Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Rev Endocr Metab Disord ; 23(5): 919-930, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35687272

RESUMEN

The growth hormone/insulin growth factor-1 axis is a key endocrine system that exerts profound effects on metabolism by its actions on different peripheral tissues but also in the brain. Growth hormone together with insulin growth factor-1 perform metabolic adjustments, including regulation of food intake, energy expenditure, and glycemia. The dysregulation of this hepatic axis leads to different metabolic disorders including obesity, type 2 diabetes or liver disease. In this review, we discuss how the growth hormone/insulin growth factor-1 axis regulates metabolism and its interactions with the central nervous system. Finally, we state our vision for possible therapeutic uses of compounds based in the components of this hepatic axis.


Asunto(s)
Diabetes Mellitus Tipo 2 , Insulina , Sistema Nervioso Central/metabolismo , Hormona del Crecimiento/metabolismo , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo
2.
Int J Mol Sci ; 23(5)2022 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-35269579

RESUMEN

Melanin-concentrating hormone (MCH) is a 19aa cyclic peptide exclusively expressed in the lateral hypothalamic area, which is an area of the brain involved in a large number of physiological functions and vital processes such as nutrient sensing, food intake, sleep-wake arousal, memory formation, and reproduction. However, the role of the lateral hypothalamic area in metabolic regulation stands out as the most relevant function. MCH regulates energy balance and glucose homeostasis by controlling food intake and peripheral lipid metabolism, energy expenditure, locomotor activity and brown adipose tissue thermogenesis. However, the MCH control of energy balance is a complex mechanism that involves the interaction of several neuroendocrine systems. The aim of the present work is to describe the current knowledge of the crosstalk of MCH with different endocrine factors. We also provide our view about the possible use of melanin-concentrating hormone receptor antagonists for the treatment of metabolic complications. In light of the data provided here and based on its actions and function, we believe that the MCH system emerges as an important target for the treatment of obesity and its comorbidities.


Asunto(s)
Hormonas Hipotalámicas/metabolismo , Melaninas/metabolismo , Sistemas Neurosecretores/metabolismo , Obesidad/metabolismo , Hormonas Hipofisarias/metabolismo , Animales , Metabolismo Energético , Humanos , Área Hipotalámica Lateral/metabolismo , Metabolismo de los Lípidos
3.
Int J Mol Sci ; 23(21)2022 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-36362103

RESUMEN

Fibroblast growth factor 21 is a pleiotropic hormone secreted mainly by the liver in response to metabolic and nutritional challenges. Physiologically, fibroblast growth factor 21 plays a key role in mediating the metabolic responses to fasting or starvation and acts as an important regulator of energy homeostasis, glucose and lipid metabolism, and insulin sensitivity, in part by its direct action on the central nervous system. Accordingly, pharmacological recombinant fibroblast growth factor 21 therapies have been shown to counteract obesity and its related metabolic disorders in both rodents and nonhuman primates. In this systematic review, we discuss how fibroblast growth factor 21 regulates metabolism and its interactions with the central nervous system. In addition, we also state our vision for possible therapeutic uses of this hepatic-brain axis.


Asunto(s)
Factores de Crecimiento de Fibroblastos , Resistencia a la Insulina , Animales , Factores de Crecimiento de Fibroblastos/metabolismo , Hígado/metabolismo , Encéfalo/metabolismo , Metabolismo Energético/fisiología
4.
Int J Mol Sci ; 22(3)2021 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-33572672

RESUMEN

Sirtuins are NAD+ dependent deacetylases that regulate a large number of physiological processes. These enzymes are highly conserved and act as energy sensors to coordinate different metabolic responses in a controlled manner. At present, seven mammalian sirtuins (SIRT 1-7) have been identified, with SIRT1 and SIRT6 shown to exert their metabolic actions in the hypothalamus, both with crucial roles in eliciting responses to dampen metabolic complications associated with obesity. Therefore, our aim is to compile the current understanding on the role of SIRT1 and SIRT6 in the hypothalamus, especially highlighting their actions on the control of energy balance.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Metabolismo Energético , Obesidad/metabolismo , Sirtuinas/metabolismo , Animales , Peso Corporal , Ingestión de Alimentos , Humanos , Hipotálamo/metabolismo , Mamíferos
5.
Rev Endocr Metab Disord ; 21(1): 45-56, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31758299

