity in rats and mice as well as the production of several sleep-regulatory substances. Nuclear factor kappa B (NF-kappa B) is a transcription factor implicated in the activation of many of these sleep-promoting substances. A unique population of neurons immunoreactive for the p65 subunit of NF-kappa B was previously localized within the caudal dorsolateral hypothalamus of rats. Therefore, we evaluated the effect of sleep deprivation on NF-kappa Bp65-immunoreactivity (IR) in cells of this region in rats as well as its nuclear translocation in a kappa B-lacZ transgenic mouse line. In rats after 6 h of sleep deprivation beginning at light onset, the number of neurons with NF-kappa Bp65-IR increased significantly in the caudal lateral hypothalamus, specifically the magnocellular lateral hypothalamus adjacent to the subthalamus. Sleep deprivation also significantly increased the number of cells expressing NF-kappa B-dependent beta-galactosidase in the magnocellular lateral hypothalamus, zona incerta dorsal, as well as the adjacent subthalamus in the transgenic mice. These results suggest that NF-kappa B expressing cells within the lateral hypothalamus may be important in the maintenance of the sleep-wake cycle.


Subject(s)
Hypothalamic Area, Lateral/metabolism , NF-kappa B/metabolism , Sleep Deprivation/metabolism , Active Transport, Cell Nucleus , Animals , Cell Count , Cell Nucleus/metabolism , Hypothalamic Area, Lateral/chemistry , Hypothalamic Area, Lateral/cytology , Male , Mice , Mice, Transgenic , Neurons/cytology , Neurons/enzymology , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Transcription Factor RelA , beta-Galactosidase/biosynthesis
11.
Endocrinology ; 145(5): 2507-15, 2004 May.
Article in English | MEDLINE | ID: mdl-14962992

ABSTRACT

Pancreatic glucokinase (GK)-like immunoreactivities are located in ependymocytes and serotonergic neurons of the rat brain. The present study investigated in vitro changes in intracellular calcium concentrations ([Ca(2+)](i)) in response to low (2 mm) or high (20 mm) extracellular glucose concentrations in isolated cells from the wall of the central canal (CC), raphe obscurus nucleus (ROb), ventromedial hypothalamus (VMH), and lateral hypothalamic area (LHA) in male rats. An increase in [Ca(2+)](i) was found in cells from the CC (21.1% or 9.8% of ependymocytes), ROb (10.9% or 14.5% of serotonergic neurons), VMH (7.8% and 25.2% of neurons), and LHA (20% or 15.7% of neurons), when extracellular glucose levels were changed from 10 to either 2 or 20 mm, respectively. Most of the ependymocytes and serotonergic neurons responding to the glucose changes were immunoreactive to the anti-GK in the CC (96.8% for low glucose and 100% for high glucose) and ROb (100% for low and high glucose). The [Ca(2+)](i) increase was blocked with calcium-free medium or L-type calcium channel blocker. Cells with an increase in [Ca(2+)](i) in response to low glucose did not respond to high glucose and vice versa. Inhibition of GK activity with acute alloxan treatment blocked low or high glucose-induced [Ca(2+)](i) increases in most GK-immunoreactive cells from the CC or ROb. The glucose-sensitive [Ca(2+)](i) increase in neurons of the VMH and LHA was also alloxan-sensitive, but no cells taken from the VMH and LHA were immunoreactive to the antibody used. The present study further indicates that ependymocytes of the CC and serotonergic neurons in the ROb are also sensitive to the changes in extracellular glucose in a GK-dependent manner, but that the subtype of GK in these cells could be different from that in the VMH and LHA.


Subject(s)
Brain Stem/chemistry , Calcium/analysis , Ependyma/chemistry , Glucose/analysis , Neurons/chemistry , Serotonin/physiology , Alloxan/pharmacology , Animals , Brain Stem/cytology , Brain Stem/enzymology , Calcium Channel Blockers/pharmacology , Enzyme Inhibitors/pharmacology , Glucokinase/analysis , Glucokinase/antagonists & inhibitors , Glucose/administration & dosage , Hypothalamic Area, Lateral/chemistry , Male , Nifedipine/pharmacology , Raphe Nuclei/chemistry , Rats , Rats, Wistar , Serotonin/analysis , Ventromedial Hypothalamic Nucleus/chemistry
12.
Neuroscience ; 122(2): 541-50, 2003.
Article in English | MEDLINE | ID: mdl-14614918

ABSTRACT

The viral transneuronal labeling method was used to demonstrate that orexin-containing neurons of the lateral hypothalamic area (LHA) are linked via multisynaptic connections to different sympathetic outflow systems. Two different types of transneuronal tracing experiments were performed: single- and double-virus studies. In the first series of experiments, Bartha pseudorabies virus (PRV), a retrograde transneuronal tracer, was injected into single sympathetic targets, viz., stellate ganglion, adrenal gland, celiac ganglion, and kidney. Six to 7 days post-injection, orexin (hypocretin) neurons were transneuronally labeled. In a second set of experiments, the double-virus tracing method was used to determine whether single orexin LHA neurons are linked to two different sympathetic outflow systems. Two isogenic forms of Bartha PRV were used that differed by a single gene. beta-Galactosidase Bartha PRV was injected into the stellate ganglion and green fluorescent protein Bartha PRV into the adrenal gland of the same rat. The reverse placement of viral injections was made in another set of rats. In both paradigms, some orexin LHA neurons were transneuronally labeled with both viruses, indicating that they are capable of modulating multiple sympathetic outflow systems. These findings raise the possibility that orexin LHA neurons regulate general sympathetic functions, such as those that occur during arousal or the fight-or-flight response.


Subject(s)
Adrenergic Fibers/physiology , Carrier Proteins/physiology , Hypothalamic Area, Lateral/physiology , Intracellular Signaling Peptides and Proteins , Neurons/physiology , Neuropeptides/physiology , Neurotransmitter Agents/physiology , Adrenergic Fibers/chemistry , Animals , Carrier Proteins/analysis , Hypothalamic Area, Lateral/chemistry , Male , Neural Pathways/chemistry , Neural Pathways/physiology , Neurons/chemistry , Neuropeptides/analysis , Orexins , Rats , Rats, Sprague-Dawley
13.
J Neuroendocrinol ; 15(1): 1-14, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12535164

ABSTRACT

Gamma-aminobutyric acid (GABA) interacts with hypothalamic neuronal pathways regulating feeding behaviour. GABA has been reported to stimulate feeding via both ionotropic GABA(A) and metabotropic GABA(B) receptors. The functional form of the GABA(B) receptor is a heterodimer consisting of GABA(B) receptor-1 (GABA(B)R1) and GABA(B) receptor-2 (GABA(B)R2) proteins. Within the heterodimer, the GABA-binding site is localized to GABA(B)R1. In the present study, we used an antiserum to the GABA(B)R1 protein in order to investigate the cellular localization of GABA(B)R1-immunoreactive neurones in discrete hypothalamic regions implicated in the control of body weight. The colocalization of GABA(B)R1 immunoreactivity with different chemical messengers that regulate food intake was analysed. GABA(B)R1-immunoreactive cell bodies were found in the periventricular, paraventricular (PVN), supraoptic, arcuate, ventromedial hypothalamic, dorsomedial hypothalamic, tuberomammillary nuclei and lateral hypothalamic area (LHA). Direct double-labelling showed that glutamic acid decarboxylase (GAD)-positive terminals were in close contact with GABA(B)R1-containing cell bodies located in all these regions. In the ventromedial part of the arcuate nucleus, GABA(B)R1-immunoreactive cell bodies were found to contain neuropeptide Y, agouti-related peptide (AGRP) and GAD. In the ventrolateral part of the arcuate nucleus, GABA(B)R1-immunoreactive cell bodies were shown to contain pro-opiomelanocortin and cocaine- and amphetamine-regulated transcript. In the LHA, GABA(B)R1 immunoreactivity was present in both melanin-concentrating hormone- and orexin-containing cell populations. In the tuberomammillary nucleus, GABA(B)R1-immunoreactive cell bodies expressed histidine decarboxylase, a marker for histamine-containing neurones. In addition, GAD and AGRP were found to be colocalized in some nerve terminals surrounding GABA(B)R1-immunoreactive cell bodies in the parvocellular part of the PVN. The results may provide a morphological basis for the understanding of how GABA regulates the hypothalamic control of food intake and body weight via GABA(B) receptors.


Subject(s)
Feeding Behavior/physiology , Hypothalamus/chemistry , Neurons/chemistry , Receptors, GABA-B/analysis , Animals , Antibodies , Arcuate Nucleus of Hypothalamus/chemistry , Arcuate Nucleus of Hypothalamus/physiology , Body Weight/physiology , Dorsomedial Hypothalamic Nucleus/chemistry , Dorsomedial Hypothalamic Nucleus/physiology , Hypothalamic Area, Lateral/chemistry , Hypothalamic Area, Lateral/physiology , Hypothalamus/physiology , Hypothalamus, Anterior/chemistry , Hypothalamus, Anterior/physiology , Male , Paraventricular Hypothalamic Nucleus/chemistry , Paraventricular Hypothalamic Nucleus/physiology , Rats , Rats, Sprague-Dawley , Receptors, GABA-B/immunology , Ventromedial Hypothalamic Nucleus/chemistry , Ventromedial Hypothalamic Nucleus/physiology , gamma-Aminobutyric Acid/physiology
14.
Brain Res ; 955(1-2): 1-7, 2002 Nov 15.
Article in English | MEDLINE | ID: mdl-12419515

ABSTRACT

Orexin (ORX) A and B (hypocretins) are excitatory neuropeptides produced by neurons of the lateral hypothalamus that have been implicated in the regulation of the sleep-wake cycle. In rats, Fos (the product of the cfos gene) expression shows daily rhythms in areas involved in sleep and wakefulness and orexinergic neurons show elevated Fos expression during the night. The present study directly compared the daily pattern of Fos expression in orexinergic neurons in diurnal (A. niloticus; grass rats) and nocturnal (R. norvegicus; lab rats) rodents. Animals kept on a 12:12 light-dark cycle were perfused at six different Zeitgeber times (ZT), with lights on at ZT 0: 1, 5, 13, 17, 20 and 23. In both nocturnal and diurnal rodents orexinergic neurons showed rhythms in Fos expression, with more Fos seen during the active phase of each species. In the diurnal species, Fos expression in cells of the lateral hypothalamus that do not produce ORX was elevated at ZT 20, a time when these animals sleep, and was low at ZT 13, a time of peak of activity. These results provide further evidence for a link between activity in orexinergic neurons and wakefulness and that in grass rats, other neurons of the lateral hypothalamus may work in an antagonistic fashion with respect to orexinergic neurons to regulate wakefulness in this diurnal species.


Subject(s)
Carrier Proteins/biosynthesis , Circadian Rhythm/physiology , Intracellular Signaling Peptides and Proteins , Neurons/metabolism , Neuropeptides/biosynthesis , Animals , Carrier Proteins/analysis , Hypothalamic Area, Lateral/chemistry , Hypothalamic Area, Lateral/metabolism , Male , Muridae , Neurons/chemistry , Neuropeptides/analysis , Orexins , Proto-Oncogene Proteins c-fos/analysis , Proto-Oncogene Proteins c-fos/biosynthesis , Rats , Rats, Sprague-Dawley
15.
J Comp Neurol ; 449(1): 43-64, 2002 Jul 15.
Article in English | MEDLINE | ID: mdl-12115692

ABSTRACT

The neuroanatomic connections of the inferior lobe and the lateral torus of the percomorph Hemichromis lifalili were investigated by 1,1', dioctadecyl-3,3,3',3'-tetramethylindo-carbocyanine perchlorate (DiI) tracing. The inferior lobe and the lateral torus both receive afferents from the secondary gustatory nucleus. Additional afferents reach the inferior lobe from the nucleus glomerulosus, nucleus suprachiasmaticus, dorsal and central posterior thalamic nucleus, nucleus lateralis valvulae, magnocellular part of the magnocellular nucleus of the preoptic region, caudal nucleus of the preglomerular region, posterior tuberal nucleus, area dorsalis of the telencephalon, and a tegmental nucleus (T2). Efferents from the inferior lobe and the lateral torus terminate in the dorsal hypothalamic neuropil and corpus mamillare. Furthermore, the inferior lobe projects to the medial nucleus of the lateral tuberal hypothalamus and perhaps makes axo-axonal synapses in the tractus tectobulbaris rectus. The inferior lobe and the torus lateralis have reciprocal connections with the preglomerular tertiary gustatory nucleus and posterior thalamic nucleus and are also mutually interconnected. The inferior lobe is also reciprocally connected with the medial nucleus of the preglomerular region, reticular formation and sparsely with the anterior dorsal thalamic and the ventromedial thalamic nuclei. Thus, whereas the lateral torus is exclusively connected with the gustatory system, the inferior lobe is of a multisensory nature. In comparison with the goldfish (Carassius auratus), the connectivity pattern of the inferior lobe of Hemichromis lifalili reflects its specialization with respect to the visual system, as it receives qualitative (i.e., dorsal posterior, anterior, and ventromedial thalamic nuclei) as well as quantitative (i.e., nucleus glomerulosus) additional visual input.


Subject(s)
Cichlids/physiology , Hypothalamus/physiology , Animals , Axonal Transport/physiology , Choline O-Acetyltransferase/analysis , Female , Hypothalamic Area, Lateral/anatomy & histology , Hypothalamic Area, Lateral/chemistry , Hypothalamic Area, Lateral/physiology , Hypothalamus/anatomy & histology , Hypothalamus/chemistry , Hypothalamus, Posterior/anatomy & histology , Hypothalamus, Posterior/chemistry , Hypothalamus, Posterior/physiology , Male , Neural Pathways/anatomy & histology , Neural Pathways/chemistry , Neural Pathways/physiology , Olfactory Pathways/anatomy & histology , Olfactory Pathways/chemistry , Olfactory Pathways/physiology , Taste/physiology
16.
Rev Neurol (Paris) ; 158(2): 155-65, 2002 Feb.
Article in French | MEDLINE | ID: mdl-11965171

ABSTRACT

Argyrophilic grain disease (AgD) is a late-onset dementia morphologically characterized by abundant neuropil grains (ArGs). ArGs are mainly found in the CA1 subfield of the cornu ammonis, entorhinal and transentorhinal cortices, the amygdala and the hypothalamic lateral tuberal nuclei. We have recently shown that abnormally phosphosphorylated tau protein is the main protein constituent of ArGs and that tau is hyperphosphorylated in up to 80p.100 of nerve cels in areas rich in ArGs. We could demonstrate that at least a subset of grains are formed within dendrites and dendritic side-branches of neurons containing hyperphosphylated tau. Morphology of dendrites containing grains suggests that a process of progressive dendritic shrinkage is taking place in neurons bearing ArGs. Furthermore it became apparent that the presence of ArGs is not necessarily associated with a cognitive decline. Our studies on AgD cases with and without dementia suggest that AgD is a progressive neurodegenerative disorder with early subclinical lesions in anterior part of the hippocampal formation. At later stages involvement of more caudal parts of the hippocampal formation generally results in a cognitive decline. Thus, one possible explanation for the dementia observed in some subjects with AgD is that there is a more widepread loss of postsynaptic structures, including synaptic contacts, throughout the hippocampus-entorhinal/parahippocampal complex and the amygdaloid nuclei. Most of the reported AgD cases are associated with neurofibrillary lesions (e.g. neurofibrillary tangles) which are also typical of Alzheimer's disease (AD). However, neurofibrillary changes do not exceed early (entorhinal and limbic) Braak stages which generally are not associated with a cognitive decline. Additional neuropathological features of AgD include oligodendroglial tau filamentous inclusions ( coiled bodies ), ballooned neurons and astrocytic tau pathology. The clinical features of AgD are poorly understood. However, preliminary data from retrospective studies suggest that in AgD behavioural disturbances will precede memory failure and memory decline. Furthermore, it has been shown that the ApoEe4 allele does not constitute a risk factor for the development of AgD. In conclusion it seems very likely that AgD is a distinct dementing disorder of old age that has to be distinguished from other tauopathies, e.g. AD, by both morphological and genetic criteria.


Subject(s)
Cytoplasmic Granules/ultrastructure , Dementia/pathology , Neuropil/ultrastructure , Age Factors , Aged , Alleles , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amygdala/chemistry , Amygdala/ultrastructure , Apolipoproteins E/genetics , Astrocytes/chemistry , Astrocytes/ultrastructure , Cognition Disorders/etiology , Coiled Bodies/chemistry , Coiled Bodies/ultrastructure , Cytoplasmic Granules/chemistry , Dementia/epidemiology , Dementia/genetics , Dementia/metabolism , Dendrites/chemistry , Dendrites/ultrastructure , Entorhinal Cortex/chemistry , Entorhinal Cortex/ultrastructure , Hippocampus/chemistry , Hippocampus/ultrastructure , Humans , Hypothalamic Area, Lateral/chemistry , Hypothalamic Area, Lateral/ultrastructure , Memory Disorders/etiology , Nerve Tissue Proteins/analysis , Neurodegenerative Diseases/classification , Neurons/chemistry , Neurons/ultrastructure , Neuropil/chemistry , Oligodendroglia/chemistry , Oligodendroglia/ultrastructure , Phosphorylation , Protein Processing, Post-Translational , Risk Factors , Silver Staining , tau Proteins/analysis
17.
Regul Pept ; 104(1-3): 21-5, 2002 Mar 15.
Article in English | MEDLINE | ID: mdl-11830272

ABSTRACT

Orexins (hypocretins) and the melanin-concentrating hormone (MCH) are neuropeptides localized to the lateral hypothalamic area and are potential regulators of energy homeostasis. Using highly sensitive radioimmunoassay for orexins and MCH, we determined their contents in the lateral hypothalamus (LH) of genetically obese ob/ob and db/db mice and their controls, C57BL/6J and C57BL/KSJ. The orexin contents in the lateral hypothalamus significantly increased in the ob/ob mice, whereas the orexin contents significantly decreased in the db/db mice. Mature orexin-A and -B peptides were the major endogenous orexin molecules in the lateral hypothalamus. Conversely, the MCH contents in the lateral hypothalamus of both obese mice increased compared to the control mice. MCH contents in the lateral hypothalamus were two- to five-fold higher than that of orexin contents. These results suggest that the regulatory mechanism of orexin and MCH may be different in the genetically obese mice.


Subject(s)
Blood Glucose/analysis , Carrier Proteins/metabolism , Hypothalamic Area, Lateral/metabolism , Hypothalamic Hormones/metabolism , Intracellular Signaling Peptides and Proteins , Melanins/metabolism , Neuropeptides/metabolism , Obesity/metabolism , Pituitary Hormones/metabolism , Animals , Body Weight , Brain Chemistry/physiology , Carrier Proteins/analysis , Chromatography, High Pressure Liquid , Hypothalamic Area, Lateral/chemistry , Hypothalamic Hormones/analysis , Male , Melanins/analysis , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Neuropeptides/analysis , Obesity/genetics , Orexins , Pituitary Hormones/analysis
18.
J Neurosci ; 21(23): 9273-9, 2001 Dec 01.
Article in English | MEDLINE | ID: mdl-11717361

ABSTRACT

The hypothalamic orexin (hypocretin) neuropeptides are associated with the regulation of sleep and feeding, and disturbances in orexinergic neurotransmission lead to a narcoleptic phenotype. Histamine has also been shown to play a role in the regulation of sleep and feeding. Therefore, we studied the relationship between the orexin and histamine systems of the CNS using electrophysiology, immunocytochemistry, and the reverse transcriptase (RT)-PCR method. Both orexin-A and orexin-B depolarized the histaminergic tuberomammillary neurons and increased their firing rate via an action on postsynaptic receptors. The depolarization was associated with a small decrease in input resistance and was likely caused by activation of both the electrogenic Na(+)/Ca(2+) exchanger and a Ca(2+) current. In a single-cell RT-PCR study using primers for the two orexin receptors, we found that most tuberomammillary neurons express both receptors and that the expression of the orexin-2 receptor is stronger than that of the orexin-1 receptor. Immunocytochemical studies show that the histamine and orexin neurons are often located very close to each other. The contacts between these two types of neurons seem to be reciprocal, because the orexin neurons are heavily innervated by histaminergic axons. These results suggest a functional connection between the two populations of hypothalamic neurons and that they may cooperate in the regulation of rapid-eye-movement sleep and feeding.


Subject(s)
Carrier Proteins/pharmacology , Histamine/metabolism , Hypothalamic Area, Lateral/drug effects , Intracellular Signaling Peptides and Proteins , Neurons/drug effects , Neuropeptides/pharmacology , Animals , Calcium Channel Blockers/pharmacology , Cell Separation , Dose-Response Relationship, Drug , Histidine Decarboxylase/metabolism , Hypothalamic Area, Lateral/chemistry , Hypothalamic Area, Lateral/metabolism , Immunohistochemistry , In Vitro Techniques , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neurons/chemistry , Neurons/metabolism , Nickel/pharmacology , Orexin Receptors , Orexins , Patch-Clamp Techniques , Potassium/metabolism , Rats , Rats, Wistar , Receptors, G-Protein-Coupled , Receptors, Neuropeptide/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Sodium-Calcium Exchanger/antagonists & inhibitors , Sodium-Calcium Exchanger/metabolism , Tetrodotoxin/pharmacology
19.
Neurosci Lett ; 306(3): 141-4, 2001 Jun 29.
Article in English | MEDLINE | ID: mdl-11406315

ABSTRACT

Mu opioid receptors occur throughout the brain, but central sites where ligand neuromodulatory effects occur during glucopenia have not been identified. The present studies investigated whether septal, preoptic, and hypothalamic neurons that express immunoreactivity for this receptor are transcriptionally activated in response to the glucose antimetabolite, 2-deoxy-D-glucose (2DG), and if intracerebroventricular (icv) administration of the selective mu receptor antagonist, CTOP, modifies this functional response to glucose substrate imbalance. Neurons labeled for mu receptor-immunoreactivity (-ir) were observed in the lateral septal nucleus (LS), medial septum (MS), anterior division of the stria terminalis (BSTa), median preoptic nucleus (MEPO), medial preoptic nucleus (MPN), parastrial nucleus (PS), anterior hypothalamic periventricular nucleus (PVa), and lateral hypothalamic area (LPO). 2DG injection (400 mg/kg i.p.) resulted in co-labeling of mu receptor-positive neurons in the LS, MS, BSTa, MEPO, PVa, and LPO for nuclear Fos-ir. Icv delivery of CTOP decreased mean numbers of co-labeled neurons in the LS, MS, BSTa, and MEPO. These results provide evidence for transactivational effects of glucopenia on mu opioid receptor-expressing neurons within the septum, preoptic area, and hypothalamus, and suggest that the functional status of these receptors within discrete septopreoptic sites may be critical for maximal glucoprivic induction of the Fos stimulus-transcription cascade within local cells. These results thus support the view that the neural loci described above may serve as substrates for regulatory effects of mu opioid receptor ligands on central compensatory activities during acute glucose deprivation.


Subject(s)
Glucose/deficiency , Hypothalamic Area, Lateral/chemistry , Preoptic Area/chemistry , Proto-Oncogene Proteins c-fos/analysis , Receptors, Opioid, mu/analysis , Somatostatin/analogs & derivatives , Somatostatin/pharmacology , Animals , Antibodies , Antimetabolites/pharmacology , Deoxyglucose/pharmacology , Hypothalamic Area, Lateral/drug effects , Hypothalamic Area, Lateral/metabolism , Male , Preoptic Area/drug effects , Preoptic Area/metabolism , Proto-Oncogene Proteins c-fos/immunology , Rats , Receptors, Opioid, mu/antagonists & inhibitors , Receptors, Opioid, mu/immunology , Septal Nuclei/chemistry , Septal Nuclei/drug effects , Septal Nuclei/metabolism , Septum of Brain/chemistry , Septum of Brain/drug effects , Septum of Brain/metabolism
20.
J Comp Neurol ; 435(1): 6-25, 2001 Jun 18.
Article in English | MEDLINE | ID: mdl-11370008

ABSTRACT

Orexins (hypocretins) are neuropeptides synthesized in the central nervous system exclusively by neurons of the lateral hypothalamus. Orexin-containing neurons have widespread projections and have been implicated in complex physiological functions including feeding behavior, sleep states, neuroendocrine function, and autonomic control. Two orexin receptors (OX(1)R and OX(2)R) have been identified, with distinct expression patterns throughout the brain, but a systematic examination of orexin receptor expression in the brain has not appeared. We used in situ hybridization histochemistry to examine the patterns of expression of mRNA for both orexin receptors throughout the brain. OX(1)R mRNA was observed in many brain regions including the prefrontal and infralimbic cortex, hippocampus, paraventricular thalamic nucleus, ventromedial hypothalamic nucleus, dorsal raphe nucleus, and locus coeruleus. OX(2)R mRNA was prominent in a complementary distribution including the cerebral cortex, septal nuclei, hippocampus, medial thalamic groups, raphe nuclei, and many hypothalamic nuclei including the tuberomammillary nucleus, dorsomedial nucleus, paraventricular nucleus, and ventral premammillary nucleus. The differential distribution of orexin receptors is consistent with the proposed multifaceted roles of orexin in regulating homeostasis and may explain the unique role of the OX(2)R receptor in regulating sleep state stability.


Subject(s)
Hypothalamic Area, Lateral/physiology , Rats, Sprague-Dawley/physiology , Receptors, Neuropeptide/genetics , Animals , Autonomic Nervous System/physiology , Cerebral Cortex/chemistry , Cerebral Cortex/physiology , Feeding Behavior/physiology , Gene Expression/physiology , Hippocampus/chemistry , Hippocampus/physiology , Hypothalamic Area, Lateral/chemistry , In Situ Hybridization , Locus Coeruleus/chemistry , Locus Coeruleus/physiology , Male , Midline Thalamic Nuclei/chemistry , Midline Thalamic Nuclei/physiology , Narcolepsy/physiopathology , Orexin Receptors , RNA, Messenger/analysis , Raphe Nuclei/chemistry , Raphe Nuclei/physiology , Rats , Receptors, G-Protein-Coupled , Sleep/physiology , Specific Pathogen-Free Organisms , Ventromedial Hypothalamic Nucleus/chemistry , Ventromedial Hypothalamic Nucleus/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...