Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 10(1): 18025, 2020 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-33093538

RESUMEN

Sleep disturbances are commonly found in trauma-exposed populations. Additionally, trauma exposure results in fear-associated memory impairments. Given the interactions of sleep with learning and memory, we hypothesized that increasing sleep duration following trauma exposure would restore overall function and improve trauma-induced fear-associated memory dysfunction. Here, we utilized single prolonged stress, a validated rodent model of post-traumatic stress disorder, in combination with optogenetic activation of hypothalamic melanin-concentrating hormone containing cells to increase sleep duration. The goal of this work was to ascertain if post-trauma sleep increases are sufficient to improve fear-associated memory function. In our laboratory, optogenetic stimulation after trauma exposure was sufficient to increase REM sleep duration during both the Light and Dark Phase, whereas NREM sleep duration was only increased during the Dark Phase of the circadian day. Interestingly though, animals that received optogenetic stimulation showed significantly improved fear-associated memory processing compared to non-stimulated controls. These results suggest that sleep therapeutics immediately following trauma exposure may be beneficial and that post-trauma sleep needs to be further examined in the context of the development of post-traumatic stress disorder.


Asunto(s)
Miedo , Hormonas Hipotalámicas/administración & dosificación , Melaninas/administración & dosificación , Trastornos de la Memoria/prevención & control , Optogenética , Hormonas Hipofisarias/administración & dosificación , Trastornos del Sueño-Vigilia/prevención & control , Sueño REM , Trastornos por Estrés Postraumático/complicaciones , Animales , Masculino , Consolidación de la Memoria , Trastornos de la Memoria/etiología , Ratas , Ratas Long-Evans , Trastornos del Sueño-Vigilia/etiología
2.
J Psychopharmacol ; 34(4): 478-489, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31909693

RESUMEN

BACKGROUND: Identifying neural substrates that are differentially affected by drugs of abuse and natural rewards is key to finding a target for an efficacious treatment for substance abuse. Melanin-concentrating hormone is a polypeptide with an inhibitory effect on the mesolimbic dopamine system. Here we test the hypothesis that melanin-concentrating hormone in the lateral hypothalamus and nucleus accumbens shell is differentially involved in the regulation of morphine and food-rewarded behaviors. METHODS: Male Sprague-Dawley rats were trained with morphine (5.0 mg/kg, subcutaneously) or food pellets (standard chow, 10-14 g) to induce a conditioned place preference, immediately followed by extinction training. Melanin-concentrating hormone (1.0 µg/side) or saline was infused into the nucleus accumbens shell or lateral hypothalamus before the reinstatement primed by morphine or food, and locomotor activity was simultaneously monitored. As the comparison, melanin-concentrating hormone was also microinjected into the nucleus accumbens shell or lateral hypothalamus before the expression of food or morphine-induced conditioned place preference. RESULTS: Microinfusion of melanin-concentrating hormone into the nucleus accumbens shell (but not into the lateral hypothalamus) prevented the reinstatement of morphine conditioned place preference but had no effect on the reinstatement of food conditioned place preference. In contrast, microinfusion of melanin-concentrating hormone into the lateral hypothalamus (but not in the nucleus accumbens shell) inhibited the reinstatement of food conditioned place preference but had no effect on the reinstatement of morphine conditioned place preference. CONCLUSIONS: These results suggest a clear double dissociation of melanin-concentrating hormone in morphine/food rewarding behaviors and melanin-concentrating hormone in the nucleus accumbens shell. Melanin-concentrating hormone could be a potential target for therapeutic intervention for morphine abuse without affecting natural rewards.


Asunto(s)
Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Área Hipotalámica Lateral/metabolismo , Hormonas Hipotalámicas/farmacología , Melaninas/farmacología , Morfina/farmacología , Núcleo Accumbens/metabolismo , Hormonas Hipofisarias/farmacología , Animales , Condicionamiento Operante/efectos de los fármacos , Hormonas Hipotalámicas/administración & dosificación , Masculino , Melaninas/administración & dosificación , Microinyecciones , Hormonas Hipofisarias/administración & dosificación , Ratas , Ratas Sprague-Dawley , Recompensa
3.
Mol Neurobiol ; 56(12): 8076-8086, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31183806

RESUMEN

Melanin-concentrating hormone (MCH) is a highly conserved neuropeptide known to exhibit important functions in the brain. Some studies have reported that MCH improves memory by promoting memory retention. However, the precise molecular mechanisms by which MCH enhances memory impairment have yet to be fully elucidated. In this study, MCH was administered to the scopolamine-induced memory-impaired mice via the nasal cavity to examine the acute effects of MCH and Alzheimer's disease (AD) mouse models to evaluate the chronic effects of MCH. MCH improved memory impairment in both models and reduced soluble amyloid beta in the cerebral cortex of APP/PS1 transgenic mice. In vitro assays also showed that MCH inhibits amyloid beta-induced cytotoxicity. Furthermore, MCH increased long-term potentiation (LTP) in the hippocampus of wild-type and 5XFAD AD mouse model. To further elucidate the mechanisms of the chronic effect of MCH, the levels of phosphorylated CREB and GSK3ß, and the expression of BDNF, TrkB and PSD95 were examined in the cerebral cortex and hippocampus. Our findings indicate that MCH might have neuroprotective effects via downstream pathways associated with the enhancement of neuronal synapses and LTP. This suggests a therapeutic potential of MCH for the treatment of neurodegenerative diseases such as AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Modelos Animales de Enfermedad , Hormonas Hipotalámicas/administración & dosificación , Melaninas/administración & dosificación , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/metabolismo , Hormonas Hipofisarias/administración & dosificación , Administración Intranasal , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Cavidad Nasal/efectos de los fármacos , Cavidad Nasal/metabolismo , Embarazo
5.
Neuropeptides ; 74: 70-81, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30642579