RESUMEN

Non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) are two of the most common liver diseases associated with obesity, type 2 diabetes and metabolic syndrome. The prevalence of these conditions are increasingly rising and presently there is not a pharmacological option available in the market. Elucidation of the mechanism of action and the molecular underpinnings behind liver disease could help to better understand the pathophysiology of these illnesses. In this sense, in the last years modulation of the ghrelin system in preclinical animal models emerge as a promising therapeutic tool. In this review, we compile the latest knowledge of the modulation of ghrelin system and its intracellular pathways that regulates lipid metabolism, hepatic inflammation and liver fibrosis. We also describe novel processes implicated in the regulation of liver disease by ghrelin, such as autophagy or dysregulated circadian rhythms. In conclusion, the information displayed in this review support that the ghrelin system could be an appealing strategy for the treatment of liver disease.


Asunto(s)
Ghrelina/metabolismo , Metabolismo de los Lípidos , Cirrosis Hepática/metabolismo , Hígado/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Animales , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatología , Ghrelina/fisiología , Humanos , Inflamación/metabolismo , Inflamación/fisiopatología , Hígado/fisiopatología , Cirrosis Hepática/fisiopatología , Síndrome Metabólico/metabolismo , Síndrome Metabólico/fisiopatología , Enfermedad del Hígado Graso no Alcohólico/fisiopatología , Obesidad/metabolismo , Obesidad/fisiopatología
6.
Int J Mol Sci ; 20(16)2019 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-31405212

RESUMEN

Glucagon exerts pleiotropic actions on energy balance and has emerged as an attractive target for the treatment of diabetes and obesity in the last few years. Glucagon reduces body weight and adiposity by suppression of appetite and by modulation of lipid metabolism. Moreover, this hormone promotes weight loss by activation of energy expenditure and thermogenesis. In this review, we cover these metabolic actions elicited by glucagon beyond its canonical regulation of glucose metabolism. In addition, we discuss recent developments of therapeutic approaches in the treatment of obesity and diabetes by dual- and tri-agonist molecules based on combinations of glucagon with other peptides. New strategies using these unimolecular polyagonists targeting the glucagon receptor (GCGR), have become successful approaches to evaluate the multifaceted nature of glucagon signaling in energy balance and metabolic syndrome.


Asunto(s)
Regulación del Apetito , Diabetes Mellitus/metabolismo , Metabolismo Energético , Glucagón/metabolismo , Obesidad/metabolismo , Animales , Fármacos Antiobesidad/farmacología , Regulación del Apetito/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Diabetes Mellitus/tratamiento farmacológico , Metabolismo Energético/efectos de los fármacos , Glucagón/agonistas , Glucosa/metabolismo , Humanos , Hipoglucemiantes/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Terapia Molecular Dirigida , Obesidad/tratamiento farmacológico , Péptidos/farmacología , Receptores de Glucagón/metabolismo , Termogénesis/efectos de los fármacos
7.
Mediators Inflamm ; 2015: 620919, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26568663

RESUMEN

Irisin is a cleaved and secreted fragment of fibronectin type III domain containing 5 (FNDC5) that is mainly released by skeletal muscle and was proposed to mediate the beneficial effects of exercise on metabolism. In the present study we aim to investigate the regulation of the circulating levels of irisin in obese animal models (diet-induced obese (DIO) rats and leptin-deficient (ob/ob) mice), as well as the influence of nutritional status and leptin. Irisin levels were measured by Enzyme-Linked Immunosorbent Assay (ELISA) and Radioimmunoassay (RIA). Serum irisin levels remained unaltered in DIO rats and ob/ob mice. Moreover, its circulating levels were also unaffected by fasting, leptin deficiency, and exogenous leptin administration in rodents. In spite of these negative results we find a negative correlation between irisin and insulin in DIO animals and a positive correlation between irisin and glucose under short-term changes in nutritional status. Our findings indicate that serum irisin levels are not modulated by different physiological settings associated to alterations in energy homeostasis. These results suggest that in rodents circulating levels of irisin are not involved in the pathophysiology of obesity and could be unrelated to metabolic status; however, further studies should clarify its precise role in states of glucose homeostasis imbalance.


