Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
Más filtros

Medicinas Complementárias
Métodos Terapéuticos y Terapias MTCI
Intervalo de año de publicación
1.
Front Neuroendocrinol ; 70: 101069, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37149229

RESUMEN

Hypothalamic melanin-concentrating hormone (MCH) neurons participate in many fundamental neuroendocrine processes. While some of their effects can be attributed to MCH itself, others appear to depend on co-released neurotransmitters. Historically, the subject of fast neurotransmitter co-release from MCH neurons has been contentious, with data to support MCH neurons releasing GABA, glutamate, both, and neither. Rather than assuming a position in that debate, this review considers the evidence for all sides and presents an alternative explanation: neurochemical identity, including classical neurotransmitter content, is subject to change. With an emphasis on the variability of experimental details, we posit that MCH neurons may release GABA and/or glutamate at different points according to environmental and contextual factors. Through the lens of the MCH system, we offer evidence that the field of neuroendocrinology would benefit from a more nuanced and dynamic interpretation of neurotransmitter identity.


Asunto(s)
Hormonas Hipotalámicas , Hormonas Hipotalámicas/metabolismo , Hormonas Hipotalámicas/farmacología , Hormonas Hipofisarias/farmacología , Hormonas Hipofisarias/fisiología , Neuronas/metabolismo , Melaninas/farmacología , Melaninas/fisiología , Hipotálamo/metabolismo , Ácido Glutámico/farmacología , Ácido Glutámico/fisiología , Neurotransmisores , Ácido gamma-Aminobutírico
2.
Int J Mol Sci ; 22(13)2021 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-34209728

RESUMEN

Parental behaviour is a comprehensive set of neural responses to social cues. The neural circuits that govern parental behaviour reside in several putative nuclei in the brain. Melanin concentrating hormone (MCH), a neuromodulator that integrates physiological functions, has been confirmed to be involved in parental behaviour, particularly in crouching behaviour during nursing. Abolishing MCH neurons in innate MCH knockout males promotes infanticide in virgin male mice. To understand the mechanism and function of neural networks underlying parental care and aggression against pups, it is essential to understand the basic organisation and function of the involved nuclei. This review presents newly discovered aspects of neural circuits within the hypothalamus that regulate parental behaviours.


Asunto(s)
Hipotálamo/citología , Red Nerviosa/fisiología , Comportamiento de Nidificación/fisiología , Agresión/psicología , Animales , Conducta Animal/fisiología , Hormonas Hipotalámicas/genética , Hormonas Hipotalámicas/fisiología , Hipotálamo/fisiología , Masculino , Melaninas/genética , Melaninas/fisiología , Ratones , Ratones Noqueados , Hormonas Hipofisarias/genética , Hormonas Hipofisarias/fisiología
3.
Genesis ; 56(8): e23217, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29806135

RESUMEN

The neuropeptide, melanin concentrating hormone (MCH), and its G protein-coupled receptor, melanin concentrating hormone receptor 1 (Mchr1), are expressed centrally in adult rodents. MCH signaling has been implicated in diverse behaviors such as feeding, sleep, anxiety, as well as addiction and reward. While a model utilizing the Mchr1 promoter to drive constitutive expression of Cre recombinase (Mchr1-Cre) exists, there is a need for an inducible Mchr1-Cre to determine the roles for this signaling pathway in neural development and adult neuronal function. Here, we generated a BAC transgenic mouse where the Mchr1 promotor drives expression of tamoxifen inducible CreER recombinase. Many aspects of the Mchr1-Cre expression pattern are recapitulated by the Mchr1-CreER model, though there are also notable differences. Most strikingly, compared to the constitutive model, the new Mchr1-CreER model shows strong expression in adult animals in hypothalamic brain regions involved in feeding behavior but diminished expression in regions involved in reward, such as the nucleus accumbens. The inducible Mchr1-CreER allele will help reveal the potential for Mchr1 signaling to impact neural development and subsequent behavioral phenotypes, as well as contribute to the understanding of the MCH signaling pathway in terminally differentiated adult neurons and the diverse behaviors that it influences.