RESUMEN

Melanin-concentrating hormone (MCH) is a neuropeptide present in neurons located in the hypothalamus that densely innervate serotonergic cells in the dorsal raphe nucleus (DRN). MCH administration into the DRN induces a depressive-like effect through a serotonergic mechanism. To further understand the interaction between MCH and serotonin, we used primary cultured serotonergic neurons to evaluate the effect of MCH on serotonergic release and metabolism by HPLC-ED measurement of serotonin (5-HT) and 5-hydroxyindolacetic acid (5-HIAA) levels. We confirmed the presence of serotonergic neurons in the E14 rat rhombencephalon by immunohistochemistry and showed for the first time evidence of MCHergic fibers reaching the area. Cultures obtained from rhombencephalic tissue presented 2.2 ±â€¯0.7% of serotonergic and 48.9 ±â€¯5.4% of GABAergic neurons. Despite the low concentration of serotonergic neurons, we were able to measure basal cellular and extracellular levels of 5-HT and 5-HIAA without the addition of any serotonergic-enhancer drug. As expected, 5-HT release was calcium-dependent and induced by depolarization. 5-HT extracellular levels were significantly increased by incubation with serotonin reuptake inhibitors (citalopram and nortriptyline) and a monoamine-oxidase inhibitor (clorgyline), and were not significantly modified by a 5-HT1A autoreceptor agonist (8-OHDPAT). Even though serotonergic cells responded as expected to these pharmacological treatments, MCH did not induce significant modifications of 5-HT and 5-HIAA extracellular levels in the cultures. Despite this unexpected result, we consider that assessment of 5-HT and 5-HIAA levels in primary serotonergic cultures may be an adequate approach to study the effect of other drugs and modulators on serotonin release, uptake and turnover.


Asunto(s)
Hormonas Hipotalámicas/metabolismo , Hipotálamo/metabolismo , Melaninas/metabolismo , Neuronas/metabolismo , Hormonas Hipofisarias/metabolismo , Núcleos del Rafe/metabolismo , Serotonina/metabolismo , Animales , Neuronas GABAérgicas/citología , Hormonas Hipotalámicas/administración & dosificación , Hipotálamo/citología , Melaninas/administración & dosificación , Vías Nerviosas/citología , Vías Nerviosas/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Hormonas Hipofisarias/administración & dosificación , Cultivo Primario de Células , Núcleos del Rafe/citología , Núcleos del Rafe/efectos de los fármacos , Ratas Sprague-Dawley , Receptor de Serotonina 5-HT1A/metabolismo
6.
Sci Rep ; 8(1): 707, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29335480

RESUMEN

The melanin-concentrating hormone (MCH) is a peptidergic neuromodulator synthesized by neurons in the lateral hypothalamus and zona incerta. MCHergic neurons project throughout the central nervous system, indicating the involvements of many physiological functions, but the role in pain has yet to be determined. In this study, we found that pMCH-/- mice showed lower baseline pain thresholds to mechanical and thermal stimuli than did pMCH+/+ mice, and the time to reach the maximum hyperalgesic response was also significantly earlier in both inflammatory and neuropathic pain. To examine its pharmacological properties, MCH was administered intranasally into mice, and results indicated that MCH treatment significantly increased mechanical and thermal pain thresholds in both pain models. Antagonist challenges with naltrexone (opioid receptor antagonist) and AM251 (cannabinoid 1 receptor antagonist) reversed the analgesic effects of MCH in both pain models, suggesting the involvement of opioid and cannabinoid systems. MCH treatment also increased the expression and activation of CB1R in the medial prefrontal cortex and dorsolateral- and ventrolateral periaqueductal grey. The MCH1R antagonist abolished the effects induced by MCH. This is the first study to suggest novel analgesic actions of MCH, which holds great promise for the application of MCH in the therapy of pain-related diseases.


