Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Neuropeptides ; 105: 102427, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38579490

RESUMEN

Obesity is a critical health condition worldwide that increases the risks of comorbid chronic diseases, but it can be managed with weight loss. However, conventional interventions relying on diet and exercise are inadequate for achieving and maintaining weight loss, thus there is significant market interest for pharmaceutical anti-obesity agents. For decades, receptor agonists for the gut peptide glucagon-like peptide 1 (GLP-1) featured prominently in anti-obesity medications by suppressing appetite and food reward to elicit rapid weight loss. As the neurocircuitry underlying food motivation overlaps with that for drugs of abuse, GLP-1 receptor agonism has also been shown to decrease substance use and relapse, thus its therapeutic potential may extend beyond weight management to treat addictions. However, as prolonged use of anti-obesity drugs may increase the risk of mood-related disorders like anxiety and depression, and individuals taking GLP-1-based medication commonly report feeling demotivated, the long-term safety of such drugs is an ongoing concern. Interestingly, current research now focuses on dual agonist approaches that include GLP-1 receptor agonism to enable synergistic effects on weight loss or associated functions. GLP-1 is secreted from the same intestinal cells as the anorectic gut peptide, Peptide YY3-36 (PYY3-36), thus this review assessed the therapeutic potential and underlying neural circuits targeted by PYY3-36 when administered independently or in combination with GLP-1 to curb the appetite for food or drugs of abuse like opiates, alcohol, and nicotine. Additionally, we also reviewed animal and human studies to assess the impact, if any, for GLP-1 and/or PYY3-36 on mood-related behaviors in relation to anxiety and depression. As dual agonists targeting GLP-1 and PYY3-36 may produce synergistic effects, they can be effective at lower doses and offer an alternative approach for therapeutic benefits while mitigating undesirable side effects.


Asunto(s)
Péptido 1 Similar al Glucagón , Péptido YY , Humanos , Animales , Péptido YY/metabolismo , Péptido YY/farmacología , Péptido 1 Similar al Glucagón/metabolismo , Ansiedad/tratamiento farmacológico , Ansiedad/metabolismo , Fragmentos de Péptidos/farmacología , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo
2.
J Comp Neurol ; 532(2): e25588, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38335050

RESUMEN

Melanin-concentrating hormone (MCH) cells in the hypothalamus regulate fundamental physiological functions like energy balance, sleep, and reproduction. This diversity may be ascribed to the neurochemical heterogeneity among MCH cells. One prominent subpopulation of MCH cells coexpresses cocaine- and amphetamine-regulated transcript (CART), and as MCH and CART can have opposing actions, MCH/CART+ and MCH/CART- cells may differentially modulate behavioral outcomes. However, it is not known if there are differences in the cellular properties underlying their functional differences; thus, we compared the neuroanatomical, electrophysiological, and morphological properties of MCH cells in male and female Mch-cre;L10-Egfp reporter mice. Half of MCH cells expressed CART and were most prominent in the medial hypothalamus. Whole-cell patch-clamp recordings revealed differences in their passive and active membrane properties in a sex-dependent manner. Female MCH/CART+ cells had lower input resistances, but male cells largely differed in their firing properties. All MCH cells increased firing when stimulated, but their firing frequency decreases with sustained stimulation. MCH/CART+ cells showed stronger spike rate adaptation than MCH/CART- cells. The kinetics of excitatory events at MCH cells also differed by cell type, as the rising rate of excitatory events was slower at MCH/CART+ cells. By reconstructing the dendritic arborization of our recorded cells, we found no sex differences, but male MCH/CART+ cells had less dendritic length and fewer branch points. Overall, distinctions in topographical division and cellular properties between MCH cells add to their heterogeneity and help elucidate their response to stimuli or effect on modulating their respective neural networks.


Asunto(s)
Cocaína , Hormonas Hipotalámicas , Animales , Femenino , Masculino , Ratones , Anfetaminas/metabolismo , Hormonas Hipotalámicas/metabolismo , Hipotálamo/metabolismo , Melaninas/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Hormonas Hipofisarias/metabolismo
3.
J Neuroendocrinol ; 35(1): e13222, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36529144