Asunto(s)
Hormonas Hipotalámicas/fisiología , Melaninas/fisiología , Hormonas Hipofisarias/fisiología , Receptores de Somatostatina/fisiología , Animales , Encéfalo/metabolismo , Encéfalo/fisiología , Hormonas Hipotalámicas/metabolismo , Hipotálamo/metabolismo , Integrasas , Melaninas/metabolismo , Ratones , Ratones Transgénicos , Modelos Animales , Neuronas/metabolismo , Neuropéptidos/metabolismo , Hormonas Hipofisarias/metabolismo , Receptores de Somatostatina/metabolismo , Transducción de Señal , Tamoxifeno
4.
J Neurosci ; 38(10): 2505-2518, 2018 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-29431649

RESUMEN

Glutamate transporter 1 (GLT1) is the main astrocytic transporter that shapes glutamatergic transmission in the brain. However, whether this transporter modulates sleep-wake regulatory neurons is unknown. Using quantitative immunohistochemical analysis, we assessed perisomatic GLT1 apposition with sleep-wake neurons in the male rat following 6 h sleep deprivation (SD) or following 6 h undisturbed conditions when animals were mostly asleep (Rest). We found that SD decreased perisomatic GLT1 apposition with wake-promoting orexin neurons in the lateral hypothalamus compared with Rest. Reduced GLT1 apposition was associated with tonic presynaptic inhibition of excitatory transmission to these neurons due to the activation of Group III metabotropic glutamate receptors, an effect mimicked by a GLT1 inhibitor in the Rest condition. In contrast, SD resulted in increased GLT1 apposition with sleep-promoting melanin-concentrating hormone (MCH) neurons in the lateral hypothalamus. Functionally, this decreased the postsynaptic response of MCH neurons to high-frequency synaptic activation without changing presynaptic glutamate release. The changes in GLT1 apposition with orexin and MCH neurons were reversed after 3 h of sleep opportunity following 6 h SD. These SD effects were specific to orexin and MCH neurons, as no change in GLT1 apposition was seen in basal forebrain cholinergic or parvalbumin-positive GABA neurons. Thus, within a single hypothalamic area, GLT1 differentially regulates excitatory transmission to wake- and sleep-promoting neurons depending on sleep history. These processes may constitute novel astrocyte-mediated homeostatic mechanisms controlling sleep-wake behavior.SIGNIFICANCE STATEMENT Sleep-wake cycles are regulated by the alternate activation of sleep- and wake-promoting neurons. Whether and how astrocytes can regulate this reciprocal neuronal activity are unclear. Here we report that, within the lateral hypothalamus, where functionally opposite wake-promoting orexin neurons and sleep-promoting melanin-concentrating hormone neurons codistribute, the glutamate transporter GLT1, mainly present on astrocytes, distinctly modulates excitatory transmission in a cell-type-specific manner and according to sleep history. Specifically, GLT1 is reduced around the somata of orexin neurons while increased around melanin-concentrating hormone neurons following sleep deprivation, resulting in different forms of synaptic plasticity. Thus, astrocytes can fine-tune the excitability of functionally discrete neurons via glutamate transport, which may represent novel regulatory mechanisms for sleep.


Asunto(s)
Transportador 2 de Aminoácidos Excitadores/metabolismo , Hormonas Hipotalámicas/fisiología , Melaninas/fisiología , Orexinas/fisiología , Hormonas Hipofisarias/fisiología , Privación de Sueño/metabolismo , Privación de Sueño/fisiopatología , Transmisión Sináptica , Animales , Hipotálamo/fisiopatología , Masculino , Neuronas , Sistema Nervioso Parasimpático/fisiopatología , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Receptores de Glutamato Metabotrópico/metabolismo , Sueño/fisiología , Vigilia/fisiología , Ácido gamma-Aminobutírico/fisiología
5.
Neurosci Res ; 118: 74-81, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28526553

RESUMEN

Sleep is one of the most important physiological functions in mammals. It is regulated by not only homeostatic regulation but also circadian clock. Several neuropeptide-producing neurons located in the hypothalamus are implicated in the regulation of sleep/wakefulness. Among them, orexin/hypocretin-producing neurons (orexin neurons) are a crucial component for maintenance of wakefulness, because lack of orexin function results in narcolepsy, which is a sleep disorder. Recent findings have identified substances that excite or inhibit neural activity of orexin neurons. Furthermore neural projections of the neurons which release these substances have been revealed. In addition to orexin, melanin concentrating hormone (MCH)-producing neurons in the lateral hypothalamic area (LHA) are also implicated in the regulation of sleep/wakefulness. MCH neurons are active during sleep but become silent during wakefulness. Recently developed innovative methods including optogenetics and pharmacogenetics have provided substantial insights into the regulation of sleep/wakefulness. In vivo optical recordings and retrograde and anterograde tracing methods will allow us to understand additional details regarding important interactions between these two types of neurons in the LHA and other neurons in the brain. Finally we discuss the circadian clock and sleep/wake cycle. Understanding of the neural networks and its circadian modulation of sleep/wake cycles remain to be investigated.