Asunto(s)
Analgésicos/farmacología , Hormonas Hipotalámicas/farmacología , Melaninas/farmacología , Percepción del Dolor/efectos de los fármacos , Hormonas Hipofisarias/farmacología , Administración Intranasal , Analgésicos/administración & dosificación , Animales , Hormonas Hipotalámicas/administración & dosificación , Hormonas Hipotalámicas/deficiencia , Melaninas/administración & dosificación , Melaninas/deficiencia , Ratones , Ratones Noqueados , Hormonas Hipofisarias/administración & dosificación , Hormonas Hipofisarias/deficiencia
7.
Neuropharmacology ; 130: 62-70, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29191753

RESUMEN

Melanin-Concentrating Hormone (MCH) is one of the most relevant orexigenic factors specifically located in the lateral hypothalamic area (LHA), with its physiological relevance demonstrated in studies using several genetically manipulated mice models. However, the central mechanisms controlling MCH-induced hyperphagia remain largely uncharacterized. Here, we show that central injection of MCH in mice deficient for kappa opoid receptor (k-OR) failed to stimulate feeding. To determine the hypothalamic area responsible for this MCH/k-OR interaction, we performed virogenetic studies and found that downregulation of k-OR by adeno-associated viruses (shOprk1-AAV) in LHA, but not in other hypothalamic nuclei, was sufficient to block MCH-induced food intake. Next, we sought to investigate the molecular signaling pathway within the LHA that mediates acute central MCH stimulation of food intake. We found that MCH activates k-OR and that increased levels of phosphorylated extracellular signal regulated kinase (ERK) are associated with downregulation of phospho-S6 Ribosomal Protein. This effect was prevented when a pharmacological inhibitor of k-OR was co-administered with MCH. Finally, the specific activation of the direct upstream regulator of S6 (p70S6K) in the LHA attenuated MCH-stimulated food consumption. Our results reveal that lateral hypothalamic k-OR system modulates the orexigenic action of MCH via the p70S6K/S6 pathway.


Asunto(s)
Ingestión de Alimentos/efectos de los fármacos , Hormonas Hipotalámicas/administración & dosificación , Melaninas/administración & dosificación , Hormonas Hipofisarias/administración & dosificación , Receptores Opioides kappa/antagonistas & inhibidores , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Animales , Depresores del Apetito/administración & dosificación , Depresores del Apetito/metabolismo , Dependovirus , Área Hipotalámica Lateral/efectos de los fármacos , Área Hipotalámica Lateral/metabolismo , Hormonas Hipotalámicas/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Melaninas/metabolismo , Ratones , Ratones Endogámicos C57BL , Hormonas Hipofisarias/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Opioides kappa/metabolismo , Proteínas Quinasas S6 Ribosómicas/efectos de los fármacos , Proteínas Quinasas S6 Ribosómicas/metabolismo
8.
Artículo en Inglés | MEDLINE | ID: mdl-28579534

RESUMEN

Reproduction in captivity is a key study issue in Anguilla anguilla as a possible solution for its dwindling population. Understanding the mechanisms controlling the production of ribosomal building blocks during artificially induced oocyte maturation could be particularly interesting. Transcription levels of ribosomal biogenesis associated genes could be used as markers to monitor oogenesis. Eels from the Albufera Lagoon were injected with carp pituitary extract for 15weeks and ovaries in previtellogenic (PV) stage (non-injected), in early-, mid-, late-vitellogenesis (EV, MV, LV), as well as in migratory nucleus stage (MN) were analysed. 5S rRNA and related genes were highly transcribed in ovaries with PV oocytes. As oocytes developed, transcriptional levels of genes related to 5S rRNA production (gtf3a), accumulation (gtf3a, 42sp43) and nucleocytoplasmic transport (rpl5, rpl11) and the 5S/18S rRNA index decreased (PV>EV>MV>LV>MN). On the contrary, 18S rRNA was at its highest at MN stage while ubtf1 in charge of activating RNA-polymerase I and synthesising 18S rRNA behaved as 5S related genes. Individuals that did not respond (NR) to the treatment showed 5S/18S index values similar to PV females, while studied genes showed EV/LV-like transcription levels. Therefore, NR females fail to express the largest rRNAs, which could thus be taken as markers of successful vitellogenesis progression. In conclusion, we have proved that the transcriptional dynamics of ribosomal genes provides useful tools to characterize induced ovarian development in European eels. In the future, such markers should be studied as putative indicators of response to hormonal treatments and of the quality of obtained eel oocytes.


Asunto(s)
Diferenciación Celular/genética , Anguilas/genética , Oocitos/citología , Oogénesis/fisiología , Hormonas Hipofisarias/administración & dosificación , Animales , Femenino
9.
Peptides ; 87: 28-33, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27845162

RESUMEN

Neuropeptideglutamic acid-isoleucine (NEI) as well as melanin concentrating hormone (MCH) is cleaved from the 165 amino acid protein, prepro-melanin concentrating hormone (prepro-MCH). Among many physiological roles of MCH, we demonstrated that intracerebroventricular (icv) injection of MCH induced increases in REM sleep episodes as well as in non REM sleep episodes. However, there are no studies on the effect of NEI on the sleep-wake cycle. As for the sites of action of MCH for induction of REM sleep, the ventrolateral periaqueductal gray (vlPAG) has been reported to be one of its site of action. Although MCH neurons contain NEI, GABA, MCH, and other neuropeptides, we do not know which transmitter(s) might induce REM sleep by acting on the vlPAG. Thus, we first examined the effect of icv injection of NEI on the sleep-wake cycle, and investigated how microinjection of either NEI, MCH, or GABA into the vlPAG affected REM sleep in rats. Icv injection of NEI (0.61µg/5µl: n=7) significantly increased the time spent in REM episodes compared to control (saline: 5µl; n=6). Microinjection of either NEI (61ng/0.2µl: n=7), MCH (100ng/0.2µl: n=6) or GABA (250mM/0.2µl: n=7) into the vlPAG significantly increased the time spent in REM episodes and the AUC. Precise hourly analysis of REM sleep also revealed that after those microinjections, NEI and MCH increased REM episodes at the latter phase, compared to GABA which increased REM episodes at the earlier phase. This result suggests that NEI and MCH may induce sustained REM sleep, while GABA may initiate REM sleep. In conclusion, our findings demonstrate that NEI, a cleaved peptide from the same precursor, prepro-MCH, as MCH, induce REM sleep at least in part through acting on the vlPAG.


Asunto(s)
Hormonas Hipotalámicas/metabolismo , Melaninas/metabolismo , Neuronas/metabolismo , Neuropéptidos/administración & dosificación , Hormonas Hipofisarias/metabolismo , Sueño REM/efectos de los fármacos , Animales , Ácido Glutámico/administración & dosificación , Ácido Glutámico/metabolismo , Hormonas Hipotalámicas/administración & dosificación , Hormonas Hipotalámicas/química , Isoleucina/administración & dosificación , Isoleucina/metabolismo , Melaninas/administración & dosificación , Melaninas/química , Microinyecciones , Neuronas/efectos de los fármacos , Neuropéptidos/metabolismo , Sustancia Gris Periacueductal/efectos de los fármacos , Sustancia Gris Periacueductal/metabolismo , Sustancia Gris Periacueductal/fisiología , Hormonas Hipofisarias/administración & dosificación , Hormonas Hipofisarias/química , Ratas , Sueño REM/fisiología , Ácido gamma-Aminobutírico/administración & dosificación
10.
Mol Neurobiol ; 54(10): 7706-7721, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27844281

RESUMEN

Acupuncture has shown the therapeutic effect on various neurodegenerative disorders including Parkinson's disease (PD). While investigating the neuroprotective mechanism of acupuncture, we firstly found the novel function of melanin-concentrating hormone (MCH) as a potent neuroprotective candidate. Here, we explored whether hypothalamic MCH mediates the neuroprotective action of acupuncture. In addition, we aimed at evaluating the neuroprotective effects of MCH and elucidating underlying mechanism in vitro and in vivo PD models. First, we tested whether hypothalamic MCH mediates the neuroprotective effects of acupuncture by challenging MCH-R1 antagonist (i.p.) in mice PD model. We also investigated whether MCH has a beneficial role in dopaminergic neuronal protection in vitro primary midbrain and human neuronal cultures and in vivo MPTP-induced, Pitx3-/-, and A53T mutant mice PD models. Transcriptomics followed by quantitative PCR and western blot analyses were performed to reveal the neuroprotective mechanism of MCH. We first found that hypothalamic MCH biosynthesis was directly activated by acupuncture treatment and that administration of an MCH-R1 antagonist reverses the neuroprotective effects of acupuncture. A novel finding is that MCH showed a beneficial role in dopaminergic neuron protection via downstream pathways related to neuronal survival. This is the first study to suggest the novel neuroprotective action of MCH as well as the involvement of hypothalamic MCH in the acupuncture effects in PD, which holds great promise for the application of MCH in the therapy of neurodegenerative diseases.


Asunto(s)
Terapia por Acupuntura/métodos , Hormonas Hipotalámicas/biosíntesis , Melaninas/biosíntesis , Fármacos Neuroprotectores/metabolismo , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/prevención & control , Hormonas Hipofisarias/biosíntesis , Animales , Células Cultivadas , Humanos , Hormonas Hipotalámicas/administración & dosificación , Hormonas Hipotalámicas/antagonistas & inhibidores , Hipotálamo/metabolismo , Masculino , Melaninas/administración & dosificación , Melaninas/antagonistas & inhibidores , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Hormonas Hipofisarias/administración & dosificación , Hormonas Hipofisarias/antagonistas & inhibidores , Resultado del Tratamiento
11.
Neurosci Lett ; 630: 66-69, 2016 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-27461793

RESUMEN

A study was performed on the effects of local microinjection of melanin-concentrating hormone (MCH) into the right sublaterodorsal tegmental nucleus (SLD) on sleep and wakefulness in rats prepared for chronic sleep recordings. MCH 200ng significantly decreased rapid-eye-movement sleep (REMS) time during the first and second 2-h of the recording period which was related to the reduction of the number of REMS periods and the increase of REMS latency. It is proposed that REMS inhibition was related to the direct deactivation of SLD glutamatergic neurons by the peptide.


Asunto(s)
Hormonas Hipotalámicas/administración & dosificación , Hormonas Hipotalámicas/fisiología , Melaninas/administración & dosificación , Melaninas/fisiología , Hormonas Hipofisarias/administración & dosificación , Hormonas Hipofisarias/fisiología , Sueño REM , Tegmento Mesencefálico/fisiología , Vigilia , Animales , Electroencefalografía , Masculino , Microinyecciones , Neuronas/fisiología , Lóbulo Occipital/fisiología , Ratas , Ratas Wistar
12.
Brain Res ; 1598: 114-28, 2015 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-25541366

RESUMEN

Hypothalamic neurons that utilize melanin-concentrating hormone (MCH) as a neuromodulator are localized in the postero-lateral hypothalamus and incerto-hypothalamic area. These neurons send dense projections to the dorsal raphe nucleus (DRN). Serotonergic neurons of the DRN are involved in the control of sleep and play a critical role in major depression. Previously, we demonstrated that microinjections of MCH into the DRN resulted in an increase in REM sleep and produce a depressive-like effect. In the present study we examined the mechanisms that mediate these effects by employing neuroanatomical and electrophysiological techniques. First, we determined that rhodamine-labeled MCH (R-MCH), when microinjected into the lateral ventricle, is internalized in serotonergic and non-serotonergic DRN neurons in rats and cats. These data strongly suggest that these neurons express MCHergic receptors. Second, in rats, we demonstrated that the microinjection of MCH into the lateral ventricle results in a significant decrease in the firing rate in 59% of the neurons recorded in the DRN; the juxtacellular administration of MCH reduced the discharge in 80% of these neurons. Some of the neurons affected by MCH were likely serotonergic on the basis of their electrophysiological and pharmacological properties. We conclude that MCH reduces the activity of serotonergic neurons of the DRN. These and previous data suggest that the MCHergic modulation of serotonergic activity within the DRN is involved in the regulation of REM sleep as well as in the pathophysiology of depressive disorders.


Asunto(s)
Núcleo Dorsal del Rafe/efectos de los fármacos , Hormonas Hipotalámicas/administración & dosificación , Melaninas/administración & dosificación , Neuronas/efectos de los fármacos , Hormonas Hipofisarias/administración & dosificación , Potenciales de Acción/efectos de los fármacos , Animales , Gatos , Núcleo Dorsal del Rafe/fisiología , Glutamato Descarboxilasa/metabolismo , Inmunohistoquímica , Microelectrodos , Microinyecciones , Neuronas/fisiología , Fotomicrografía , Ratas Wistar , Rodaminas
13.
Peptides ; 50: 96-9, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24126282

RESUMEN

An emerging body of evidence involves the hypothalamic neuropeptide melanin-concentrating hormone (MCH) in the regulation of emotional states. We have reported a pro-depressive effect induced by MCH after its microinjection into the dorsal raphe nucleus (DR) evaluated in the forced swimming test (FST) in rats. Here we extended this study to the median raphe nucleus (MnR). Firstly, the presence of MCH-containing fibers in the rat MnR was analyzed by means of immunohistochemistry. Secondly, the behavioral effect induced by the microinjection of MCH into the MnR was assessed using the FST. Morphological results showed a large density of MCHergic fibers within the MnR. Behavioral results indicated that 100 ng of MCH (but not 50 ng) significantly increased the immobility time and decreased the swimming time, demonstrating a depressive-like effect. In contrast, climbing behavior was not significantly affected. Present findings revealed that the MnR neurons participate in the MCHergic control of affective-related behavioral responses. However, the behavioral patterns induced by MCH in the MnR and DR were different. This could be explained by anatomical and physiological differences between both nuclei.


Asunto(s)
Conducta Animal/efectos de los fármacos , Hormonas Hipotalámicas/administración & dosificación , Melaninas/administración & dosificación , Hormonas Hipofisarias/administración & dosificación , Núcleos del Rafe/efectos de los fármacos , Animales , Depresión Química , Inmunohistoquímica , Masculino , Microinyecciones , Especificidad de Órganos , Núcleos del Rafe/anatomía & histología , Núcleos del Rafe/fisiología , Ratas , Ratas Wistar , Natación/psicología
14.
Peptides ; 39: 11-5, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23123302

RESUMEN

The ventrolateral preoptic area (VLPO) has been recognized as one of the key structures responsible for the generation of non-REM (NREM) sleep. The melanin-concentrating hormone (MCH)-containing neurons, which are located in the lateral hypothalamus and incerto-hypothalamic area, project widely throughout the central nervous system and include projections to the VLPO. The MCH has been associated with the central regulation of feeding and energy homeostasis. In addition, recent findings strongly suggest that the MCHergic system promotes sleep. The aim of the present study was to determine if MCH generates sleep by regulating VLPO neuronal activity. To this purpose, we characterized the effect of unilateral and bilateral microinjections of MCH into the VLPO on sleep and wakefulness in the rat. Unilateral administration of MCH into the VLPO and adjacent dorsal preoptic area did not modify sleep. On the contrary, bilateral microinjections of MCH (100 ng) into these areas significantly increased light sleep (LS, 39.2±4.8 vs. 21.6±2.5 min, P<0.05) and total NREM sleep (142.4±23.2 vs. 86.5±10.5 min, P<0.05) compared to control (saline) microinjections. No effect was observed on REM sleep. We conclude that MCH administration into the VLPO and adjacent dorsal lateral preoptic area promotes the generation of NREM sleep.


Asunto(s)
Hormonas Hipotalámicas/fisiología , Melaninas/fisiología , Hormonas Hipofisarias/fisiología , Área Preóptica/fisiología , Sueño REM , Animales , Hormonas Hipotalámicas/administración & dosificación , Masculino , Melaninas/administración & dosificación , Microinyecciones , Hormonas Hipofisarias/administración & dosificación , Ratas , Ratas Wistar
15.
Gastroenterology ; 144(3): 636-649.e6, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23142626

RESUMEN

BACKGROUND & AIMS: Specific neuronal circuits modulate autonomic outflow to liver and white adipose tissue. Melanin-concentrating hormone (MCH)-deficient mice are hypophagic, lean, and do not develop hepatosteatosis when fed a high-fat diet. Herein, we sought to investigate the role of MCH, an orexigenic neuropeptide specifically expressed in the lateral hypothalamic area, on hepatic and adipocyte metabolism. METHODS: Chronic central administration of MCH and adenoviral vectors increasing MCH signaling were performed in rats and mice. Vagal denervation was performed to assess its effect on liver metabolism. The peripheral effects on lipid metabolism were assessed by real-time polymerase chain reaction and Western blot. RESULTS: We showed that the activation of MCH receptors promotes nonalcoholic fatty liver disease through the parasympathetic nervous system, whereas it increases fat deposition in white adipose tissue via the suppression of sympathetic traffic. These metabolic actions are independent of parallel changes in food intake and energy expenditure. In the liver, MCH triggers lipid accumulation and lipid uptake, with c-Jun N-terminal kinase being an essential player, whereas in adipocytes MCH induces metabolic pathways that promote lipid storage and decreases lipid mobilization. Genetic activation of MCH receptors or infusion of MCH specifically in the lateral hypothalamic area modulated hepatic lipid metabolism, whereas the specific activation of this receptor in the arcuate nucleus affected adipocyte metabolism. CONCLUSIONS: Our findings show that central MCH directly controls hepatic and adipocyte metabolism through different pathways.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Adiposidad/fisiología , Área Hipotalámica Lateral/fisiología , Hormonas Hipotalámicas/fisiología , Hígado/metabolismo , Melaninas/fisiología , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Hormonas Hipofisarias/fisiología , Adipocitos/efectos de los fármacos , Tejido Adiposo/efectos de los fármacos , Animales , Ingestión de Alimentos , Ácidos Grasos/metabolismo , Hígado Graso/metabolismo , Hígado Graso/fisiopatología , Área Hipotalámica Lateral/efectos de los fármacos , Hormonas Hipotalámicas/administración & dosificación , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/fisiología , Lipogénesis/efectos de los fármacos , Lipogénesis/fisiología , Hígado/efectos de los fármacos , Masculino , Melaninas/administración & dosificación , Ratones , Enfermedad del Hígado Graso no Alcohólico , Hormonas Hipofisarias/administración & dosificación , Ratas , Ratas Sprague-Dawley , Receptores de la Hormona Hipofisaria/agonistas , Receptores de la Hormona Hipofisaria/fisiología , Nervio Vago/efectos de los fármacos , Nervio Vago/fisiología , Nervio Vago/fisiopatología
16.
Am J Physiol Regul Integr Comp Physiol ; 303(6): R624-32, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22814666

RESUMEN

Melanin-concentrating hormone (MCH) is a neuropeptide that acts to increase feeding behavior and decrease energy expenditure. The role of MCH in central cardiorespiratory regulation is still poorly understood. Experiments were conducted on urethane-anesthetized, vagotomized, and artificially ventilated male Sprague-Dawley rats (n = 22) to ascertain whether MCH modulates sympathetic vasomotor tone, as well as barosympathetic, chemosympathetic, and somatosympathetic reflexes at the level of the spinal cord. Intrathecal injection of 10 µl of MCH produced a dose-dependent hypotension, bradycardia, and sympathoinhibition. Peak response was observed following administration of 1 mM MCH, causing a decrease in mean arterial pressure of 39 ± 2 mmHg (P < 0.001), splanchnic sympathetic nerve activity of 78 ± 11% (P < 0.001), and heart rate of 87 ± 11 beats per minute (bpm) (P < 0.01). The two peaks of the somatosympathetic reflex were decreased by intrathecal MCH, 7 ± 3% (P < 0.01) and 31 ± 6% (P < 0.01), respectively, and the spinal component of the reflex was accentuated 96 ± 23% (P < 0.05), with respect to the baseline for MCH, compared with the two peaks and spinal component of the somatosympathetic reflex elicited following saline injection with respect to the baseline for saline. MCH decreased the sympathetic gain to 120 s of hyperoxic hypercapnea (10% CO(2) in 90% O(2)) and to 10-12 s poikilocapneic anoxia (100% N(2)) from 0.74 ± 0.14%/s to 0.23 ± 0.04%/s (P < 0.05) and 16.47 ± 3.2% to 4.35 ± 1.56% (P < 0.05), respectively. There was a 34% decrease in gain and a 62% decrease in range of the sympathetic baroreflex with intrathecal MCH. These data demonstrate that spinal MCH blunts the central regulation of sympathetic tone and adaptive sympathetic reflexes.


Asunto(s)
Fenómenos Fisiológicos Cardiovasculares/efectos de los fármacos , Hormonas Hipotalámicas/farmacología , Melaninas/farmacología , Hormonas Hipofisarias/farmacología , Reflejo/efectos de los fármacos , Simpaticolíticos/farmacología , Animales , Hormonas Hipotalámicas/administración & dosificación , Inyecciones Espinales , Masculino , Melaninas/administración & dosificación , Hormonas Hipofisarias/administración & dosificación , Ratas , Ratas Sprague-Dawley , Reflejo/fisiología , Fenómenos Fisiológicos Respiratorios/efectos de los fármacos , Sistema Nervioso Simpático/efectos de los fármacos , Simpaticolíticos/administración & dosificación
17.
Life Sci ; 90(23-24): 895-9, 2012 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-22579511

RESUMEN

AIMS: To examine the effects of bilateral microinjection of melanin-concentrating hormone (MCH) 50 and 100 ng into the horizontal limb of the diagonal band of Broca (HDB) on sleep variables during the light phase of the light-dark cycle of the rat. MAIN METHODS: Male Wistar rats were implanted for chronic sleep recordings. In addition, a guide cannula was implanted above the right and left HDB. Following the microinjection of MCH or control solution the electroencephalogram and the electromyogram were recorded for 6 h. Data was collected and classified as either wakefulness (W), light sleep, slow wave sleep (SWS) or REM sleep (REMS). Latencies for SWS and REMS, as well as the number of REM periods and the mean duration of REM episodes were also determined. KEY FINDINGS: MCH 50 and 100 ng significantly decreased W during the first 2-h of recording. Moreover, MCH 100 ng significantly reduced REMS latency and increased REMS time during the first 2-h block of the recording, due to an increase in the number of REM periods. SIGNIFICANCE: Our findings tend to suggest that the basal forebrain participates in the effects of MCH on W and REMS through the deactivation of cholinergic, glutamatergic and γ-aminobutyric acid (GABA)-ergic cells.


Asunto(s)
Hormonas Hipotalámicas/farmacología , Melaninas/farmacología , Hormonas Hipofisarias/farmacología , Prosencéfalo/metabolismo , Sueño REM/efectos de los fármacos , Vigilia/efectos de los fármacos , Acetilcolina/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Electroencefalografía , Electromiografía , Ácido Glutámico/metabolismo , Hormonas Hipotalámicas/administración & dosificación , Masculino , Melaninas/administración & dosificación , Microinyecciones , Hormonas Hipofisarias/administración & dosificación , Ratas , Ratas Wistar , Ácido gamma-Aminobutírico/metabolismo
18.
Reprod Fertil Dev ; 24(4): 599-607, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22541548

RESUMEN

The use of hormonally induced spermatozoa expressed in urine (HISu) is a valuable component of reproduction technologies for amphibians. Five protocols for sampling HISu from the European common frog (Rana temporaria) were compared: (1) pituitary extracts, (2) 0.12 µg g⁻¹ luteinising hormone-releasing hormone analogue (LHRHa), (3) 1.20 µg g⁻¹ LHRHa, (4) 11.7 IU g⁻¹ human chorionic gonadotrophin (hCG) and (5) 23.4 IU g⁻¹ hCG (g⁻¹ = per gram bodyweight). From 1 to 24h after administration we assessed the number and concentration of spermatozoa in spermic urine and in holding water, and in urine the percentage of motile spermatozoa and their progressive motility. The protocol using 1.20 µg g⁻¹ LHRHa gave the highest total sperm numbers (650 × 106) and the highest percentage (40%) of samples with sperm concentrations above 200 × 106 mL⁻¹. The percentage motility and progressive motility was similar from all protocols. Considerable amounts of spermatozoa were expressed by R. temporaria into their holding water. We tested hormonal priming and spermiation in the common toad (Bufo bufo) using 0.13 µg g⁻¹ LHRHa administered 24h before a final spermiating dose of 12.8 IU g⁻¹ hCG. No spermatozoa were expressed in holding water. Priming resulted in 35% more spermatozoa than without; however, there were no differences in sperm concentrations. Primed B. bufo produced spermatozoa with significantly higher percentage motility, but not progressive motility, membrane integrity, or abnormal spermatozoa than unprimed males.


Asunto(s)
Bufo bufo/fisiología , Especies en Peligro de Extinción , Hormonas Hipofisarias/farmacología , Rana temporaria/fisiología , Recuento de Espermatozoides/veterinaria , Espermatogénesis/efectos de los fármacos , Animales , Bufo bufo/orina , Gonadotropina Coriónica/administración & dosificación , Gonadotropina Coriónica/farmacología , Relación Dosis-Respuesta a Droga , Hormona Liberadora de Gonadotropina/administración & dosificación , Hormona Liberadora de Gonadotropina/análogos & derivados , Hormona Liberadora de Gonadotropina/farmacología , Indicadores de Salud , Masculino , Moscú , Hipófisis/química , Hormonas Hipofisarias/administración & dosificación , Rana temporaria/orina , Salud Reproductiva , Federación de Rusia , Estaciones del Año , Recuento de Espermatozoides/métodos , Motilidad Espermática/efectos de los fármacos , Espermatozoides/efectos de los fármacos , Extractos de Tejidos/farmacología
19.
Pharmacol Biochem Behav ; 100(3): 581-6, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21925200

RESUMEN

Melanin concentrating hormone (MCH) stimulates feeding driven by energy needs and reward and modifies anxiety behavior. Orexigenic peptides of similar characteristics, including nociceptin/orphanin FQ, Agouti-related protein and opioids, increase consumption also by reducing avoidance of potentially tainted food in animals displaying a conditioned taste aversion (CTA). Herein, using real-time PCR, we assessed whether expression levels of genes encoding MCH and its receptor, MCHR1, were affected in CTA in the rat. We also investigated whether injecting MCH intracerebroventricularly (ICV) during the acquisition and retrieval of LiCl-induced CTA, would alleviate aversive responses. MCHR1 gene was upregulated in the hypothalamus and brain stem of aversive animals, MCH mRNA was significantly higher in the hypothalamus, whereas a strong trend suggesting upregulation of MCH and MCHR1 genes was detected in the amygdala. Despite these expression changes associated with aversion, MCH injected prior to the induction of CTA with LiCl as well as later, during the CTA retrieval upon subsequent presentations of the aversive tastant, did not reduce the magnitude of CTA. We conclude that MCH and its receptor form an orexigenic system whose expression is affected in CTA. This altered MCH expression may contribute to tastant-targeted hypophagia in CTA. However, changing the MCH tone in the brain by exogenous peptide was insufficient to prevent the onset or facilitate extinction of LiCl-induced CTA. This designates MCH as one of many accessory molecules associated with shaping an aversive response, but not a critical one for LiCl-dependent CTA to occur.


Asunto(s)
Encéfalo/metabolismo , Disgeusia/metabolismo , Regulación de la Expresión Génica , Hormonas Hipotalámicas/metabolismo , Melaninas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Hormonas Hipofisarias/metabolismo , Receptores de Somatostatina/metabolismo , Animales , Tronco Encefálico/metabolismo , Condicionamiento Psicológico , Disgeusia/tratamiento farmacológico , Hormonas Hipotalámicas/administración & dosificación , Hormonas Hipotalámicas/genética , Hormonas Hipotalámicas/uso terapéutico , Hipotálamo/metabolismo , Inyecciones Intraventriculares , Masculino , Melaninas/administración & dosificación , Melaninas/genética , Melaninas/uso terapéutico , Proteínas del Tejido Nervioso/administración & dosificación , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/uso terapéutico , Especificidad de Órganos , Hormonas Hipofisarias/administración & dosificación , Hormonas Hipofisarias/genética , Hormonas Hipofisarias/uso terapéutico , ARN Mensajero/metabolismo , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Somatostatina/genética , Regulación hacia Arriba
20.
Zygote ; 20(1): 39-43, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21208496

RESUMEN

In the hatchery-bred tambaqui Colossoma macropomum, spontaneous semen release does not occur, and hand-stripping produces reduced semen volume. The goal of this work is to evaluate the effects of hormonal induction with carp pituitary extract (CPE) on both qualitative (visual aspect, pH, motility, viability and morphological abnormalities) and quantitative (volume, concentration and number of spermatozoa per ejaculate) traits of tambaqui semen. Eleven males were treated with CPE (induced), and 11 were left untreated as a control (non-induced). All analysed parameters except motility and percentage of viable spermatozoa presented significant differences (p < 0.05) between the induced and non-induced treatments. CPE induction resulted in a 25-fold increase in semen volume and a 10-fold increase in the number of spermatozoa collected. However, both sperm concentration and the frequency of sperm with morphological abnormalities (commonly detached heads or bent tails) were significantly lower in CPE-induced fish. The hormonal induction of tambaqui males with CPE is efficient and positively influences some qualitative and quantitative properties of semen. Additionally, semen collection via gentle abdominal massage occurs more readily in CPE-induced fish.


Asunto(s)
Carpas/fisiología , Hormonas Hipofisarias/farmacología , Semen/efectos de los fármacos , Animales , Eyaculación , Concentración de Iones de Hidrógeno , Inyecciones Intramusculares , Masculino , Hormonas Hipofisarias/administración & dosificación , Semen/citología , Semen/fisiología , Recuento de Espermatozoides , Maduración del Esperma , Motilidad Espermática , Espermatozoides/citología , Espermatozoides/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...