RESUMEN

Melanin-concentrating hormone (MCH) neurons within the hypothalamus are heterogeneous and can coexpress additional neuropeptides and transmitters. The majority of MCH neurons express vesicular transporters to package glutamate for synaptic release, and MCH neurons can directly innervate downstream neurons via glutamate release. Although glutamatergic signalling from MCH neurons may support physiological and behavioural roles that are independent of MCH (e.g., in glucose homeostasis and nutrient-sensing), it can also mediate similar roles to MCH in the regulation of energy balance. In addition to energy balance, the MCH system has also been implicated in mood disorders, as MCH receptor antagonists have anxiolytic and anti-depressive effects. However, the contribution of glutamatergic signalling from MCH neurons to mood-related functions have not been investigated. We crossed Mch-cre mice with floxed-Vglut2 mice to delete the expression of the vesicular glutamate transporter 2 (Vglut2) and disable glutamatergic signalling specifically from MCH neurons. The resulting Mch-Vglut2-KO mice showed Vglut2 deletion from over 75% of MCH neurons, and although we did not observe changes in depressive-like behaviours, we found that Mch-Vglut2-KO mice displayed anxiety-like behaviours. Mch-Vglut2-KO mice showed reduced exploratory activity when placed in a new cage and were quicker to consume food placed in the centre of a novel open arena. These findings showed that Vglut2 deletion from MCH neurons resulted in anxiolytic actions and suggested that the anxiogenic effects of glutamate are similar to those of the MCH peptide. Taken together, these findings suggest that glutamate and MCH may synergize to regulate and promote anxiety-like behaviour.


Asunto(s)
Ansiolíticos , Ratones , Animales , Ansiolíticos/metabolismo , Ansiolíticos/farmacología , Neuronas/metabolismo , Ácido Glutámico/metabolismo , Proteína 2 de Transporte Vesicular de Glutamato/genética , Proteína 2 de Transporte Vesicular de Glutamato/metabolismo , Ansiedad
4.
PLoS One ; 17(12): e0279468, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36584182

RESUMEN

The COVID-19 pandemic has been linked with increased reports of depression, anxiety, and stress. Stay-at-home directives during the pandemic-imposed lifestyle changes, including eating and sedentary behaviors that can further undermine mental health outcomes. Physical activity is a vital component for metabolic health, as well as for mental health by serving as an active coping strategy to manage stress and promote resilience. Global reports of increased sedentary leisure behaviors have been associated with feelings of depression and anxiety, but it unclear whether the relationship between physical activity and depression or anxiety persists over time. In this longitudinal study, we investigated (i) whether physical activity at the onset of the pandemic was related to feelings of depression or anxiety over time and (ii) whether this relationship was mediated by stress appraisals during the pandemic. We surveyed 319 adults living in Canada or the United States to assess physical activity, stress appraisals, and mental health outcomes at two time points over a 6-month period. We found a reduction in leisure-time physical activity that was linked to subsequent feelings of depression. Furthermore, individuals with lower levels of physical activity were more likely to appraise their COVID-19 situation to be uncontrollable at pandemic onset and as the pandemic continued. Stress appraisals of threat and uncontrollability were also positively related to feelings of depression. Modelling these three factors together showed that appraising a situation as uncontrollable mediated the relationship between initial physical activity and subsequent depressive feelings. Although correlational, these data highlight the protective role of leisure-time physical activity against worsened mental health outcomes during periods of prolonged stress.


Asunto(s)
COVID-19 , Pandemias , Adulto , Humanos , Estudios Longitudinales , COVID-19/epidemiología , Ejercicio Físico , Ansiedad/epidemiología , Evaluación de Resultado en la Atención de Salud , Depresión/epidemiología
6.
J Comp Neurol ; 530(10): 1634-1657, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35143049

RESUMEN

Beta-klotho (KLB) is a coreceptor required for endocrine fibroblast growth factor (FGF) 15/19 and FGF21 signaling in the brain. Klb is prominent within the hypothalamus, which is consistent with its metabolic functions, but diverse roles for Klb are now emerging. Central Klb expression is low but discrete and may govern FGF-targeted sites. However, given its low expression, it is unclear if Klb mRNA is more widespread. We performed in situ hybridization to label Klb mRNA to generate spatial maps capturing the distribution and levels of Klb within the mouse hypothalamus, hippocampal region, subiculum, and amygdala. Semiquantitative analysis revealed that Klb-labeled cells may express low, medium, or high levels of Klb mRNA. Hypothalamic Klb hybridization was heterogeneous and varied rostrocaudally within the same region. Most Klb-labeled cells were found in the lateral hypothalamic zone, but the periventricular hypothalamic region, including the suprachiasmatic nucleus, contained the greatest proportion of cells expressing medium or high Klb levels. We also found heterogeneous Klb hybridization in the amygdala and subiculum, where Klb was especially distinct within the central amygdalar nucleus and ventral subiculum, respectively. By contrast, Klb-labeled cells in the hippocampal region only expressed low levels of Klb and were typically found in the pyramidal layer of Ammon's horn or dentate gyrus. The Klb-labeled regions identified in this study are consistent with reported roles of Klb in metabolism, taste preference, and neuroprotection. However, additional identified sites, including within the hypothalamus and amygdala, may suggest novel roles for FGF15/19 or FGF21 signaling.