Asunto(s)
Hormonas Hipotalámicas/fisiología , Hipotálamo/fisiología , Melaninas/fisiología , Orexinas/fisiología , Hormonas Hipofisarias/fisiología , Sueño/fisiología , Vigilia/fisiología , Animales , Humanos
6.
Alcohol Clin Exp Res ; 40(10): 2199-2207, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27579857

RESUMEN

BACKGROUND: Reward and energy homeostasis are both regulated by a network of hypothalamic neuropeptide systems. The melanin-concentrating hormone (MCH) and its MCH-1 receptor (MCH1-R) modulate alcohol intake, but it remains unknown to what extent this reflects actions on energy balance or reward. Here, we evaluated the MCH1-R in regulation of caloric intake and motivation to consume alcohol in states of escalated consumption. METHODS: Rats had intermittent access (IA) to alcohol and were divided into high- and low-drinking groups. Food and alcohol consumption was assessed after administration of an MCH1-R antagonist, GW803430. Next, GW803430 was evaluated on alcohol self-administration in protracted abstinence induced by IA in high-drinking rats. Finally, the effect of GW803430 was assessed on alcohol self-administration in acute withdrawal in rats exposed to alcohol vapor. Gene expression of MCH and MCH1-R was measured in the hypothalamus and nucleus accumbens (NAc) in both acute and protracted abstinence. RESULTS: High-drinking IA rats consumed more calories from alcohol than chow and GW803430 decreased both chow and alcohol intake. In low-drinking rats, only food intake was affected. In protracted abstinence from IA, alcohol self-administration was significantly reduced by pretreatment with GW803430 and gene expression of both MCH and the MCH1-R were dysregulated in hypothalamus and NAc. In contrast, during acute withdrawal from vapor exposure, treatment with GW803430 did not affect alcohol self-administration, and no changes in MCH or MCH1-R gene expression were observed. CONCLUSIONS: Our data suggest a dual role of MCH and the MCH1-R in regulation of alcohol intake, possibly through mechanisms involving caloric intake and reward motivation. A selective suppression of alcohol self-administration during protracted abstinence by GW803430 was observed and accompanied by adaptations in gene expression of MCH and MCH1-R. Selective suppression of escalated consumption renders the MCH1-R an attractive target for treatment of alcohol use disorders.


Asunto(s)
Consumo de Bebidas Alcohólicas/fisiopatología , Ingestión de Energía/fisiología , Hormonas Hipotalámicas/fisiología , Melaninas/fisiología , Motivación/fisiología , Hormonas Hipofisarias/fisiología , Receptores de Somatostatina/fisiología , Animales , Ingestión de Alimentos/fisiología , Expresión Génica/efectos de los fármacos , Expresión Génica/fisiología , Hormonas Hipotalámicas/biosíntesis , Hipotálamo/metabolismo , Masculino , Melaninas/biosíntesis , Núcleo Accumbens/metabolismo , Hormonas Hipofisarias/biosíntesis , Pirimidinonas/farmacología , Ratas , Receptores de Somatostatina/antagonistas & inhibidores , Autoadministración , Tiofenos/farmacología
7.
Hepatology ; 64(4): 1086-104, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27387967