Asunto(s)
Fibronectinas/sangre , Leptina/sangre , Estado Nutricional , Obesidad/sangre , Animales , Composición Corporal , Masculino , Obesidad/etiología , Ratas , Ratas Sprague-Dawley
8.
Biochem Pharmacol ; 223: 116129, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38490517

RESUMEN

Leptin is a hormone that is secreted by adipocytes in proportion to adipose tissue size, and that informs the brain about the energy status of the body. Leptin acts through its receptor LepRb, expressed mainly in the hypothalamus, and induces a negative energy balance by potent inhibition of feeding and activation of energy expenditure. These actions have led to huge expectations for the development of therapeutic targets for metabolic complications based on leptin-derived compounds. However, the majority of patients with obesity presents elevated leptin production, suggesting that in this setting leptin is ineffective in the regulation of energy balance. This resistance to the action of leptin in obesity has led to the development of "leptin sensitizers," which have been tested in preclinical studies. Much research has focused on generating combined treatments that act on multiple levels of the gastrointestinal-brain axis. The gastrointestinal-brain axis secretes a variety of different anorexigenic signals, such as uroguanylin, glucagon-like peptide-1, amylin, or cholecystokinin, which can alleviate the resistance to leptin action. Moreover, alternative mechanism such as pharmacokinetics, proteostasis, the role of specific kinases, chaperones, ER stress and neonatal feeding modifications are also implicated in leptin resistance. This review will cover the current knowledge regarding the interaction of leptin with different endocrine factors from the gastrointestinal-brain axis and other novel mechanisms that improve leptin sensitivity in obesity.


Asunto(s)
Leptina , Obesidad , Humanos , Recién Nacido , Tejido Adiposo/metabolismo , Metabolismo Energético/fisiología , Hipotálamo/metabolismo , Leptina/metabolismo , Obesidad/tratamiento farmacológico , Obesidad/metabolismo
9.
Pharmacol Res ; 68(1): 125-31, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23174266

RESUMEN

In recent years, numerous studies have demonstrated the health benefits of polyphenols, and special attention has been paid to their beneficial effects against cardiovascular disease, the leading cause of death in the world today. Polyphenols present vasodilator effects and are able to improve lipid profiles and attenuate the oxidation of low density lipoproteins. In addition, they present clear anti-inflammatory effects and can modulate apoptotic processes in the vascular endothelium. It has been suggested that most of these effects are a consequence of the antioxidant properties of polyphenols, but this idea is not completely accepted, and many other mechanisms have been proposed recently to explain the health effects of these compounds. In fact, different signaling pathways have been linked to polyphenols. This review brings together some recent studies which establish the beneficial properties of polyphenols for cardiovascular disease and analyzes the mechanisms involved in these properties.


Asunto(s)
Enfermedades Cardiovasculares/prevención & control , Polifenoles/farmacología , Animales , Antiinflamatorios/farmacología , Apoptosis , Fibrinolíticos/farmacología , Humanos , Vasodilatadores/farmacología
10.
Nat Metab ; 5(6): 1045-1058, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37277610

RESUMEN

Hypothalamic AgRP/NPY neurons are key players in the control of feeding behaviour. Ghrelin, a major orexigenic hormone, activates AgRP/NPY neurons to stimulate food intake and adiposity. However, cell-autonomous ghrelin-dependent signalling mechanisms in AgRP/NPY neurons remain poorly defined. Here we show that calcium/calmodulin-dependent protein kinase ID (CaMK1D), a genetic hot spot in type 2 diabetes, is activated upon ghrelin stimulation and acts in AgRP/NPY neurons to mediate ghrelin-dependent food intake. Global Camk1d-knockout male mice are resistant to ghrelin, gain less body weight and are protected against high-fat-diet-induced obesity. Deletion of Camk1d in AgRP/NPY, but not in POMC, neurons is sufficient to recapitulate above phenotypes. In response to ghrelin, lack of CaMK1D attenuates phosphorylation of CREB and CREB-dependent expression of the orexigenic neuropeptides AgRP/NPY in fibre projections to the paraventricular nucleus (PVN). Hence, CaMK1D links ghrelin action to transcriptional control of orexigenic neuropeptide availability in AgRP neurons.