Asunto(s)
Amígdala del Cerebelo , Hipocampo , Animales , Corteza Cerebral , Hipotálamo , Ratones , ARN Mensajero
7.
Sci Rep ; 11(1): 19174, 2021 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-34580387

RESUMEN

Current classes of cancer therapeutics have negative side effects stemming from off-target cytotoxicity. One way to avoid this would be to use a drug delivery system decorated with targeting moieties, such as an aptamer, if a targeted aptamer is available. In this study, aptamers were selected against acute myeloid leukemia (AML) cells expressing the MLL-AF9 oncogene through systematic evolution of ligands by exponential enrichment (SELEX). Twelve rounds of SELEX, including two counter selections against fibroblast cells, were completed. Aptamer pools were sequenced, and three candidate sequences were identified. These sequences consisted of two 23-base primer regions flanking a 30-base central domain. Binding studies were performed using flow cytometry, and the lead sequence had a binding constant of 37.5 + / - 2.5 nM to AML cells, while displaying no binding to fibroblast or umbilical cord blood cells at 200 nM. A truncation study of the lead sequence was done using nine shortened sequences, and showed the 5' primer was not important for binding. The lead sequence was tested against seven AML patient cultures, and five cultures showed binding at 200 nM. In summary, a DNA aptamer specific to AML cells was developed and characterized for future drug-aptamer conjugates.


Asunto(s)
Aptámeros de Nucleótidos/farmacología , Leucemia Mieloide Aguda , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Técnica SELEX de Producción de Aptámeros/métodos , Línea Celular Tumoral , Células Cultivadas , Sangre Fetal , Humanos , Ligandos
8.
Neurosci Biobehav Rev ; 128: 346-357, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34182019

RESUMEN

Fructose consumption has been linked with metabolic syndrome and obesity. Fructose-based sweeteners like high fructose corn syrup taste sweeter, improve food palatability, and are increasingly prevalent in our diet. The increase in fructose consumption precedes the rise in obesity and is a contributing driver to the obesity epidemic worldwide. The role of dietary fructose in obesity can be multifactorial by promoting visceral adiposity, hypertension, and insulin resistance. Interestingly, one emergent finding from human and animal studies is that dietary fructose promotes overfeeding. As the brain is a critical regulator of food intake, we reviewed the evidence that fructose can act in the brain and elucidated the major brain systems underlying fructose-induced overfeeding. We found that fructose acts on multiple interdependent brain systems to increase orexigenic drive and the incentive salience of food while decreasing the latency between food bouts and reducing cognitive control to disinhibit feeding. We concluded that the collective actions of fructose may promote feeding behavior by producing a hunger-like state in the brain.


Asunto(s)
Fructosa , Síndrome Metabólico , Animales , Dieta , Humanos , Obesidad , Edulcorantes
9.
Nutrients ; 12(12)2020 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-33322367

RESUMEN

To limit the spread of coronavirus disease 2019 (COVID-19), many individuals were instructed to stay at home, and teleworking became commonplace. Meanwhile, many others were laid off or worked reduced hours, and some front line workers were required to work longer hours. Concurrently, a surge in reports of "pandemic baking" suggested a cascade effect on eating behaviors, which may be an inadvertent strategy to cope with stress. We conducted an online survey of people living in Canada or the United States (N = 680) to assess how employment change may have been experienced as stressful and linked to a shift in food choices. Regression models suggested that reduced hours and being laid off were associated with greater stress appraisals, avoidant- and emotion-focused coping responses, and negative affect. In turn, negative affect was associated with eating to cope and unhealthy snack choices, like salty or sweet treats. Our study emphasizes that under stressful conditions, such as those experienced during the COVID-19 pandemic, some coping strategies may contribute to the greater vulnerability to downstream effects, particularly those relating to eating choices and nutritional balances.


Asunto(s)
Adaptación Psicológica , COVID-19 , Salud Mental , Pandemias , SARS-CoV-2 , Estrés Psicológico , Adulto , COVID-19/epidemiología , COVID-19/psicología , Femenino , Humanos , Masculino , Estrés Psicológico/epidemiología , Estrés Psicológico/patología
10.
Mol Metab ; 42: 101104, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33075544

RESUMEN

OBJECTIVE: Exposure to persistent organic pollutants is consistently associated with increased diabetes risk in humans. We investigated the short- and long-term impact of transient low-dose dioxin (2,3,7,8-tetrachlorodibenzo-p-dioxin, TCDD) exposure during pregnancy and lactation on glucose homeostasis and beta cell function in female mice, including their response to a metabolic stressor later in life. METHODS: Female mice were injected with either corn oil (CO; vehicle control) or 20 ng/kg/d TCDD 2x/week throughout mating, pregnancy and lactation, and then tracked for 6-10 weeks after chemical exposure stopped. A subset of CO- and TCDD-exposed dams was then transferred to a 45% high-fat diet (HFD) or remained on a standard chow diet for an additional 11 weeks to assess the long-term effects of TCDD on adaptability to a metabolic stressor. To summarize, female mice were transiently exposed to TCDD and then subsequently tracked beyond when TCDD had been excreted to identify lasting metabolic effects of TCDD exposure. RESULTS: TCDD-exposed dams were hypoglycemic at birth but otherwise had normal glucose homeostasis during and post-TCDD exposure. However, TCDD-exposed dams on a chow diet were modestly heavier than controls starting 5 weeks after the last TCDD injection, and their weight gain accelerated after transitioning to a HFD. TCDD-exposed dams also had an accelerated onset of hyperglycemia, impaired glucose-induced plasma insulin levels, reduced islet size, increased MAFA-ve beta cells, and increased proinsulin accumulation following HFD feeding compared to controls. Overall, our study demonstrates that low-dose TCDD exposure during pregnancy has minimal effects on metabolism during the period of active exposure, but has detrimental long-term effects on metabolic adaptability to HFD feeding. CONCLUSIONS: Our study suggests that transient low-dose TCDD exposure in female mice impairs metabolic adaptability to HFD feeding, demonstrating that dioxin exposure may be a contributing factor to obesity and diabetes pathogenesis in females.


Asunto(s)
Dioxinas/efectos adversos , Obesidad/metabolismo , Animales , Diabetes Mellitus , Dieta Alta en Grasa , Dioxinas/metabolismo , Dioxinas/farmacología , Susceptibilidad a Enfermedades/inducido químicamente , Femenino , Glucosa/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/fisiología , Lactancia/efectos de los fármacos , Lactancia/metabolismo , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Embarazo
11.
J Neurosci Res ; 98(10): 2045-2071, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32530066

RESUMEN

Melanin-concentrating hormone (MCH) is a ubiquitous vertebrate neuropeptide predominantly synthesized by neurons of the diencephalon that can act through two G protein-coupled receptors, called MCHR1 and MCHR2. The expression of Mchr1 has been investigated in both rats and mice, but its synthesis remains poorly described. After identifying an antibody that detects MCHR1 with high specificity, we employed immunohistochemistry to map the distribution of MCHR1 in the CNS of rats and mice. Multiple neurochemical markers were also employed to characterize some of the neuronal populations that synthesize MCHR1. Our results show that MCHR1 is abundantly found in a subcellular structure called the primary cilium, which has been associated, among other functions, with the detection of free neurochemical messengers present in the extracellular space. Ciliary MCHR1 was found in a wide range of areas, including the olfactory bulb, cortical mantle, striatum, hippocampal formation, amygdala, midline thalamic nuclei, periventricular hypothalamic nuclei, midbrain areas, and in the spinal cord. No differences were observed between male and female mice, and interspecies differences were found in the caudate-putamen nucleus and the subgranular zone. Ciliary MCHR1 was found in close association with several neurochemical markers, including tyrosine hydroxylase, calretinin, kisspeptin, estrogen receptor, oxytocin, vasopressin, and corticotropin-releasing factor. Given the role of neuronal primary cilia in sensing free neurochemical messengers in the extracellular fluid, the widespread distribution of ciliary MCHR1, and the diverse neurochemical populations who synthesize MCHR1, our data indicate that nonsynaptic communication plays a prominent role in the normal function of the MCH system.


Asunto(s)
Encéfalo/metabolismo , Cilios/metabolismo , Receptores de Somatostatina/biosíntesis , Caracteres Sexuales , Animales , Cilios/genética , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratas , Ratas Long-Evans , Ratas Sprague-Dawley , Receptores de Somatostatina/genética
12.
J Comp Neurol ; 528(11): 1833-1855, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31950494

RESUMEN

The hypothalamus contains catecholaminergic neurons marked by the expression of tyrosine hydroxylase (TH). As multiple chemical messengers coexist in each neuron, we determined if hypothalamic TH-immunoreactive (ir) neurons express vesicular glutamate or GABA transporters. We used Cre/loxP recombination to express enhanced GFP (EGFP) in neurons expressing the vesicular glutamate (vGLUT2) or GABA transporter (vGAT), then determined whether TH-ir neurons colocalized with native EGFPVglut2 - or EGFPVgat -fluorescence, respectively. EGFPVglut2 neurons were not TH-ir. However, discrete TH-ir signals colocalized with EGFPVgat neurons, which we validated by in situ hybridization for Vgat mRNA. To contextualize the observed pattern of colocalization between TH-ir and EGFPVgat , we first performed Nissl-based parcellation and plane-of-section analysis, and then mapped the distribution of TH-ir EGFPVgat neurons onto atlas templates from the Allen Reference Atlas (ARA) for the mouse brain. TH-ir EGFPVgat neurons were distributed throughout the rostrocaudal extent of the hypothalamus. Within the ARA ontology of gray matter regions, TH-ir neurons localized primarily to the periventricular hypothalamic zone, periventricular hypothalamic region, and lateral hypothalamic zone. There was a strong presence of EGFPVgat fluorescence in TH-ir neurons across all brain regions, but the most striking colocalization was found in a circumscribed portion of the zona incerta (ZI)-a region assigned to the hypothalamus in the ARA-where every TH-ir neuron expressed EGFPVgat . Neurochemical characterization of these ZI neurons revealed that they display immunoreactivity for dopamine but not dopamine ß-hydroxylase. Collectively, these findings indicate the existence of a novel mouse hypothalamic population that may signal through the release of GABA and/or dopamine.


Asunto(s)
Hipotálamo/citología , Neuronas/citología , Neuronas/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/metabolismo , Animales , Femenino , Hipotálamo/metabolismo , Masculino , Ratones , Proteínas de Transporte Vesicular de Glutamato/metabolismo
13.
Mol Metab ; 29: 114-123, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31668382

RESUMEN

OBJECTIVE: Melanin-concentrating hormone (MCH) plays a key role in regulating energy balance. MCH acts via its receptor MCHR1, and MCHR1 deletion increases energy expenditure and locomotor activity, which is associated with a hyperdopaminergic state. Since MCHR1 expression is widespread, the neurons supporting the effects of MCH on energy expenditure are not clearly defined. There is a high density of MCHR1 neurons in the striatum, and these neurons are known to be GABAergic. We thus determined if MCH acts via this GABAergic neurocircuit to mediate energy balance. METHODS: We generated a Mchr1-flox mouse and crossed it with the Vgat-cre mouse to assess if MCHR1 deletion from GABAergic neurons expressing the vesicular GABA transporter (vGAT) in female Vgat-Mchr1-KO mice resulted in lower body weights or increased energy expenditure. Additionally, we determined if MCHR1-expressing neurons within the accumbens form part of the neural circuit underlying MCH-mediated energy balance by delivering an adeno-associated virus expressing Cre recombinase to the accumbens nucleus of Mchr1-flox mice. To evaluate if a dysregulated dopaminergic tone leads to their hyperactivity, we determined if the dopamine reuptake blocker GBR12909 prolonged the drug-induced locomotor activity in Vgat-Mchr1-KO mice. Furthermore, we also performed amperometry recordings to test whether MCHR1 deletion increases dopamine output within the accumbens and whether MCH can suppress dopamine release. RESULTS: Vgat-Mchr1-KO mice have lower body weight, increased energy expenditure, and increased locomotor activity. Similarly, restricting MCHR1 deletion to the accumbens nucleus also increased locomotor activity. Vgat-Mchr1-KO mice show increased and prolonged sensitivity to GBR12909-induced locomotor activity, and amperometry recordings revealed that GBR12909 elevated accumbens dopamine levels to twice that of controls, thus MCHR1 deletion may lead to a hyperdopaminergic state that mediates their observed hyperactivity. Consistent with the inhibitory effect of MCH, we found that MCH acutely suppresses dopamine release within the accumbens. CONCLUSIONS: As with established models of systemic MCH or MCHR1 deletion, we found that MCHR1 deletion from GABAergic neurons, specifically those within the accumbens nucleus, also led to increased locomotor activity. A hyperdopaminergic state underlies this increased locomotor activity, and is consistent with our finding that MCH signaling within the accumbens nucleus suppresses dopamine release. In effect, MCHR1 deletion may disinhibit dopamine release leading to the observed hyperactivity.


Asunto(s)
Neuronas GABAérgicas/metabolismo , Locomoción , Receptores de Somatostatina/metabolismo , Animales , Dopamina/metabolismo , Metabolismo Energético , Locomoción/efectos de los fármacos , Ratones , Ratones Transgénicos , Núcleo Accumbens/metabolismo , Piperazinas/farmacología , Receptores de Somatostatina/genética , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/deficiencia , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/genética
14.
Diabetes ; 68(12): 2210-2222, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31530579

RESUMEN

Melanin-concentrating hormone (MCH) is an important regulator of food intake, glucose metabolism, and adiposity. However, the mechanisms mediating these actions remain largely unknown. We used pharmacological and genetic approaches to show that the sirtuin 1 (SIRT1)/FoxO1 signaling pathway in the hypothalamic arcuate nucleus (ARC) mediates MCH-induced feeding, adiposity, and glucose intolerance. MCH reduces proopiomelanocortin (POMC) neuronal activity, and the SIRT1/FoxO1 pathway regulates the inhibitory effect of MCH on POMC expression. Remarkably, the metabolic actions of MCH are compromised in mice lacking SIRT1 specifically in POMC neurons. Of note, the actions of MCH are independent of agouti-related peptide (AgRP) neurons because inhibition of γ-aminobutyric acid receptor in the ARC did not prevent the orexigenic action of MCH, and the hypophagic effect of MCH silencing was maintained after chemogenetic stimulation of AgRP neurons. Central SIRT1 is required for MCH-induced weight gain through its actions on the sympathetic nervous system. The central MCH knockdown causes hypophagia and weight loss in diet-induced obese wild-type mice; however, these effects were abolished in mice overexpressing SIRT1 fed a high-fat diet. These data reveal the neuronal basis for the effects of MCH on food intake, body weight, and glucose metabolism and highlight the relevance of SIRT1/FoxO1 pathway in obesity.


Asunto(s)
Adiposidad/efectos de los fármacos , Proteína Forkhead Box O1/metabolismo , Intolerancia a la Glucosa/metabolismo , Hiperfagia/metabolismo , Hormonas Hipotalámicas/farmacología , Melaninas/farmacología , Neuronas/efectos de los fármacos , Hormonas Hipofisarias/farmacología , Proopiomelanocortina/metabolismo , Sirtuina 1/metabolismo , Adiposidad/fisiología , Animales , Proteína Forkhead Box O1/genética , Intolerancia a la Glucosa/genética , Hiperfagia/genética , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Noqueados , Neuronas/metabolismo , Técnicas de Placa-Clamp , Ratas Sprague-Dawley , Sirtuina 1/genética
15.
Neuropharmacology ; 154: 34-49, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30503993

RESUMEN

The lateral hypothalamus (LH) is a functionally and anatomically complex brain region that is involved in the regulation of many behavioral and physiological processes including feeding, arousal, energy balance, stress, reward and motivated behaviors, pain perception, body temperature regulation, digestive functions and blood pressure. Despite noteworthy experimental efforts over the past decades, the circuit, cellular and synaptic bases by which these different processes are regulated by the LH remains incompletely understood. This knowledge gap links in large part to the high cellular heterogeneity of the LH. Fortunately, the rapid evolution of newer genetic and electrophysiological tools is now permitting the selective manipulation, typically genetically-driven, of discrete LH cell populations. This, in turn, permits not only assignment of function to discrete cell groups, but also reveals that considerable synergistic and antagonistic interactions exist between key LH cell populations that regulate feeding and arousal. For example, we now know that while LH melanin-concentrating hormone (MCH) and orexin/hypocretin neurons both function as sensors of the internal metabolic environment, their roles regulating sleep and arousal are actually opposing. Additional studies have uncovered similarly important roles for subpopulations of LH GABAergic cells in the regulation of both feeding and arousal. Herein we review the role of LH MCH, orexin/hypocretin and GABAergic cell populations in the regulation of energy homeostasis (including feeding) and sleep-wake and discuss how these three cell populations, and their subpopulations, may interact to optimize and coordinate metabolism, sleep and arousal. This article is part of the Special Issue entitled 'Hypothalamic Control of Homeostasis'.


Asunto(s)
Ingestión de Alimentos/fisiología , Metabolismo Energético/fisiología , Área Hipotalámica Lateral/metabolismo , Sueño/fisiología , Vigilia/fisiología , Animales , Humanos , Vías Nerviosas/metabolismo , Orexinas/metabolismo
16.
Brain Struct Funct ; 224(1): 99-110, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30284033

RESUMEN

Neurons containing melanin-concentrating hormone (MCH) in the posterior lateral hypothalamus play an integral role in rapid eye movement sleep (REMs) regulation. As MCH neurons also contain a variety of other neuropeptides [e.g., cocaine- and amphetamine-regulated transcript (CART) and nesfatin-1] and neurotransmitters (e.g., glutamate), the specific neurotransmitter responsible for REMs regulation is not known. We hypothesized that glutamate, the primary fast-acting neurotransmitter in MCH neurons, is necessary for REMs regulation. To test this hypothesis, we deleted vesicular glutamate transporter (Vglut2; necessary for synaptic release of glutamate) specifically from MCH neurons by crossing MCH-Cre mice (expressing Cre recombinase in MCH neurons) with Vglut2flox/flox mice (expressing LoxP-modified alleles of Vglut2), and studied the amounts, architecture and diurnal variation of sleep-wake states during baseline conditions. We then activated the MCH neurons lacking glutamate neurotransmission using chemogenetic methods and tested whether these MCH neurons still promoted REMs. Our results indicate that glutamate in MCH neurons contributes to normal diurnal variability of REMs by regulating the levels of REMs during the dark period, but MCH neurons can promote REMs even in the absence of glutamate.


Asunto(s)
Ritmo Circadiano , Ácido Glutámico/metabolismo , Hormonas Hipotalámicas/metabolismo , Hipotálamo Posterior/metabolismo , Melaninas/metabolismo , Neuronas/metabolismo , Hormonas Hipofisarias/metabolismo , Sueño REM , Animales , Hormonas Hipotalámicas/genética , Hipotálamo Posterior/citología , Masculino , Melaninas/genética , Ratones Transgénicos , Fotoperiodo , Hormonas Hipofisarias/genética , Factores de Tiempo , Proteína 2 de Transporte Vesicular de Glutamato/genética , Proteína 2 de Transporte Vesicular de Glutamato/metabolismo , Vigilia
17.
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
18.
PLoS One ; 11(2): e0148252, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26872145

RESUMEN

Fibroblast growth factor 21 (FGF21) is an important endocrine metabolic regulator expressed in multiple tissues including liver and adipose tissue. Although highest levels of expression are in pancreas, little is known about the function of FGF21 in this tissue. In order to understand the physiology of FGF21 in the pancreas, we analyzed its expression and regulation in both acinar and islet tissues. We found that acinar tissue express 20-fold higher levels than that observed in islets. We also observed that pancreatic FGF21 is nutritionally regulated; a marked reduction in FGF21 expression was noted with fasting while obesity is associated with 3-4 fold higher expression. Acinar and islet cells are targets of FGF21, which when systemically administered, leads to phosphorylation of the downstream target ERK 1/2 in about half of acinar cells and a small subset of islet cells. Chronic, systemic FGF21 infusion down-regulates its own expression in the pancreas. Mice lacking FGF21 develop significant islet hyperplasia and periductal lymphocytic inflammation when fed with a high fat obesogenic diet. Inflammatory infiltrates consist of TCRb+ Thy1+ T lymphocytes with increased levels of Foxp3+ regulatory T cells. Increased levels of inflammatory cells were coupled with elevated expression of cytokines such as TNFα, IFNγ and IL1ß. We conclude that FGF21 acts to limit islet hyperplasia and may also prevent pancreatic inflammation.


Asunto(s)
Dieta Alta en Grasa , Factores de Crecimiento de Fibroblastos/genética , Hiperplasia/genética , Islotes Pancreáticos/metabolismo , Obesidad/genética , Pancreatitis/genética , Células Acinares/metabolismo , Células Acinares/patología , Animales , Grasas de la Dieta/efectos adversos , Ayuno , Factores de Crecimiento de Fibroblastos/metabolismo , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Hiperplasia/etiología , Hiperplasia/metabolismo , Hiperplasia/patología , Inflamación , Interferón gamma/genética , Interferón gamma/metabolismo , Islotes Pancreáticos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Obesidad/etiología , Obesidad/metabolismo , Obesidad/patología , Especificidad de Órganos , Pancreatitis/etiología , Pancreatitis/metabolismo , Pancreatitis/patología , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Transducción de Señal , Antígenos Thy-1/genética , Antígenos Thy-1/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
19.
PLoS One ; 10(10): e0139462, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26444289

RESUMEN

Some animals and humans fed a high-energy diet (HED) are diet-resistant (DR), remaining as lean as individuals who were naïve to HED. Other individuals become obese during HED exposure and subsequently defend the obese weight (Diet-Induced Obesity- Defenders, DIO-D) even when subsequently maintained on a low-energy diet. We hypothesized that the body weight setpoint of the DIO-D phenotype resides in the hypothalamic paraventricular nucleus (PVN), where anorexigenic melanocortins, including melanotan II (MTII), increase presynaptic GABA release, and the orexigenic neuropeptide Y (NPY) inhibits it. After prolonged return to low-energy diet, GABA inputs to PVN neurons from DIO-D rats exhibited highly attenuated responses to MTII compared with those from DR and HED-naïve rats. In DIO-D rats, melanocortin-4 receptor expression was significantly reduced in dorsomedial hypothalamus, a major source of GABA input to PVN. Unlike melanocortin responses, NPY actions in PVN of DIO-D rats were unchanged, but were reduced in neurons of the ventromedial hypothalamic nucleus; in PVN of DR rats, NPY responses were paradoxically increased. MTII-sensitivity was restored in DIO-D rats by several weeks' refeeding with HED. The loss of melanocortin sensitivity restricted to PVN of DIO-D animals, and its restoration upon prolonged refeeding with HED suggest that their melanocortin systems retain the ability to up- and downregulate around their elevated body weight setpoint in response to longer-term changes in dietary energy density. These properties are consistent with a mechanism of body weight setpoint.


Asunto(s)
Peso Corporal/fisiología , Melanocortinas/metabolismo , Obesidad/fisiopatología , Núcleo Hipotalámico Paraventricular/metabolismo , Núcleo Hipotalámico Paraventricular/fisiopatología , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Núcleo Arqueado del Hipotálamo/fisiopatología , Restricción Calórica/métodos , Dieta/métodos , Grasas de la Dieta/metabolismo , Leptina/metabolismo , Neuropéptido Y/metabolismo , Péptidos Cíclicos/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor de Melanocortina Tipo 4/metabolismo , Núcleo Hipotalámico Ventromedial/metabolismo , Núcleo Hipotalámico Ventromedial/fisiopatología , alfa-MSH/análogos & derivados , alfa-MSH/metabolismo , Ácido gamma-Aminobutírico/metabolismo
20.
Eur Neuropsychopharmacol ; 25(10): 1808-16, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26092201

RESUMEN

Olanzapine (OLZ), an atypical antipsychotic, can be effective in treating patients with restricting type anorexia nervosa who exercise excessively. Clinical improvements include weight gain and reduced pathological hyperactivity. However the neuronal populations and mechanisms underlying OLZ actions are not known. We studied the effects of OLZ on hyperactivity using male mice lacking the hypothalamic neuropeptide melanin-concentrating hormone (MCHKO) that are lean and hyperactive. We compared the in vivo effects of systemic or intra-accumbens nucleus (Acb) OLZ administration on locomotor activity in WT and MCHKO littermates. Acute systemic OLZ treatment in WT mice significantly reduced locomotor activity, an effect that is substantially attenuated in MCHKO mice. Furthermore, OLZ infusion directly into the Acb of WT mice reduced locomotor activity, but not in MCHKO mice. To identify contributing neuronal mechanisms, we assessed the effect of OLZ treatment on Acb synaptic transmission ex vivo and in vitro. Intraperitoneal OLZ treatment reduced Acb GABAergic activity in WT but not MCHKO neurons. This effect was also seen in vitro by applying OLZ to acute brain slices. OLZ reduced the frequency and amplitude of GABAergic activity that was more robust in WT than MCHKO Acb. These findings indicate that OLZ reduced Acb GABAergic transmission and that MCH is necessary for the hypolocomotor effects of OLZ.


Asunto(s)
Benzodiazepinas/farmacología , Fármacos del Sistema Nervioso Central/farmacología , Hormonas Hipotalámicas/metabolismo , Melaninas/metabolismo , Actividad Motora/efectos de los fármacos , Núcleo Accumbens/efectos de los fármacos , Hormonas Hipofisarias/metabolismo , Carrera , Animales , Relación Dosis-Respuesta a Droga , Hormonas Hipotalámicas/genética , Masculino , Melaninas/genética , Ratones Endogámicos C57BL , Ratones Transgénicos , Actividad Motora/fisiología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Núcleo Accumbens/fisiología , Olanzapina , Técnicas de Placa-Clamp , Hormonas Hipofisarias/genética , Carrera/fisiología , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología , Técnicas de Cultivo de Tejidos , Ácido gamma-Aminobutírico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...