RESUMEN

UNLABELLED: The opioid system is widely known to modulate the brain reward system and thus affect the behavior of humans and other animals, including feeding. We hypothesized that the hypothalamic opioid system might also control energy metabolism in peripheral tissues. Mice lacking the kappa opioid receptor (κOR) and adenoviral vectors overexpressing or silencing κOR were stereotaxically delivered in the lateral hypothalamic area (LHA) of rats. Vagal denervation was performed to assess its effect on liver metabolism. Endoplasmic reticulum (ER) stress was inhibited by pharmacological (tauroursodeoxycholic acid) and genetic (overexpression of the chaperone glucose-regulated protein 78 kDa) approaches. The peripheral effects on lipid metabolism were assessed by histological techniques and western blot. We show that in the LHA κOR directly controls hepatic lipid metabolism through the parasympathetic nervous system, independent of changes in food intake and body weight. κOR colocalizes with melanin concentrating hormone receptor 1 (MCH-R1) in the LHA, and genetic disruption of κOR reduced melanin concentrating hormone-induced liver steatosis. The functional relevance of these findings was given by the fact that silencing of κOR in the LHA attenuated both methionine choline-deficient, diet-induced and choline-deficient, high-fat diet-induced ER stress, inflammation, steatohepatitis, and fibrosis, whereas overexpression of κOR in this area promoted liver steatosis. Overexpression of glucose-regulated protein 78 kDa in the liver abolished hypothalamic κOR-induced steatosis by reducing hepatic ER stress. CONCLUSIONS: This study reveals a novel hypothalamic-parasympathetic circuit modulating hepatic function through inflammation and ER stress independent of changes in food intake or body weight; these findings might have implications for the clinical use of opioid receptor antagonists. (Hepatology 2016;64:1086-1104).


Asunto(s)
Dieta , Estrés del Retículo Endoplásmico , Hormonas Hipotalámicas/fisiología , Hipotálamo/fisiología , Hepatopatías/etiología , Melaninas/fisiología , Hormonas Hipofisarias/fisiología , Receptores Opioides kappa/fisiología , Animales , Inflamación/complicaciones , Inflamación/etiología , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley
8.
Fish Physiol Biochem ; 42(3): 883-93, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26690629

RESUMEN

Melanin-concentrating hormone (MCH) is a crucial neuropeptide involved in various biological functions in both mammals and fish. In this study, the full-length MCH cDNA was obtained from Schizothorax prenanti by rapid amplification of cDNA ends polymerase chain reaction. The full-length MCH cDNA contained 589 nucleotides including an open reading frame of 375 nucleotides encoding 256 amino acids. MCH mRNA was highly expressed in the brain by real-time quantitative PCR analysis. Within the brain, expression of MCH mRNA was preponderantly detected in the hypothalamus. In addition, the MCH mRNA expression in the S. prenanti hypothalamus of fed group was significantly decreased compared with the fasted group at 1 and 3 h post-feeding, respectively. Furthermore, the MCH gene expression presented significant increase in the hypothalamus of fasted group compared with the fed group during long-term fasting. After re-feeding, there was a dramatic decrease in MCH mRNA expression in the hypothalamus of S. prenanti. The results indicate that the expression of MCH is affected by feeding status. Taken together, our results suggest that MCH may be involved in food intake regulation in S. prenanti.


Asunto(s)
Cyprinidae , Ingestión de Alimentos/genética , Ayuno/fisiología , Proteínas de Peces , Hormonas Hipotalámicas , Melaninas , Hormonas Hipofisarias , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Clonación Molecular , Cyprinidae/genética , Cyprinidae/fisiología , ADN Complementario/genética , Femenino , Proteínas de Peces/genética , Proteínas de Peces/fisiología , Hormonas Hipotalámicas/genética , Hormonas Hipotalámicas/fisiología , Hipotálamo/metabolismo , Masculino , Melaninas/genética , Melaninas/fisiología , Hormonas Hipofisarias/genética , Hormonas Hipofisarias/fisiología , ARN Mensajero/metabolismo
9.
Hippocampus ; 25(11): 1361-73, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25808129

RESUMEN

The cognitive role of melanin-concentrating hormone (MCH) neurons, a neuronal population located in the mammalian postero-lateral hypothalamus sending projections to all cortical areas, remains poorly understood. Mainly activated during paradoxical sleep (PS), MCH neurons have been implicated in sleep regulation. The genetic deletion of the only known MCH receptor in rodent leads to an impairment of hippocampal dependent forms of memory and to an alteration of hippocampal long-term synaptic plasticity. By using MCH/ataxin3 mice, a genetic model characterized by a selective deletion of MCH neurons in the adult, we investigated the role of MCH neurons in hippocampal synaptic plasticity and hippocampal-dependent forms of memory. MCH/ataxin3 mice exhibited a deficit in the early part of both long-term potentiation and depression in the CA1 area of the hippocampus. Post-tetanic potentiation (PTP) was diminished while synaptic depression induced by repetitive stimulation was enhanced suggesting an alteration of pre-synaptic forms of short-term plasticity in these mice. Behaviorally, MCH/ataxin3 mice spent more time and showed a higher level of hesitation as compared to their controls in performing a short-term memory T-maze task, displayed retardation in acquiring a reference memory task in a Morris water maze, and showed a habituation deficit in an open field task. Deletion of MCH neurons could thus alter spatial short-term memory by impairing short-term plasticity in the hippocampus. Altogether, these findings could provide a cellular mechanism by which PS may facilitate memory encoding. Via MCH neuron activation, PS could prepare the day's learning by increasing and modulating short-term synaptic plasticity in the hippocampus.


Asunto(s)
Conducta Animal/fisiología , Región CA1 Hipocampal/fisiología , Hormonas Hipotalámicas/fisiología , Hipotálamo/citología , Melaninas/fisiología , Memoria a Corto Plazo/fisiología , Plasticidad Neuronal/fisiología , Neuronas/fisiología , Hormonas Hipofisarias/fisiología , Sueño REM/fisiología , Animales , Ataxina-3/genética , Hormonas Hipotalámicas/genética , Hipotálamo/metabolismo , Melaninas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Hormonas Hipofisarias/genética
16.
J Neurosci ; 33(25): 10257-63, 2013 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-23785141

RESUMEN

Melanin concentrating hormone (MCH) is a cyclic neuropeptide present in the hypothalamus of all vertebrates. MCH is implicated in a number of behaviors but direct evidence is lacking. To selectively stimulate the MCH neurons the gene for the light-sensitive cation channel, channelrhodopsin-2, was inserted into the MCH neurons of wild-type mice. Three weeks later MCH neurons were stimulated for 1 min every 5 min for 24 h. A 10 Hz stimulation at the start of the night hastened sleep onset, reduced length of wake bouts by 50%, increased total time in non-REM and REM sleep at night, and increased sleep intensity during the day cycle. Sleep induction at a circadian time when all of the arousal neurons are active indicates that MCH stimulation can powerfully counteract the combined wake-promoting signal of the arousal neurons. This could be potentially useful in treatment of insomnia.


Asunto(s)
Hormonas Hipotalámicas/genética , Hormonas Hipotalámicas/fisiología , Melaninas/genética , Melaninas/fisiología , Neuronas/fisiología , Hormonas Hipofisarias/genética , Hormonas Hipofisarias/fisiología , Sueño/fisiología , Animales , Channelrhodopsins , Ritmo Circadiano/fisiología , Color , Ritmo Delta/fisiología , Electrodos Implantados , Electroencefalografía , Hipotálamo/fisiología , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Técnicas de Placa-Clamp , Estimulación Luminosa , Plásmidos/genética , Sueño REM/fisiología , Vigilia/fisiología
17.
Sleep Med Rev ; 17(4): 293-8, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23477948

RESUMEN

The melanin-concentrating hormone (MCH) is a 19 aminoacid peptide found in mammals predominantly in neurons located in the lateral hypothalamus and incerto-hypothalamic area. The biological function of MCH is mediated by two G-protein-coupled receptors known as MCHR1 and MCHR2, although the latter is expressed only in carnivores, primates and man. The MCHR1 couples to Gi, Gq and Go proteins, with Gi leading to the inhibition of both excitatory and inhibitory synaptic events. Within the central nervous system (CNS) MCH participates in a number of functions including sleep-wake behavior. In this respect, MCHergic neurons project widely throughout the CNS to brain regions involved in the regulation of behavioral states. MCHergic neurons are silent during wakefulness (W), increase their firing during slow wave sleep (SWS) and still more during REM sleep (REMS). Studies in knockout mice for MCH (MCH(-/-)) have shown a reduction in SWS and an increase of W during the light and the dark phase of the light-dark cycle. Moreover, in response to food deprivation a marked reduction in REMS time was observed in these animals. Conflicting effects on sleep variables have been reported in MCHR1(-/-) mice by different authors. The i.c.v. administration of MCH increases REMS and SWS in the rat. In addition, an enhancement of REMS has been described following the microinjection of the neuropeptide into the nucleus pontis oralis of the cat, while its infusion into the dorsal raphe nucleus (DR) and the basal forebrain (horizontal limb of the diagonal band of Broca) is followed by an increase of REMS and a reduction of W in the rat. Immunoneutralization of MCH in the DR augmented W and suppressed REMS in the rat, as did the s.c. injection of selective MCHR1 antagonists. The robust REMS-inducing effect of MCH is likely related to the deactivation of monoaminergic, orexinergic, glutamatergic, cholinergic (W-on) and GABAergic (REM-off) neurons involved in the generation of W and the inhibition of REMS. On the basis of preclinical studies, it can be proposed that selective MCHR1 receptor agonists could constitute potential therapeutic modalities in the arsenal of insomnia pharmacotherapy. Due to the lack of adequate animal models, the role of the MCHR2 on sleep is still unknown.