Asunto(s)
Diabetes Mellitus Tipo 2 , Ghrelina , Ratones , Animales , Masculino , Ghrelina/metabolismo , Proteína Relacionada con Agouti/genética , Proteína Relacionada con Agouti/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Neuropéptido Y/genética , Neuropéptido Y/metabolismo , Neuronas/metabolismo , Obesidad/metabolismo , Ratones Noqueados , Ingestión de Alimentos , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/metabolismo
11.
Sci Adv ; 9(20): eadf2982, 2023 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-37196091

RESUMEN

The synchronization of circadian clock depends on a central pacemaker located in the suprachiasmatic nuclei. However, the potential feedback of peripheral signals on the central clock remains poorly characterized. To explore whether peripheral organ circadian clocks may affect the central pacemaker, we used a chimeric model in which mouse hepatocytes were replaced by human hepatocytes. Liver humanization led to reprogrammed diurnal gene expression and advanced the phase of the liver circadian clock that extended to muscle and the entire rhythmic physiology. Similar to clock-deficient mice, liver-humanized mice shifted their rhythmic physiology more rapidly to the light phase under day feeding. Our results indicate that hepatocyte clocks can affect the central pacemaker and offer potential perspectives to apprehend pathologies associated with altered circadian physiology.


Asunto(s)
Relojes Circadianos , Ritmo Circadiano , Humanos , Ratones , Animales , Ritmo Circadiano/genética , Hígado/metabolismo , Hepatocitos , Relojes Circadianos/genética , Núcleo Supraquiasmático/metabolismo
12.
Pharmacol Res ; 64(1): 31-5, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21349333

RESUMEN

In this study we evaluated the effect of the administration of different soluble fiber enriched-diets on inflammatory and redox state of Zucker fatty rats. Four groups of ten 8 week-old female Zucker fatty rats were used. The four groups were respectively fed the following diets until the 15th week of life: standard diet (obese control), 10% high methoxylated apple pectin (HMAP)-, 5% soluble cocoa fiber (SCF)-, and 10% ß-glucan-enriched diets. A group of Zucker lean rats fed the standard diet was also used as control for normal values of this rat strain. The plasma levels of tumoral necrosis factor-α (TNF-α), adiponectin, and malondialdehyde (MDA) were measured at the end of treatment. The reduced glutathione liver levels were also obtained at that moment. TNF-α plasma levels decreased somewhat in Zucker fatty rats fed the different fibers, and MDA plasma levels significantly decreased in these animals. Nevertheless, adiponectin plasma levels increased in the Zucker fatty rats fed the SCF enriched diet, but did not change in the HMAP and the ß-glucan group. The Zucker fatty rats fed the different fiber showed a trend towards increased the reduced glutathione liver levels, but significant differences with obese control rats were only obtained in the ß-glucan group. The results obtained in this study suggest that the intake of the different soluble fiber-enriched diets that we have evaluated could prevent and/or attenuate the inflammatory and/or the prooxidative state of the metabolic syndrome.


Asunto(s)
Fibras de la Dieta/uso terapéutico , Obesidad/sangre , Obesidad/dietoterapia , Estrés Oxidativo , Adiponectina/sangre , Animales , Biomarcadores/sangre , Cacao/química , Fibras de la Dieta/farmacología , Femenino , Glutatión/metabolismo , Inflamación/sangre , Hígado/efectos de los fármacos , Hígado/metabolismo , Malondialdehído/sangre , Malus/química , Pectinas/farmacología , Ratas , Ratas Zucker , Factor de Necrosis Tumoral alfa/sangre , beta-Glucanos/farmacología
13.
Redox Biol ; 41: 101945, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33744652

RESUMEN

Sirtuin 3 (SIRT3) is one of the seven mammalian sirtuin homologs of the yeast Sir2 gene that has emerged as an important player in the regulation of energy metabolism in peripheral tissues. However, its role in the hypothalamus has not been explored. Herein, we show that the genetic inhibition of SIRT3 in the hypothalamic arcuate nucleus (ARC) induced a negative energy balance and improvement of several metabolic parameters. These effects are specific for POMC neurons, because ablation of SIRT3 in POMC, but not in AgRP neurons, decreased body weight and adiposity, increased energy expenditure and brown adipose tissue (BAT) activity, and induced browning in white adipose tissue (WAT). Notably, the depletion of SIRT3 in POMC neurons caused these effects in male mice fed a chow diet but failed to affect energy balance in males fed a high fat diet and females under both type of diets. Overall, we provide the first evidence pointing for a key role of SIRT3 in POMC neurons in the regulation of energy balance.


Asunto(s)
Proopiomelanocortina , Sirtuina 3 , Tejido Adiposo Pardo/metabolismo , Animales , Dieta Alta en Grasa , Metabolismo Energético , Femenino , Masculino , Ratones , Neuronas/metabolismo , Proopiomelanocortina/metabolismo , Sirtuina 3/metabolismo
14.
Expert Opin Drug Discov ; 14(5): 421-431, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30821530

RESUMEN

INTRODUCTION: Current pharmacological therapies that target single receptors have limited efficacy for the treatment of diabetes and obesity. Novel approaches with hybrid peptides that activate more than one receptor at once to generate beneficial effects through synergistic effects have shown promising results. Several unimolecular dual and tri-agonists, mainly associated with GPCR like GLP-1/GCG/GIP receptors, have shown exceptional efficacy in preclinical models, and are currently being evaluated in clinical trials to investigate their safety and beneficial effects in humans. Areas covered: Herein, the authors review the development of drugs used in the treatment of metabolic disease, from single agonists to the new generation of tri-agonist peptides and compile the latest knowledge available on GPCR-based drug discovery. The authors also provide the reader with their expert perspectives on this exciting area of drug development. Expert opinion: The co-agonists that have been clinically tested so far have been well tolerated and reduce body weight as well as fasting glucose levels in patients with Type 2 Diabetes Mellitus to a higher degree than single agonists alone. The promising data collected so far now warrant large scale randomized clinical trials to assess whether a unimolecular polypharmacy-based approach could translate into safe and efficacious treatments for obesity and its comorbidities.


Asunto(s)
Descubrimiento de Drogas/métodos , Enfermedades Metabólicas/tratamiento farmacológico , Receptores Acoplados a Proteínas G/agonistas , Animales , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/fisiopatología , Desarrollo de Medicamentos/métodos , Receptor del Péptido 1 Similar al Glucagón/agonistas , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Humanos , Enfermedades Metabólicas/fisiopatología , Obesidad/tratamiento farmacológico , Obesidad/fisiopatología , Receptores Acoplados a Proteínas G/metabolismo
15.
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
16.
Cell Stress ; 2(11): 329-331, 2018 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-31225457

RESUMEN

Cancer cells feature strong metabolic changes to cope with the high energy demand for cell growth and division. Given the importance of metabolic reprogramming in tumor development, it seems logical that tumor suppressors and oncogenes are also regulating the molecular pathways controlling these processes. The p53 tumor suppressor gene has been extensively studied for its role in responding to DNA damage, hypoxia, and oncogenic activation. During the last years, we have learnt that p53 has also the capacity to modulate metabolic changes in cells by regulating a large variety of pathways such as glycolysis, oxidative phosphorylation or fatty acid metabolism. Our group has recently found that the lack of p53 in AgRP neurons, but not POMC neurons, causes that mice are more prone to develop diet-induced obesity (Nat Commun. 9(1):3432). The reason for this is that these mice showed a late increase in food intake and especially because they had a reduced thermogenic activity in BAT. The mechanism modulating these actions involves the upregulation of MKK7 that activates c-Jun N-terminal kinase.

18.
Artículo en Inglés | MEDLINE | ID: mdl-29896158

RESUMEN

Energy homeostasis is tightly regulated by the central nervous system which responds to nervous and circulating inputs to adapt food intake and energy expenditure. However, the rewarding and motivational aspect of food is tightly dependent of dopamine (DA) release in mesocorticolimbic (MCL) system and could be operant in uncontrolled caloric intake and obesity. Accumulating evidence indicate that manipulating the microbiota-gut-brain axis through prebiotic supplementation can have beneficial impact of the host appetite and body weight. However, the consequences of manipulating the implication of the microbiota-gut-brain axis in the control motivational and hedonic/reinforcing aspects of food are still underexplored. In this study, we investigate whether and how dietary prebiotic fructo-oligosaccharides (FOS) could oppose, or revert, the change in hedonic and homeostatic control of feeding occurring after a 2-months exposure to high-fat high-sugar (HFHS) diet. The reinforcing and motivational components of food reward were assessed using a two-food choice paradigm and a food operant behavioral test in mice exposed to FOS either during or after HFHS exposure. We also performed mRNA expression analysis for key genes involved in limbic and hypothalamic control of feeding. We show in a preventive-like approach, FOS addition of HFHS diet had beneficial impact of hypothalamic neuropeptides, and decreased the operant performance for food but only after an overnight fast while it did not prevent the imbalance in mesolimbic markers for DA signaling induced by palatable diet exposure nor the spontaneous tropism for palatable food when given the choice. However, when FOS was added to control diet after chronic HFHS exposure, although it did not significantly alter body weight loss, it greatly decreased palatable food tropism and consumption and was associated with normalization of MCL markers for DA signaling. We conclude that the nature of the diet (regular chow or HFHS) as well as the timing at which prebiotic supplementation is introduced (preventive or curative) greatly influence the efficacy of the gut-microbiota-brain axis. This crosstalk selectively alters the hedonic or motivational drive to eat and triggers molecular changes in neural substrates involved in the homeostatic and non-homeostatic control of body weight.

19.
Nat Commun ; 9(1): 3432, 2018 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-30143607

RESUMEN

p53 is a well-known tumor suppressor that has emerged as an important player in energy balance. However, its metabolic role in the hypothalamus remains unknown. Herein, we show that mice lacking p53 in agouti-related peptide (AgRP), but not proopiomelanocortin (POMC) or steroidogenic factor-1 (SF1) neurons, are more prone to develop diet-induced obesity and show reduced brown adipose tissue (BAT) thermogenic activity. AgRP-specific ablation of p53 resulted in increased hypothalamic c-Jun N-terminal kinase (JNK) activity before the mice developed obesity, and central inhibition of JNK reversed the obese phenotype of these mice. The overexpression of p53 in the ARC or specifically in AgRP neurons of obese mice decreased body weight and stimulated BAT thermogenesis, resulting in body weight loss. Finally, p53 in AgRP neurons regulates the ghrelin-induced food intake and body weight. Overall, our findings provide evidence that p53 in AgRP neurons is required for normal adaptations against diet-induced obesity.


Asunto(s)
Dieta/efectos adversos , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Obesidad/etiología , Obesidad/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Tejido Adiposo Pardo/metabolismo , Proteína Relacionada con Agouti/metabolismo , Animales , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa 8 Activada por Mitógenos/genética , Neuronas/metabolismo , Proopiomelanocortina/metabolismo , Ratas Sprague-Dawley , Factor Esteroidogénico 1/metabolismo , Proteína p53 Supresora de Tumor/genética
20.
Mol Nutr Food Res ; 61(2)2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27624317

RESUMEN

SCOPE: Studying the flavanol metabolism is essential to identify bioactive compounds, as beneficial effects of flavanols have been attributed to their metabolic products. However, host-related factors, including pathological conditions, may affect flavanol metabolism and, thus, their bioactivity. This study aims to elucidate whether hypertension affects grape seed flavanol metabolism, influencing their bioactivity in relation to hypertension. METHODS AND RESULTS: Grape seed flavanols' effect on blood pressure (BP) was studied in spontaneously hypertensive rats (SHR) and healthy Wistar rats 6 h after grape seed extract administration (375 mg/kg). Animals were then sacrificed, and plasma bioavailability and aorta distribution of flavanol metabolites were studied by HPLC-MS/MS in both the groups. Grape seed flavanols were only able to decrease BP in SHR. Plasma total flavanol metabolites showed similar levels, being the difference noticed in specific metabolites' concentrations. Specifically, microbial metabolites showed quantitative and qualitative differences between both health states. Moreover, aorta total concentrations were found decreased in SHR. Interestingly, flavanol microbial metabolites were specifically increased SHR aortas, showing qualitative differences in small phenolic forms. CONCLUSION: This study demonstrates important differences in bioactivity and target tissue metabolite levels between healthy and diseased rats, indicating potential metabolites responsible of the anti-hypertensive effect.


Asunto(s)
Flavonoides/farmacocinética , Extracto de Semillas de Uva/farmacocinética , Vitis/química , Animales , Antihipertensivos/farmacología , Aorta/efectos de los fármacos , Disponibilidad Biológica , Presión Sanguínea/efectos de los fármacos , Flavonoides/sangre , Flavonoides/metabolismo , Extracto de Semillas de Uva/farmacología , Estado de Salud , Masculino , Ratas Endogámicas SHR , Ratas Wistar , Semillas/química , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA