Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 616
Filtrar
1.
Biol Sex Differ ; 15(1): 33, 2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38570844

RESUMEN

Recent preclinical research exploring how neuropeptide transmitter systems regulate motivated behavior reveal the increasing importance of sex as a critical biological variable. Neuropeptide systems and their central circuits both contribute to sex differences in a range of motivated behaviors and regulate sex-specific behaviors. In this short review, we explore the current research of how sex as a biological variable influences several distinct motivated behaviors that are modulated by the melanin-concentrating hormone (MCH) neuropeptide system. First, we review how MCH regulates feeding behavior within the context of energy homeostasis differently between male and female rodents. Then, we focus on MCH's role in lactation as a sex-specific process within the context of energy homeostasis. Next, we discuss the sex-specific effects of MCH on maternal behavior. Finally, we summarize the role of MCH in drug-motivated behaviors. While these topics are traditionally investigated from different scientific perspectives, in this short review we discuss how these behaviors share commonalities within the larger context of motivated behaviors, and that sex differences discovered in one area of research may impact our understanding in another. Overall, our review highlights the need for further research into how sex differences in energy regulation associated with reproduction and parental care contribute to regulating motivated behaviors.


Asunto(s)
Hormonas Hipotalámicas , Melaninas , Neuropéptidos , Femenino , Masculino , Animales , Caracteres Sexuales , Hormonas Hipotalámicas/farmacología , Hormonas Hipotalámicas/fisiología , Hormonas Hipofisarias/farmacología , Hormonas Hipofisarias/fisiología
2.
Nat Rev Endocrinol ; 19(12): 708-721, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37715028

RESUMEN

Traditional textbook physiology has ascribed unitary functions to hormones from the anterior and posterior pituitary gland, mainly in the regulation of effector hormone secretion from endocrine organs. However, the evolutionary biology of pituitary hormones and their receptors provides evidence for a broad range of functions in vertebrate physiology. Over the past decade, we and others have discovered that thyroid-stimulating hormone, follicle-stimulating hormone, adrenocorticotropic hormone, prolactin, oxytocin and arginine vasopressin act directly on somatic organs, including bone, adipose tissue and liver. New evidence also indicates that pituitary hormone receptors are expressed in brain regions, nuclei and subnuclei. These studies have prompted us to attribute the pathophysiology of certain human diseases, including osteoporosis, obesity and neurodegeneration, at least in part, to changes in pituitary hormone levels. This new information has identified actionable therapeutic targets for drug discovery.


Asunto(s)
Hipófisis , Hormonas Hipofisarias , Humanos , Hormonas Hipofisarias/fisiología , Prolactina , Tejido Adiposo , Encéfalo
3.
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
4.
Biomed Pharmacother ; 155: 113771, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36271553

RESUMEN

Memory storage in the brain is one of the most extensively studied subjects in neuroscience. However, due to the highly complex structure of the memory-related systems in the brain, the mystery remains unsolved. Consolidation is one of the most important parts of the memory process, and one that can be affected by numerous neurodegenerative diseases. Hypothalamic melanin-concentrating hormone (MCH) neuronal activity has been of particular interest to researchers in terms of the association between sleep, neurodegenerative diseases, and memory consolidation. We used Pmch-Cre animals to investigate the role of MCH neuronal activity in memory consolidation. In order to observe the differences in memory consolidation, we chemogenetically inhibited MCH neurons using the DREADD method and measured hippocampus-dependent memory performance with a novel object recognition test applicable to early memory impairment in Alzheimer's disease. Our results revealed no significant improvement or worsening with MCH inhibition, suggesting that the role of MCH should now be evaluated in a wider setting.


Asunto(s)
Hormonas Hipotalámicas , Animales , Ratones , Hormonas Hipotalámicas/fisiología , Hormonas Hipofisarias/fisiología , Sueño REM , Melaninas , Neuronas/fisiología
5.
Endocrinol Metab (Seoul) ; 37(5): 719-731, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36168775

RESUMEN

Over the past years, pituitary hormones and their receptors have been shown to have non-traditional actions that allow them to bypass the hypothalamus-pituitary-effector glands axis. Bone cells-osteoblasts and osteoclasts-express receptors for growth hormone, follicle stimulating hormone (FSH), thyroid stimulating hormone (TSH), adrenocorticotrophic hormone (ACTH), prolactin, oxytocin, and vasopressin. Independent skeletal actions of pituitary hormones on bone have been studied using genetically modified mice with haploinsufficiency and by activating or inactivating the receptors pharmacologically, without altering systemic effector hormone levels. On another front, the discovery of a TSH variant (TSH-ßv) in immune cells in the bone marrow and skeletal action of FSHß through tumor necrosis factor α provides new insights underscoring the integrated physiology of bone-immune-endocrine axis. Here we discuss the interaction of each pituitary hormone with bone and the potential it holds in understanding bone physiology and as a therapeutic target.


Asunto(s)
Hormonas Hipofisarias , Tirotropina , Ratones , Animales , Hormonas Hipofisarias/fisiología , Hormona Folículo Estimulante , Prolactina , Hormona Adrenocorticotrópica
6.
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
7.
Front Neurol Neurosci ; 45: 75-90, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34052816

RESUMEN

During the last decade, optogenetic-based circuit mapping has become one of the most common approaches to systems neuroscience, and amassing studies have expanded our understanding of brain structures causally involved in the regulation of sleep-wake cycles. Recent imaging technologies enable the functional mapping of cellular activity, from population down to single-cell resolution, across a broad repertoire of behaviors and physiological processes, including sleep-wake states. This chapter summarizes experimental evidence implicating hypocretins/orexins, melanin-concentrating hormone, and inhibitory neurons from the lateral hypothalamus (LH) in forming an intricate network involved in regulating sleep and metabolism, including feeding behaviors. It further confirms the dual sleep-metabolic functions of LH cells, and sheds light on a possible mechanism underlying brain plasticity during sleep and metabolic disorders.


Asunto(s)
Conducta Alimentaria/fisiología , Área Hipotalámica Lateral/fisiología , Hormonas Hipotalámicas/fisiología , Melaninas/fisiología , Red Nerviosa/fisiología , Neuronas/fisiología , Orexinas/fisiología , Hormonas Hipofisarias/fisiología , Sueño/fisiología , Animales , Humanos , Área Hipotalámica Lateral/metabolismo , Hormonas Hipotalámicas/metabolismo , Melaninas/metabolismo , Red Nerviosa/metabolismo , Neuronas/metabolismo , Orexinas/metabolismo , Hormonas Hipofisarias/metabolismo
8.
Neuron ; 107(2): 306-319.e9, 2020 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-32407670

RESUMEN

Melanin-concentrating hormone (MCH)-expressing neurons are key regulators of energy and glucose homeostasis. Here, we demonstrate that they provide dense projections to the median eminence (ME) in close proximity to tanycytes and fenestrated vessels. Chemogenetic activation of MCH neurons as well as optogenetic stimulation of their projections in the ME enhance permeability of the ME by increasing fenestrated vascular loops and enhance leptin action in the arcuate nucleus of the hypothalamus (ARC). Unbiased phosphoRiboTrap-based assessment of cell activation upon chemogenetic MCH neuron activation reveals MCH-neuron-dependent regulation of endothelial cells. MCH neurons express the vascular endothelial growth factor A (VEGFA), and blocking VEGF-R signaling attenuates the leptin-sensitizing effect of MCH neuron activation. Our experiments reveal that MCH neurons directly regulate permeability of the ME barrier, linking the activity of energy state and sleep regulatory neurons to the regulation of hormone accessibility to the ARC.


Asunto(s)
Permeabilidad de la Membrana Celular/fisiología , Hormonas Hipotalámicas/fisiología , Eminencia Media/fisiología , Melaninas/fisiología , Neuronas/fisiología , Hormonas Hipofisarias/fisiología , Animales , Núcleo Arqueado del Hipotálamo/fisiología , Vasos Sanguíneos/fisiología , Capilares/fisiología , Núcleo Celular/fisiología , Núcleo Celular/ultraestructura , Células Endoteliales/fisiología , Leptina/fisiología , Eminencia Media/irrigación sanguínea , Ratones , Cultivo Primario de Células , Ratas , Ratas Sprague-Dawley , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/biosíntesis
9.
Science ; 365(6459): 1308-1313, 2019 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-31604241

RESUMEN

The neural mechanisms underlying memory regulation during sleep are not yet fully understood. We found that melanin concentrating hormone-producing neurons (MCH neurons) in the hypothalamus actively contribute to forgetting in rapid eye movement (REM) sleep. Hypothalamic MCH neurons densely innervated the dorsal hippocampus. Activation or inhibition of MCH neurons impaired or improved hippocampus-dependent memory, respectively. Activation of MCH nerve terminals in vitro reduced firing of hippocampal pyramidal neurons by increasing inhibitory inputs. Wake- and REM sleep-active MCH neurons were distinct populations that were randomly distributed in the hypothalamus. REM sleep state-dependent inhibition of MCH neurons impaired hippocampus-dependent memory without affecting sleep architecture or quality. REM sleep-active MCH neurons in the hypothalamus are thus involved in active forgetting in the hippocampus.


Asunto(s)
Hipocampo/citología , Hormonas Hipotalámicas/fisiología , Melaninas/fisiología , Memoria , Hormonas Hipofisarias/fisiología , Células Piramidales/fisiología , Sueño REM , Animales , Conducta Animal , Hipocampo/fisiología , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
10.
Sleep Med ; 49: 28-30, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30172630

RESUMEN

This article focuses on the contributions made by Michel Jouvet concerning the systems responsible for the muscle atonia of paradoxical sleep (REM sleep). He was the first to describe the brainstem system mechanisms responsible for muscle atonia during paradoxical sleep using pontine cats and localized pontine lesions. Also discussed is the research going on in the eighties, when Michel Jouvet was hunting for the hypnogenetic factor. At that time, he thought that it was secreted by the hypophysis; but it finally turned out to be controlled by the hypocretin/orexin and melanin concentrating hormone neurones located in the lateral hypothalamus. Several unforgettable moments with Michel Jouvet are described which occurred between 1983 as well as his last moments with us.


Asunto(s)
Modelos Animales de Enfermedad , Hormonas Hipotalámicas/fisiología , Melaninas/fisiología , Hormonas Hipofisarias/fisiología , Trastorno de la Conducta del Sueño REM/patología , Sueño REM/fisiología , Animales , Gatos , Francia , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Masculino , Vías Nerviosas , Puente/patología , Investigación
11.
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
12.
J Endocrinol ; 237(3): R83-R98, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29555849

RESUMEN

Studies over the past decade have challenged the long-held belief that pituitary hormones have singular functions in regulating specific target tissues, including master hormone secretion. Our discovery of the action of thyroid-stimulating hormone (TSH) on bone provided the first glimpse into the non-traditional functions of pituitary hormones. Here we discuss evolving experimental and clinical evidence that growth hormone (GH), follicle-stimulating hormone (FSH), adrenocorticotrophic hormone (ACTH), prolactin, oxytocin and arginine vasopressin (AVP) regulate bone and other target tissues, such as fat. Notably, genetic and pharmacologic FSH suppression increases bone mass and reduces body fat, laying the framework for targeting the FSH axis for treating obesity and osteoporosis simultaneously with a single agent. Certain 'pituitary' hormones, such as TSH and oxytocin, are also expressed in bone cells, providing local paracrine and autocrine networks for the regulation of bone mass. Overall, the continuing identification of new roles for pituitary hormones in biology provides an entirely new layer of physiologic circuitry, while unmasking new therapeutic targets.


Asunto(s)
Tejido Adiposo/metabolismo , Huesos/metabolismo , Hormonas Hipofisarias/fisiología , Tejido Adiposo/efectos de los fármacos , Hormona Adrenocorticotrópica/farmacología , Hormona Adrenocorticotrópica/fisiología , Animales , Arginina Vasopresina/farmacología , Arginina Vasopresina/fisiología , Huesos/efectos de los fármacos , Hormona Folículo Estimulante/farmacología , Hormona Folículo Estimulante/fisiología , Hormona del Crecimiento/farmacología , Hormona del Crecimiento/fisiología , Humanos , Oxitocina/farmacología , Oxitocina/fisiología , Hormonas Hipofisarias/farmacología , Prolactina/farmacología , Prolactina/fisiología , Tirotropina/farmacología , Tirotropina/fisiología
13.
eNeuro ; 5(1)2018.
Artículo en Inglés | MEDLINE | ID: mdl-29423437

RESUMEN

Electrical signals generated by molecularly-distinct classes of lateral hypothalamus (LH) neurons have distinct physiological consequences. For example, LH orexin neurons promote net body energy expenditure, while LH non-orexin neurons [VGAT, melanin-concentrating hormone (MCH)] drive net energy conservation. Appropriate switching between such physiologically-opposing LH outputs is traditionally thought to require cell-type-specific chemical modulation of LH firing. However, it was recently found that, in vivo, the LH neurons are also physiologically exposed to electrical oscillations of different frequency bands. The role of the different physiological oscillation frequencies in firing of orexin vs non-orexin LH neurons remains unknown. Here, we used brain-slice whole-cell patch-clamp technology to target precisely-defined oscillation waveforms to individual molecularly-defined classes LH cells (orexin, VGAT, MCH, GAD65), while measuring the action potential output of the cells. By modulating the frequency of sinusoidal oscillatory input, we found that high-frequency oscillations (γ, ≈30-200 Hz) preferentially silenced the action potential output orexinLH cells. In contrast, low frequencies (δ-θ, ≈0.5-7 Hz) similarly permitted outputs from different LH cell types. This differential control of orexin and non-orexin cells by oscillation frequency was mediated by cell-specific, impedance-unrelated resonance mechanisms. These results substantiate electrical oscillations as a novel input modality for cell-type-specific control of LH firing, which offers an unforeseen way to control specific cell ensembles within this highly heterogeneous neuronal cluster.


Asunto(s)
Área Hipotalámica Lateral/fisiología , Neuronas/fisiología , Potenciales de Acción , Animales , Estimulación Eléctrica , Femenino , Glutamato Descarboxilasa/fisiología , Hormonas Hipotalámicas/fisiología , Masculino , Melaninas/fisiología , Ratones Transgénicos , Orexinas/fisiología , Hormonas Hipofisarias/fisiología , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/fisiología
14.
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
15.
Physiol Behav ; 181: 75-79, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-28867197

RESUMEN

Melanin-concentrating hormone [MCH] is an important neuromodulator related to motivated behaviors. The MCH-containing neurons are mainly located in the lateral hypothalamic area, zona incerta, and incerto-hypothalamic area. In the medial preoptic area [MPOA], a key region for the regulation of maternal behavior, Pmch mRNA expression and MCH synthesis can be detected exclusively during the lactation period. As litter size affects different parameters of maternal physiology, the aim of this study was to verify whether litter size can modulate the number of MCH-containing neurons in the MPOA of lactating rats. The dams were divided into the following groups: postpartum day 12, 15, or 19, with a large, small or reduced litter. Our results show that the number of MCH-immunoreactive neurons in the MPOA is positively correlated with the number of pups in the litter and that artificially reducing the number of pups can also decrease the number of MCH-immunoreactive neurons in the MPOA.


Asunto(s)
Hormonas Hipotalámicas/fisiología , Lactancia/fisiología , Tamaño de la Camada/fisiología , Melaninas/fisiología , Neuronas/fisiología , Hormonas Hipofisarias/fisiología , Área Preóptica/fisiología , Animales , Recuento de Células/estadística & datos numéricos , Femenino , Periodo Posparto/fisiología , Ratas
16.
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
17.
PLoS One ; 12(1): e0168430, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28061506

RESUMEN

STUDY OBJECTIVES: Sleep reduction after stroke is linked to poor recovery in patients. Conversely, a neuroprotective effect is observed in animals subjected to acute sleep deprivation (SD) before ischemia. This neuroprotection is associated with an increase of the sleep, melanin concentrating hormone (MCH) and orexin/hypocretin (OX) systems. This study aims to 1) assess the relationship between sleep and recovery; 2) test the association between MCH and OX systems with the pathological mechanisms of stroke. METHODS: Sprague-Dawley rats were assigned to four experimental groups: (i) SD_IS: SD performed before ischemia; (ii) IS: ischemia; (iii) SD_Sham: SD performed before sham surgery; (iv) Sham: sham surgery. EEG and EMG were recorded. The time-course of the MCH and OX gene expression was measured at 4, 12, 24 hours and 3, 4, 7 days following ischemic surgery by qRT-PCR. RESULTS: A reduction of infarct volume was observed in the SD_IS group, which correlated with an increase of REM sleep observed during the acute phase of stroke. Conversely, the IS group showed a reduction of REM sleep. Furthermore, ischemia induces an increase of MCH and OX systems during the acute phase of stroke, although, both systems were still increased for a long period of time only in the SD_IS group. CONCLUSIONS: Our data indicates that REM sleep may be involved in the neuroprotective effect of SD pre-ischemia, and that both MCH and OX systems were increased during the acute phase of stroke. Future studies should assess the role of REM sleep as a prognostic marker, and test MCH and OXA agonists as new treatment options in the acute phase of stroke.


Asunto(s)
Hormonas Hipotalámicas/fisiología , Melaninas/fisiología , Orexinas/fisiología , Hormonas Hipofisarias/fisiología , Privación de Sueño , Sueño REM , Accidente Cerebrovascular/fisiopatología , Animales , Electroencefalografía , Electromiografía , Expresión Génica , Hormonas Hipotalámicas/genética , Masculino , Melaninas/genética , Orexinas/genética , Hormonas Hipofisarias/genética , Pronóstico , Ratas Sprague-Dawley , Accidente Cerebrovascular/genética
18.
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
19.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...