Asunto(s)
Hormonas Hipotalámicas/fisiología , Melaninas/fisiología , Hormonas Hipofisarias/fisiología , Sueño/fisiología , Vigilia/fisiología , Animales , Encéfalo/fisiología , Gatos , Humanos , Hipotálamo/fisiología , Ratones , Neurotransmisores/fisiología , Fases del Sueño/fisiología
18.
J Neuroendocrinol ; 24(6): 930-43, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22295972

RESUMEN

Interleukin (IL)-6 deficient mice develop mature-onset obesity. Furthermore, i.c.v. administration of IL-6 increases energy expenditure, suggesting that IL-6 centrally regulates energy homeostasis. To investigate whether it would be possible for IL-6 to directly influence the energy homeostasis via hypothalamic regulation in humans and rodents, we mapped the distribution of the ligand binding IL-6 receptor α (IL-6Rα) in this brain region. In the human hypothalamus, IL-6Rα-immunoreactivity was detected in perikarya and first-order dendrites of neurones. The IL-6Rα-immunoreactive (-IR) neurones were observed posterior to the level of the interventricular foramen. There, IL-6Rα-IR neurones were located in the lateral hypothalamic, perifornical, dorsal and posterior hypothalamic areas, the hypothalamic dorsomedial nucleus and in the zona incerta. In the caudal part of the hypothalamus, the density of the IL-6Rα-IR neurones gradually increased. Double-labelling immunofluorescent studies demonstrated that IL-6Rα immunoreactivity was localised in the same neurones as the orexigenic neuropeptide, melanin-concentrating hormone (MCH). By contrast, IL-6Rα-immunoreactivity was not observed in the orexin B-IR neurones. To determine whether the observed expression of IL-6Rα is evolutionary conserved, we studied the co-localisation of IL-6Rα with MCH and orexin in the mouse hypothalamus, where IL-6Rα-immunoreactivity was present in numerous MCH-IR and orexin-IR neurones. Our data demonstrate that the MCH neurones of the human hypothalamus, as well as the MCH and orexin neurones of the mouse hypothalamus, contain IL-6Rα. This opens up the possibility that IL-6 influences the energy balance through the MCH neurones in humans, and both MCH and orexin neurones in mice.


Asunto(s)
Hormonas Hipotalámicas/metabolismo , Hipotálamo/metabolismo , Melaninas/metabolismo , Hormonas Hipofisarias/metabolismo , Receptores de Interleucina-6/metabolismo , Adulto , Animales , Humanos , Hormonas Hipotalámicas/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Melaninas/fisiología , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Neuropéptidos/metabolismo , Orexinas , Hormonas Hipofisarias/fisiología
19.
Am J Physiol Regul Integr Comp Physiol ; 302(7): R815-24, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22262306

RESUMEN

Neurons that synthesize melanin-concentrating hormone (MCH) colocalize GABA, regulate energy homeostasis, modulate water intake, and influence anxiety, stress, and social interaction. Similarly, vasopressin and oxytocin can influence the same behaviors and states, suggesting that these neuropeptides may exert part of their effect by modulating MCH neurons. Using whole cell recording in MCH-green fluorescent protein (GFP) transgenic mouse hypothalamic brain slices, we found that both vasopressin and oxytocin evoked a substantial excitatory effect. Both peptides reversibly increased spike frequency and depolarized the membrane potential in a concentration-dependent and tetrodotoxin-resistant manner, indicating a direct effect. Substitution of lithium for extracellular sodium, Na(+)/Ca(2+) exchanger blockers KB-R7943 and SN-6, and intracellular calcium chelator BAPTA, all substantially reduced the vasopressin-mediated depolarization, suggesting activation of the Na(+)/Ca(2+) exchanger. Vasopressin reduced input resistance, and the vasopressin-mediated depolarization was attenuated by SKF-96265, suggesting a second mechanism based on opening nonselective cation channels. Neither vasopressin nor oxytocin showed substantial excitatory actions on lateral hypothalamic inhibitory neurons identified in a glutamate decarboxylase 67 (GAD67)-GFP mouse. The primary vasopressin receptor was vasopressin receptor 1a (V1aR), as suggested by the excitation by V1aR agonist [Arg(8)]vasotocin, the selective V1aR agonist [Phe(2)]OVT and by the presence of V1aR mRNA in MCH cells, but not in other nearby GABA cells, as detected with single-cell RT-PCR. Oxytocin receptor mRNA was also detected in MCH neurons. Together, these data suggest that vasopressin or oxytocin exert a minimal effect on most GABA neurons in the lateral hypothalamus but exert a robust excitatory effect on presumptive GABA cells that contain MCH. Thus, some of the central actions of vasopressin and oxytocin may be mediated through MCH cells.


Asunto(s)
Arginina Vasopresina/fisiología , Neuronas GABAérgicas/fisiología , Hormonas Hipotalámicas/fisiología , Hipotálamo/fisiología , Melaninas/fisiología , Oxitocina/fisiología , Hormonas Hipofisarias/fisiología , Animales , Arginina Vasopresina/agonistas , Arginina Vasopresina/farmacología , Compuestos de Bencilo/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio/efectos de los fármacos , Quelantes/farmacología , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Neuronas GABAérgicas/efectos de los fármacos , Glutamato Descarboxilasa/genética , Glutamato Descarboxilasa/fisiología , Hipotálamo/efectos de los fármacos , Imidazoles/farmacología , Canales Iónicos/efectos de los fármacos , Litio/farmacología , Potenciales de la Membrana/efectos de los fármacos , Ratones , Ratones Transgénicos , Oxitocina/farmacología , Receptores de Oxitocina/fisiología , Receptores de Vasopresinas/agonistas , Receptores de Vasopresinas/fisiología , Intercambiador de Sodio-Calcio/antagonistas & inhibidores , Intercambiador de Sodio-Calcio/fisiología , Tiazolidinas/farmacología , Tiourea/análogos & derivados , Tiourea/farmacología
20.
Mol Cell Endocrinol ; 348(1): 281-96, 2012 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-21945816

RESUMEN

In order to better understand the role of melanin-concentrating hormone (MCH) in the regulation of appetite in fish, the mRNAs of two forms of MCH, prepro-MCH and MCH2, and two forms of MCH receptors, MCH-R1 and MCH-R2, were isolated from winter flounder (Pseudopleuronectes americanus). In addition, the mRNA expressions of these peptides and their receptors were determined under fed and fasted conditions. Both MCHs are expressed in forebrain and midbrain, as well as peripheral tissues including gut and gonads. Both MCH-Rs are ubiquitously expressed in the brain and periphery. Fasting induced an increase in the expression levels of MCH and MCH-R1 mRNAs in optic tectum/thalamus and hypothalamus but had no effect on either MCH2 or MCH-R2 mRNA expressions. Our results suggest that MCH and MCH-R1, but not MCH2 and MCH-R2 might have a role in the regulation of appetite in flounder.


Asunto(s)
Regulación del Apetito , Proteínas de Peces/fisiología , Lenguado/fisiología , Hormonas Hipotalámicas/fisiología , Melaninas/fisiología , Hormonas Hipofisarias/fisiología , Receptores de la Hormona Hipofisaria/fisiología , Secuencia de Aminoácidos , Animales , Encéfalo/metabolismo , Femenino , Proteínas de Peces/genética , Proteínas de Peces/metabolismo , Lenguado/genética , Lenguado/metabolismo , Privación de Alimentos , Expresión Génica , Hormonas Hipotalámicas/genética , Hormonas Hipotalámicas/metabolismo , Hipotálamo/metabolismo , Masculino , Melaninas/genética , Melaninas/metabolismo , Datos de Secuencia Molecular , Especificidad de Órganos , Filogenia , Hormonas Hipofisarias/genética , Hormonas Hipofisarias/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de la Hormona Hipofisaria/genética , Receptores de la Hormona Hipofisaria/metabolismo , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA