Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
J Acupunct Meridian Stud ; 14(6): 207-218, 2021 Dec 31.
Article in English | MEDLINE | ID: mdl-35770600

ABSTRACT

Background: Myocardial ischemia reperfusion injury (MIRI) is an important mechanism of post-myocardial infarction injury and a main cause of death in patients with ischemic heart disease. Electroacupuncture (EA) pretreatment is effective for the prevention and treatment of MIRI, but mechanisms mediating the effects of cardiovascular disease EA treatments remain unclear. Objectives: To determine whether the lateral hypothalamus (LHA) and the cerebellar fastigial nucleus (FN) are involved in the protective effects of EA stimulation on MIRI. Methods: EA pretreatment was performed for 7 days before the establishment of the MIRI model. ST-segment changes on electrocardiograms were recorded and the Curtis-Walker arrhythmia score was used to evaluate changes in reperfusion injury. Hematoxylin-eosin staining was applied to evaluate the pathological and morphological changes in myocardial tissue. c-fos expression in the LHA and FN was determined by immunofluorescence staining. Glutamic (Glu) and γ-Aminobutyric acid (GABA) levels were measured using a high-performance liquid chromatography-electrochemical method. Results: EA pretreatment reduced ST-segment elevation, arrhythmia scores, and morphological changes in MIRI myocardial cells in rats, and decreased the c-fos protein expression in LHA/FN nuclei. MIRI was associated with an imbalance between GABA and Glu levels, whereas EA pretreatment increased GABA levels and decreased Glu levels in the LHA/FN. Conclusion: FN and LHA are involved in the EA-mediated attenuation of MIRI. Pretreatment with EA plays a protective role in the myocardium by regulating Glu and GABA release in the LHA and FN.


Subject(s)
Electroacupuncture , Myocardial Reperfusion Injury , Animals , Cerebellar Nuclei , Hypothalamic Area, Lateral , Myocardial Reperfusion Injury/therapy , Rats , gamma-Aminobutyric Acid
2.
Neuron ; 107(2): 368-382.e8, 2020 07 22.
Article in English | MEDLINE | ID: mdl-32442399

ABSTRACT

The ventral tegmental area (VTA) has dopamine, GABA, and glutamate neurons, which have been implicated in reward and aversion. Here, we determined whether VTA-glutamate or -GABA neurons play a role in innate defensive behavior. By VTA cell-type-specific genetic ablation, we found that ablation of glutamate, but not GABA, neurons abolishes escape behavior in response to threatening stimuli. We found that escape behavior is also decreased by chemogenetic inhibition of VTA-glutamate neurons and detected increases in activity in VTA-glutamate neurons in response to the threatening stimuli. By ultrastructural and electrophysiological analysis, we established that VTA-glutamate neurons receive a major monosynaptic glutamatergic input from the lateral hypothalamic area (LHA) and found that photoinhibition of this input decreases escape responses to threatening stimuli. These findings indicate that VTA-glutamate neurons are activated by and required for innate defensive responses and that information on threatening stimuli to VTA-glutamate neurons is relayed by LHA-glutamate neurons.


Subject(s)
Aggression/physiology , Glutamic Acid/physiology , Neurons/physiology , Ventral Tegmental Area/cytology , Ventral Tegmental Area/physiology , Animals , Escape Reaction , Humans , Hypothalamic Area, Lateral/cytology , Hypothalamic Area, Lateral/physiology , Hypothalamus/cytology , Hypothalamus/physiology , Mice , Neurons/ultrastructure , Optogenetics , Photic Stimulation , Reflex, Monosynaptic/physiology , Ventral Tegmental Area/ultrastructure , gamma-Aminobutyric Acid/physiology
3.
Curr Biol ; 29(17): 2775-2789.e7, 2019 09 09.
Article in English | MEDLINE | ID: mdl-31422881

ABSTRACT

Lower urinary tract symptoms (LUTS) are exceptionally common and debilitating, and they are likely caused or exacerbated by dysfunction of neural circuits controlling bladder function. An incomplete understanding of neural control of bladder function limits our ability to clinically address LUTS. Barrington's nucleus (Bar) provides descending control of bladder and sphincter function, and its glutamatergic neurons expressing corticotropin releasing hormone (BarCrh/Vglut2) are implicated in bladder control. However, it remains unclear whether this subset of Bar neurons is necessary for voiding, and the broader circuitry providing input to this control center remains largely unknown. Here, we examine the contribution to micturition behavior of BarCrh/Vglut2 neurons relative to the overall BarVglut2 population. First, we identify robust, excitatory synaptic input to Bar. Glutamatergic axons from the periaqueductal gray (PAG) and lateral hypothalamic area (LHA) intensely innervate and are functionally connected to Bar, and optogenetic stimulation of these axon terminals reliably provokes voiding. Similarly, optogenetic stimulation of BarVglut2 neurons triggers voiding, whereas stimulating the BarCrh/Vglut2 subpopulation causes bladder contraction, typically without voiding. Next, we genetically ablate either BarVglut2 or BarCrh/Vglut2 neurons and found that only BarVglut2 ablation replicates the profound urinary retention produced by conventional lesions in this region. Fiber photometry recordings reveal that BarVglut2 neuron activity precedes increased bladder pressure, while activity of BarCrh/Vglut2 is phase delayed. Finally, deleting Crh from Bar neurons has no effect on voiding and related bladder physiology. Our results help identify the circuitry that modulates Bar neuron activity and identify subtypes that may serve different roles in micturition.


Subject(s)
Barrington's Nucleus/physiology , Hypothalamus/metabolism , Mesencephalon/metabolism , Neurons/physiology , Urination/physiology , Animals , Corticotropin-Releasing Hormone/metabolism , Female , Male , Mice , Neurons, Afferent
4.
J Comp Neurol ; 527(18): 2973-3001, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31152440

ABSTRACT

Melanin-concentrating hormone (MCH) is a conserved neuropeptide, predominantly located in the diencephalon of vertebrates, and associated with a wide range of functions. While functional studies have focused on the use of the traditional mouse laboratory model, critical gaps exist in our understanding of the morphology of the MCH system in this species. Even less is known about the nontraditional animal model Neotomodon alstoni (Mexican volcano mouse). A comparative morphological study among these rodents may, therefore, contribute to a better understanding of the evolution of the MCH peptidergic system. To this end, we employed diverse immunohistochemical protocols to identify key aspects of the MCH system, including its spatial relationship to another neurochemical population of the tuberal hypothalamus, the orexins. Three-dimensional (3D) reconstructions were also employed to convey a better sense of spatial distribution to these neurons. Our results show that the distribution of MCH neurons in all rodents studied follows a basic plan, but individual characteristics are found for each species, such as the preeminence of a periventricular group only in the rat, the lack of posterior groups in the mouse, and the extensive presence of MCH neurons in the anterior hypothalamic area of Neotomodon. Taken together, these data suggest a strong anatomical substrate for previously described functions of the MCH system, and that particular neurochemical and morphological features may have been determinant to species-specific phenotypes in rodent evolution.


Subject(s)
Hypothalamic Hormones/metabolism , Hypothalamus/cytology , Hypothalamus/metabolism , Melanins/metabolism , Melanophores/metabolism , Pituitary Hormones/metabolism , Animals , Female , Hypothalamic Hormones/analysis , Hypothalamus/chemistry , Male , Melanins/analysis , Mice , Mice, Inbred C57BL , Phylogeny , Pituitary Hormones/analysis , Rats , Rats, Sprague-Dawley , Species Specificity
5.
Neuroendocrinology ; 108(3): 190-200, 2019.
Article in English | MEDLINE | ID: mdl-30625474

ABSTRACT

BACKGROUND/AIMS: Nesfatin-1, processed from nucleobindin-2 (NUCB2), is a potent anorexigenic peptide being expressed in rodent hypothalamic nuclei and involved in the regulation of feeding behavior and body weight in animals. The present study aimed to investigate NUCB2/nesfatin-1 protein expression in the human hypothalamus as well as its correlation with body weight. METHODS: Sections of hypothalamus and adjacent cholinergic basal forebrain nuclei, including the nucleus basalis of Meynert (NBM) and the diagonal band of Broca (DBB), from 25 autopsy cases (17 males, 8 females; 8 lean, 9 overweight, 8 obese) were examined using immunohistochemistry and double immunofluorescence labeling. RESULTS: Prominent NUCB2/nesfatin-1 immunoexpression was detected in supraoptic, paraventricular, and infundibular nuclei, lateral hypothalamic area (LHA)/perifornical region, and NBM/DBB. NUCB2/nesfatin-1 was found to extensively colocalize with (a) oxytocin and vasopressin in paraventricular and supraoptic nuclei, (b) melanin-concentrating hormone in the LHA, and (c) cocaine- and amphetamine-regulated transcript in infundibular and paraventricular nuclei and LHA. Interestingly, in the LHA, NUCB2/nesfatin-1 protein expression was significantly decreased in obese, compared with lean (p < 0.01) and overweight (p < 0.05) subjects. CONCLUSIONS: The findings of the present study are suggestive of a potential role for NUCB2/nesfatin-1 as an integral regulator of food intake and energy homeostasis in the human hypothalamus. In the LHA, an appetite- and reward-related brain area, reduced NUCB2/nesfatin-1 immunoexpression may contribute to dysregulation of homeostatic and/or hedonic feeding behavior and obesity. NUCB2/nesfatin-1 localization in NBM/DBB might imply its participation in the neuronal circuitry controlling cognitive influences on food intake and give impetus towards unraveling additional biological actions of NUCB2/nesfatin-1 in human neuronal networks.


Subject(s)
Hypothalamic Hormones/metabolism , Hypothalamus/metabolism , Melanins/metabolism , Nerve Tissue Proteins/metabolism , Nucleobindins/biosynthesis , Obesity/metabolism , Oxytocin/metabolism , Pituitary Hormones/metabolism , Vasopressins/metabolism , Adult , Aged , Aged, 80 and over , Body Weight , Case-Control Studies , Female , Humans , Male , Middle Aged
6.
Mol Metab ; 20: 178-193, 2019 02.
Article in English | MEDLINE | ID: mdl-30528281

ABSTRACT

OBJECTIVE: The supramammillary nucleus (SuM) is nestled between the lateral hypothalamus (LH) and the ventral tegmental area (VTA). This neuroanatomical position is consistent with a potential role of this nucleus to regulate ingestive and motivated behavior. Here neuroanatomical, molecular, and behavior approaches are utilized to determine whether SuM contributes to ingestive and food-motivated behavior control. METHODS: Through the application of anterograde and retrograde neural tract tracing with novel designer viral vectors, the current findings show that SuM neurons densely innervate the LH in a sex dimorphic fashion. Glucagon-like peptide-1 (GLP-1) is a clinically targeted neuro-intestinal hormone with a well-established role in regulating energy balance and reward behaviors. Here we determine that GLP-1 receptors (GLP-1R) are expressed throughout the SuM of both sexes, and also directly on SuM LH-projecting neurons and investigate the role of SuM GLP-1R in the regulation of ingestive and motivated behavior in male and female rats. RESULTS: SuM microinjections of the GLP-1 analogue, exendin-4, reduced ad libitum intake of chow, fat, or sugar solution in both male and female rats, while food-motivated behaviors, measured using the sucrose motivated operant conditioning test, was only reduced in male rats. These data contrasted with the results obtained from a neighboring structure well known for its role in motivation and reward, the VTA, where females displayed a more potent response to GLP-1R activation by exendin-4. In order to determine the physiological role of SuM GLP-1R signaling regulation of energy balance, we utilized an adeno-associated viral vector to site-specifically deliver shRNA for the GLP-1R to the SuM. Surprisingly, and in contrast to previous results for the two SuM neighboring sites, LH and VTA, SuM GLP-1R knockdown increased food seeking and adiposity in obese male rats without altering food intake, body weight or food motivation in lean or obese, female or male rats. CONCLUSION: Taken together, these results indicate that SuM potently contributes to ingestive and motivated behavior control; an effect contingent on sex, diet/homeostatic energy balance state and behavior of interest. These data also extend the map of brain sites directly responsive to GLP-1 agonists, and highlight key differences in the role that GLP-1R play in interconnected and neighboring nuclei.


Subject(s)
Basal Ganglia/metabolism , Eating , Glucagon-Like Peptide 1/metabolism , Hypothalamus/metabolism , Motivation , Animals , Basal Ganglia/cytology , Basal Ganglia/physiology , Conditioning, Operant , Energy Metabolism , Female , Glucagon-Like Peptide-1 Receptor/genetics , Glucagon-Like Peptide-1 Receptor/metabolism , Hypothalamus/cytology , Hypothalamus/physiology , Male , Neural Pathways/cytology , Neural Pathways/metabolism , Neural Pathways/physiology , Rats , Rats, Sprague-Dawley , Sex Factors
7.
Nutrients ; 10(11)2018 Nov 10.
Article in English | MEDLINE | ID: mdl-30423806

ABSTRACT

It has been widely described that chronic intake of fructose causes metabolic alterations which can be associated with brain function impairment. In this study, we evaluated the effects of fructose intake on the sleep⁻wake cycle, locomotion, and neurochemical parameters in Wistar rats. The experimental group was fed with 10% fructose in drinking water for five weeks. After treatment, metabolic indicators were quantified in blood. Electroencephalographic recordings were used to evaluate the sleep architecture and the spectral power of frequency bands. Likewise, the locomotor activity and the concentrations of orexin A and monoamines were estimated. Our results show that fructose diet significantly increased the blood levels of glucose, cholesterol, and triglycerides. Fructose modified the sleep⁻wake cycle of rats, increasing the waking duration and conversely decreasing the non-rapid eye movement sleep. Furthermore, these effects were accompanied by increases of the spectral power at different frequency bands. Chronic consumption of fructose caused a slight increase in the locomotor activity as well as an increase of orexin A and dopamine levels in the hypothalamus and brainstem. Specifically, immunoreactivity for orexin A was increased in the ventral tegmental area after the intake of fructose. Our study suggests that fructose induces metabolic changes and stimulates the activity of orexinergic and dopaminergic neurons, which may be responsible for alterations of the sleep⁻wake cycle.


Subject(s)
Brain/drug effects , Dietary Sugars/pharmacology , Dopamine/metabolism , Feeding Behavior , Fructose/pharmacology , Orexins/metabolism , Sleep/drug effects , Animals , Blood Glucose/metabolism , Brain/cytology , Brain/metabolism , Brain Stem/drug effects , Brain Stem/metabolism , Diet , Hypothalamus/drug effects , Hypothalamus/metabolism , Lipids/blood , Locomotion/drug effects , Male , Motor Activity/drug effects , Rats, Wistar , Sleep Stages/drug effects , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism , Wakefulness/drug effects
8.
Neuroscience ; 379: 103-114, 2018 05 21.
Article in English | MEDLINE | ID: mdl-29534973

ABSTRACT

The hypothalamic energy sensor adenosine 5'-monophosphate-activated protein kinase (AMPK), an important regulator of counter-regulatory responses to hypoglycemia, responds to pharmacological manipulation of hindbrain AMPK activity. Dorsomedial hindbrain A2 noradrenergic neurons express hypoglycemia-sensitive metabolo-sensory biomarkers, including AMPK. Here, adult male rats were pretreated by intra-caudal fourth ventricular administration of the selective neurotoxin 6-hydroxydopamine (6-OHDA) to determine if catecholamine signaling from the aforesaid site governs hypothalamic AMPK activation during insulin-induced hypoglycemia (IIH). Micropunched arcuate (ARH), ventromedial (VMH), paraventricular (PVH), dorsomedial (DMH) nuclei and lateral hypothalamic area (LHA) tissues were obtained at the neutral protamine Hagedorn insulin-induced hypoglycemic nadir, coincident with A2 AMPK activation, for Western blot analysis of AMPK, phospho-AMPK (pAMPK), and relevant metabolic neuropeptides. ARH, VMH, LHA, and DMH norepinephrine levels were altered according to insulin dose; 6-OHDA-mediated reversal of these responses was site-specific. IIH elevated LHA and reduced VMH pAMPK protein, profiles that were respectively unchanged or increased by 6-OHDA. PVH and ARH pAMPK was resistant to IIH, but augmented in ARH of neurotoxin- plus insulin-treated rats. ARH neuropeptide Y (NPY) and pro-opiomelanocortin (POMC) proteins were correspondingly increased or refractory to IIH; 6-OHDA pretreatment normalized NPY and elevated POMC expression after insulin injection. Results demonstrate site-specific bi-directional adjustments in hypothalamic AMPK reactivity to hypoglycemia. Intensification of ARH/VMH pAMPK by 6-OHDA implies dorsomedial hindbrain improvement of energy balance in those sites during IIH. Neurotoxin-mediated augmentation versus suppression of basal catabolic (ARH POMC/VMH steroidogenic factor-1) or IIH-associated anabolic (ARH NPY) neuropeptide profiles, respectively, may involve local AMPK-dependent against independent mechanisms.


Subject(s)
Adenylate Kinase/metabolism , Catecholamines/metabolism , Hypoglycemia/metabolism , Hypothalamus/metabolism , Insulin/metabolism , Rhombencephalon/metabolism , Adrenergic Agents/pharmacology , Animals , Hypothalamus/drug effects , Insulin/administration & dosage , Male , Neuropeptide Y/metabolism , Oxidopamine/pharmacology , Phosphorylation , Pro-Opiomelanocortin/metabolism , Rats, Sprague-Dawley , Rhombencephalon/drug effects , Signal Transduction/drug effects
9.
Neurosurg Focus ; 43(3): E15, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28859567

ABSTRACT

The neurosurgical endeavor to treat psychiatric patients may have been part of human history since its beginning. The modern era of psychosurgery can be traced to the heroic attempts of Gottlieb Burckhardt and Egas Moniz to alleviate mental symptoms through the ablation of restricted areas of the frontal lobes in patients with disabling psychiatric illnesses. Thanks to the adaptation of the stereotactic frame to human patients, the ablation of large volumes of brain tissue has been practically abandoned in favor of controlled interventions with discrete targets. Consonant with the role of the hypothalamus in the mediation of the most fundamental approach-avoidance behaviors, some hypothalamic nuclei and regions, in particular, have been selected as targets for the treatment of aggressiveness (posterior hypothalamus), pathological obesity (lateral or ventromedial nuclei), sexual deviations (ventromedial nucleus), and drug dependence (ventromedial nucleus). Some recent improvements in outcomes may have been due to the use of stereotactically guided deep brain stimulation and the change of therapeutic focus from categorical diagnoses (such as schizophrenia) to dimensional symptoms (such as aggressiveness), which are nonspecific in terms of formal diagnosis. However, agreement has never been reached on 2 related issues: 1) the choice of target, based on individual diagnoses; and 2) reliable prediction of outcomes related to individual targets. Despite the lingering controversies on such critical aspects, the experience of the past decades should pave the way for advances in the field. The current failure of pharmacological treatments in a considerable proportion of patients with chronic disabling mental disorders is reminiscent of the state of affairs that prevailed in the years before the early psychosurgical attempts. This article reviews the functional organization of the hypothalamus, the effects of ablation and stimulation of discrete hypothalamic regions, and the stereotactic targets that have most often been used in the treatment of psychopathological and behavioral symptoms; finally, the implications of current and past experience are presented from the perspective of how this fund of knowledge may usefully contribute to the future of hypothalamic psychosurgery.


Subject(s)
Hypothalamus/diagnostic imaging , Hypothalamus/surgery , Mental Disorders/diagnostic imaging , Mental Disorders/surgery , Neurosurgical Procedures/methods , Humans , Stereotaxic Techniques
10.
Behav Brain Res ; 324: 87-95, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28212941

ABSTRACT

Previously, we showed that the blockade of α1-adrenoreceptors in the median raphe nucleus (MnR) increased food intake in free-feeding rats, indicating that adrenergic mechanisms in the MnR participate in the regulation of food intake. However, the impact of such a pharmacological manipulation on other neural circuits related to food intake remains unknown. In the current study, we sought to identify forebrain regions which are responsive to α1-adrenergic receptor blockade and presumably involved in the modulation of the feeding response. For this purpose, we examined the induction of c-Fos immunoreactivity in forebrain structures following injections of the α1-adrenoceptor antagonist prazosin into the MnR of free-feeding rats. To determine the chemical identity of hypothalamic c-Fos-positive cells, we then conducted double-label immunohistochemistry for Fos/orexin (OX) or Fos/melanin-concentrating hormone (MCH). Finally, we combined anterograde tracing from the MnR with immunohistochemical detection of orexin. Prazosin injections into the MnR significantly increased food intake. The ingestive response was accompanied by an increase in Fos expression in the basolateral amygdala (BLA) and lateral hypothalamic area (LHA). In the LHA, Fos expression occurred in neurons expressing OX, but not MCH. Combined anterograde tracing experiments revealed that LHA OX neurons are prominently targeted by MnR axons. These findings suggest that intra-MnR injection of prazosin, via activation of orexinergic neurons in the LHA and non-orexinergic neurons in the BLA, evoked a motivational response toward food intake.


Subject(s)
Adrenergic alpha-1 Receptor Antagonists/administration & dosage , Eating , Neurons/metabolism , Prazosin/administration & dosage , Raphe Nuclei/physiology , Receptors, Adrenergic, alpha-1/physiology , Animals , Basolateral Nuclear Complex/metabolism , Hypothalamic Hormones/metabolism , Hypothalamus/metabolism , Male , Melanins/metabolism , Orexins/metabolism , Pituitary Hormones/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Raphe Nuclei/drug effects , Rats, Wistar
11.
Life Sci ; 148: 241-6, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26874026

ABSTRACT

AIMS: Melanin-concentrating hormone (MCH) is implicated in the control of food intake, body weight regulation and energy homeostasis. Lactation is an important physiological model to study the hypothalamic integration of peripheral sensory signals, such as suckling stimuli and those related to energy balance. MCH can be detected in the medial preoptic area (MPOA), especially around the 19th day of lactation, when this hormone is described as displaying a peak synthesis followed by a decrease after weaning. The physiological significance of this phenomenon is unclear. Therefore, we aimed to investigate hypothalamic changes associated to sensory stimulation by the litter, in special its influence over MCH synthesis. MAIN METHODS: Female Wistar rats (n=56) were euthanized everyday from lactation days 15-21, with or without suckling stimulus (WS and NS groups, respectively). MCH and Fos immunoreactivity were evaluated in the MPOA and lateral and incerto-hypothalamic areas (LHA and IHy). KEY FINDINGS: Suckling stimulus induced Fos synthesis in all regions studied. An increase on the number of suckling-induced Fos-ir neurons could be detected in the LHA after the 18th day. Conversely, the amount of MCH decreased in the MPOA from days 15-21, independent of suckling stimulation. No colocalization between MCH and Fos could be detected in any region analyzed. SIGNIFICANCE: Suckling stimulus is capable of stimulating hypothalamic regions not linked to maternal behavior, possibly to mediate energy balance aspects of lactation. Although dams are hyperphagic before weaning, this behavioral change does not appear to be mediated by MCH.


Subject(s)
Hypothalamic Hormones/biosynthesis , Hypothalamus/metabolism , Lactation/metabolism , Melanins/biosynthesis , Melanophores/metabolism , Pituitary Hormones/biosynthesis , Proto-Oncogene Proteins c-fos/biosynthesis , Animals , Animals, Suckling , Female , Hypothalamic Hormones/analysis , Melanins/analysis , Pituitary Hormones/analysis , Proto-Oncogene Proteins c-fos/analysis , Rats , Rats, Wistar
12.
Neuroscience ; 289: 392-405, 2015 Mar 19.
Article in English | MEDLINE | ID: mdl-25613687

ABSTRACT

Melanin-concentrating hormone [MCH] is a neuropeptide that modulates several behaviors, such as feeding and reward. Because the hedonic and rewarding features of a food also influence feeding behavior, the nucleus accumbens [Acb] has been highlighted as a key area integrating these roles. Functional data confirm that MCH acts on a subdivision of the Acb; however, considering the importance of finding anatomical and neurochemical data that correlate the previously demonstrated function of MCH, we delineated this investigation based on the following points: (1) Is there a pattern of innervation by MCH fibers regarding the subregions within the Acb? (2) Specifically, which hypothalamic nuclei synthesize MCH and innervate the Acb? (3) Finally, what are the neurochemical identities of the accumbal neurons innervated by MCH inputs? We examined the MCH immunoreactivity [MCH-ir] in the Acb in rat brains using the peroxidase technique. Additionally, after injecting retrograde neuronal tracer [Fluoro-Gold® - FG®] into subdivisions of the Acb [shell or core], we mapped single- or double-labeled cells. Moreover, using a double immunoperoxidase protocol, we investigated the MCH-ir fibers for gamma-aminobutyric acid [GABA]-ir and choline acetyltransferase [ChAT]-ir cells in the shell subdivision of the Acb [AcbSh]. We found that the MCH-ir fibers preferentially innervate the medial AcbSh, particularly the septal pole. This innervation originated from the incerto-hypothalamic area [IHy], internuclear area, lateral hypothalamic area, perifornical area, periventricular nucleus and posterior hypothalamus. Moreover, the IHy has the highest relationship between double/single retrogradely labeled cells [n=5.33±0.66/16±0.93, i.e. 33.33%] in the whole hypothalamus. Furthermore, our data suggest that MCH-ir fibers are in apposition to GABAergic and cholinergic cells in the AcbSh. Therefore, we provide anatomical support to the ongoing functional studies investigating the relation among the hypothalamus, MCH transmission into the Acb and the involvement of known neuronal phenotypes within the AcbSh.


Subject(s)
Acetylcholine/metabolism , Hypothalamic Hormones/metabolism , Hypothalamus/cytology , Melanins/metabolism , Neurons/cytology , Nucleus Accumbens/cytology , Pituitary Hormones/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Cell Count , Fluorescent Antibody Technique , Hypothalamus/metabolism , Immunoenzyme Techniques , Male , Neuroanatomical Tract-Tracing Techniques , Neurons/metabolism , Nucleus Accumbens/metabolism , Photomicrography , Rats, Long-Evans , Stilbamidines
13.
Neuroscience ; 250: 599-613, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-23912034

ABSTRACT

Experiments were done to investigate whether hypothalamic hypocretin-1 (hcrt-1; orexin-A) neurons that sent axonal projections to cardiovascular responsive sites in the nucleus of the solitary tract (NTS) co-expressed leucine-enkephalin (L-Enk), and to determine the effects of co-administration of hcrt-1 and D-Ala2,D-Leu5-Enkephalin (DADL) into NTS on mean arterial pressure (MAP) and heart rate. In the first series, in the Wistar rat the retrograde tract-tracer fluorogold (FG) was microinjected (50nl) into caudal NTS sites at which L-glutamate (0.25 M; 10 nl) elicited decreases in MAP and where fibers hcrt-1 immunoreactive fibers were observed that also contained L-Enk immunoreactivity. Of the number of hypothalamic hcrt-1 immunoreactive neurons identified ipsilateral to the NTS injection site (1207 ± 78), 32.3 ± 2.3% co-expressed L-Enk immunoreactivity and of these, 2.6 ± 1.1% were retrogradely labeled with FG. Hcrt-1/L-Enk neurons projecting to NTS were found mainly within the perifornical region. In the second series, the region of caudal NTS found to contain axons that co-expressed hcrt-1 and L-Enk immunoreactivity was microinjected with a combination of hcrt-1 and DADL in α-chloralose anesthetized Wistar rats. Microinjection of DADL into NTS elicited depressor and bradycardia responses similar to those elicited by microinjection of hcrt-1. An hcrt-1 injection immediately after the DADL injection elicited an almost twofold increase in the magnitude of the depressor and bradycardia responses compared to those elicited by hcrt-1 alone. Prior injections of the non-specific opioid receptor antagonist naloxone or the specific opioid δ-receptor antagonist ICI 154,129 significantly attenuated the cardiovascular responses to the combined hcrt-1-DADL injections. Taken together, these data suggest that activation of hypothalamic-opioidergic neuronal systems contribute to the NTS hcrt-1 induced cardiovascular responses, and that this descending hypothalamo-medullary pathway may represent the anatomical substrate by which hcrt-1/L-Enk neurons function in the coordination of autonomic-cardiovascular responses during different behavioral states.


Subject(s)
Arterial Pressure/physiology , Enkephalin, Leucine/metabolism , Hypothalamus/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Neural Pathways/physiology , Neurons/physiology , Neuropeptides/metabolism , Solitary Nucleus/physiology , Animals , Arterial Pressure/drug effects , Data Interpretation, Statistical , Enkephalin, Leucine/analogs & derivatives , Enkephalin, Leucine/pharmacology , Enkephalin, Leucine-2-Alanine/analogs & derivatives , Enkephalin, Leucine-2-Alanine/pharmacology , Heart Rate/drug effects , Heart Rate/physiology , Hypothalamus/cytology , Hypothalamus/metabolism , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/pharmacology , Male , Melphalan/analogs & derivatives , Melphalan/pharmacology , Microinjections , Narcotic Antagonists/pharmacology , Neural Pathways/cytology , Neural Pathways/metabolism , Neurons/metabolism , Neuropeptides/pharmacology , Orexins , Rats , Rats, Wistar , Solitary Nucleus/cytology , Solitary Nucleus/metabolism , Stilbamidines
14.
J Chem Neuroanat ; 54: 5-19, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23619059

ABSTRACT

Numerous functions have been attributed to the Edinger-Westphal nucleus (EW), including those related to feeding behavior, pain control, alcohol consumption and the stress response. The EW is thought to consist of two parts: one controls accommodation, choroidal blood flow and pupillary constriction, primarily comprising cholinergic cells and projecting to the ciliary ganglion; and the other would be involved in the non-ocular functions mentioned above, comprising peptide-producing neurons and projecting to the brainstem, spinal cord and prosencephalic regions. Despite the fact that the EW is well known, its connections have yet to be described in detail. The aim of this work was to produce a map of the hypothalamic sources of afferents to the EW in the rat. We injected the retrograde tracer Fluoro-Gold into the EW, and using biotinylated dextran amine, injected into afferent sources as the anterograde control. We found retrogradely labeled cells in the following regions: subfornical organ, paraventricular hypothalamic nucleus, arcuate nucleus, lateral hypothalamic area, zona incerta, posterior hypothalamic nucleus, medial vestibular nucleus and cerebellar interpositus nucleus. After injecting BDA into the paraventricular hypothalamic nucleus, lateral hypothalamic area and posterior hypothalamic nucleus, we found anterogradely labeled fibers in close apposition to and potential synaptic contact with urocortin 1-immunoreactive cells in the EW. On the basis of our findings, we can suggest that the connections between the EW and the hypothalamic nuclei are involved in controlling stress responses and feeding behavior.


Subject(s)
Afferent Pathways/anatomy & histology , Brain Stem/anatomy & histology , Hypothalamus/anatomy & histology , Animals , Immunohistochemistry , Male , Rats , Rats, Long-Evans
SELECTION OF CITATIONS
SEARCH DETAIL