Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
1.
Mol Metab ; 72: 101714, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36966947

RESUMEN

OBJECTIVE: Mice with global deletion of the transient receptor potential channel melastatin family member 8 (TRPM8) are obese, and treatment of diet-induced obese (DIO) mice with TRPM8 agonists decrease body weight. Whether TRPM8 signaling regulates energy metabolism via central or peripheral effects is unknow. Here we assessed the metabolic phenotype of mice with either Nestin Cre-mediated neuronal loss of TRPM8, or with deletion of TRPM8 in Advillin Cre positive sensory neurons of the peripheral nervous system (PNS). METHODS: Nestin Cre- and Advillin Cre-Trpm8 knock-out (KO) mice were metabolically phenotyped under chronic exposure to either chow or high-fat diet (HFD), followed by assessment of energy and glucose metabolism. RESULTS: At room temperature, chow-fed neuronal Trpm8 KO are obese and show decreased energy expenditure when acutely treated with the TRPM8 selective agonist icilin. But body weight of neuronal Trpm8 KO mice is indistinguishable from wildtype controls at thermoneutrality, or when mice are chronically exposed to HFD-feeding. In contrast to previous studies, we show that the TRPM8 agonist icilin has no direct effect on brown adipocytes, but that icilin stimulates energy expenditure, at least in part, via neuronal TRPM8 signaling. We further show that lack of TRPM8 in sensory neurons of the PNS does not lead to a metabolically relevant phenotype. CONCLUSIONS: Our data indicate that obesity in TRPM8-deficient mice is centrally mediated and likely originates from alterations in energy expenditure and/or thermal conductance, but does not depend on TRPM8 signaling in brown adipocytes or sensory neurons of the PVN.


Asunto(s)
Intolerancia a la Glucosa , Canales Catiónicos TRPM , Animales , Masculino , Ratones , Peso Corporal , Dieta Alta en Grasa/efectos adversos , Intolerancia a la Glucosa/metabolismo , Ratones Noqueados , Nestina/metabolismo , Obesidad/metabolismo , Canales Catiónicos TRPM/genética , Canales Catiónicos TRPM/metabolismo
2.
Mol Metab ; 30: 72-130, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31767182

RESUMEN

BACKGROUND: The glucagon-like peptide-1 (GLP-1) is a multifaceted hormone with broad pharmacological potential. Among the numerous metabolic effects of GLP-1 are the glucose-dependent stimulation of insulin secretion, decrease of gastric emptying, inhibition of food intake, increase of natriuresis and diuresis, and modulation of rodent ß-cell proliferation. GLP-1 also has cardio- and neuroprotective effects, decreases inflammation and apoptosis, and has implications for learning and memory, reward behavior, and palatability. Biochemically modified for enhanced potency and sustained action, GLP-1 receptor agonists are successfully in clinical use for the treatment of type-2 diabetes, and several GLP-1-based pharmacotherapies are in clinical evaluation for the treatment of obesity. SCOPE OF REVIEW: In this review, we provide a detailed overview on the multifaceted nature of GLP-1 and its pharmacology and discuss its therapeutic implications on various diseases. MAJOR CONCLUSIONS: Since its discovery, GLP-1 has emerged as a pleiotropic hormone with a myriad of metabolic functions that go well beyond its classical identification as an incretin hormone. The numerous beneficial effects of GLP-1 render this hormone an interesting candidate for the development of pharmacotherapies to treat obesity, diabetes, and neurodegenerative disorders.


Asunto(s)
Péptido 1 Similar al Glucagón/metabolismo , Péptido 1 Similar al Glucagón/farmacología , Glucemia/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Polipéptido Inhibidor Gástrico/metabolismo , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Glucosa/metabolismo , Humanos , Hipoglucemiantes/uso terapéutico , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Obesidad/metabolismo , Receptores de Glucagón/metabolismo
3.
Internist (Berl) ; 60(9): 895-902, 2019 Sep.
Artículo en Alemán | MEDLINE | ID: mdl-31346639

RESUMEN

BACKGROUND: The worldwide rise in overweight and obesity is paralleled by an increasing prevalence of type-2 diabetes. Apart from bariatric surgery, treatment options to decrease body weight are often underwhelming. Innovative pharmacological options are required to cope with the global "diabesity" pandemic. OBJECTIVES: Particular novel pharmacological approaches are discussed, with a special focus on polyagonist-based pharmacotherapies. MATERIALS AND METHODS: Articles on co- and tri-agonists for the treatment of obesity and diabetes are presented and discussed. RESULTS: Unimolecular peptides have been developed for the treatment of obesity and type-2 diabetes. These peptides activate the receptors of multiple hormones and bundle their positive effects in one single molecule. In preclinical studies, polyagonists targeting the receptors for glucagon-like peptide-1 (GLP-1), glucagon, or glucose-dependent insulinotropic peptide (GIP) were promising to reduce body weight and blood glucose. GLP-1-mediated delivery of the nuclear hormones estrogen or dexamethasone also yielded beneficial effects in preclinical studies of obesity. CONCLUSIONS: Polyagonists represent an innovative strategy for the development of novel pharmacotherapies to treat obesity and diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Incretinas/uso terapéutico , Obesidad/tratamiento farmacológico , Polifarmacología , Polipéptido Inhibidor Gástrico , Péptido 1 Similar al Glucagón , Humanos , Insulina
4.
J Intern Med ; 284(6): 581-602, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30230640

RESUMEN

Obesity and its comorbidities, such as type 2 diabetes, are pressing worldwide health concerns. Available anti-obesity treatments include weight loss pharmacotherapies and bariatric surgery. Whilst surgical interventions typically result in significant and sustained weight loss, available pharmacotherapies are far less effective, typically decreasing body weight by no more than 5-10%. An emerging class of multi-agonist drugs may eventually bridge this gap. This new class of specially tailored drugs hybridizes the amino acid sequences of key metabolic hormones into one single entity with enhanced potency and sustained action. Successful examples of this strategy include multi-agonist drugs targeting the receptors for glucagon-like peptide-1 (GLP-1), glucagon and the glucose-dependent insulinotropic polypeptide (GIP). Due to the simultaneous activity at several metabolically relevant receptors, these multi-agonists offer improved body weight loss and glucose tolerance relative to their constituent monotherapies. Further advancing this concept, chimeras were generated that covalently link nuclear acting hormones such as oestrogen, thyroid hormone (T3 ) or dexamethasone to peptide hormones such as GLP-1 or glucagon. The benefit of this strategy is to restrict the nuclear hormone action exclusively to cells expressing the peptide hormone receptor, thereby maximizing combinatorial metabolic efficacy of both drug constituents in the target cells whilst preventing the nuclear hormone cargo from entering and acting on cells devoid of the peptide hormone receptor, in which the nuclear hormone might have unwanted effects. Many of these multi-agonists are in preclinical and clinical development and may represent new and effective tools in the fight against obesity and its comorbidities.


Asunto(s)
Hormonas Gastrointestinales/agonistas , Obesidad/tratamiento farmacológico , Péptidos/uso terapéutico , Animales , Hormonas Gastrointestinales/fisiología , Glucagón/agonistas , Glucagón/fisiología , Péptido 1 Similar al Glucagón/agonistas , Péptido 1 Similar al Glucagón/fisiología , Humanos , Obesidad/fisiopatología , Péptidos/farmacología
5.
Pharmacol Rev ; 70(4): 712-746, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30087160

RESUMEN

With their ever-growing prevalence, obesity and diabetes represent major health threats of our society. Based on estimations by the World Health Organization, approximately 300 million people will be obese in 2035. In 2015 alone there were more than 1.6 million fatalities attributable to hyperglycemia and diabetes. In addition, treatment of these diseases places an enormous burden on our health care system. As a result, the development of pharmacotherapies to tackle this life-threatening pandemic is of utmost importance. Since the beginning of the 19th century, a variety of drugs have been evaluated for their ability to decrease body weight and/or to improve deranged glycemic control. The list of evaluated drugs includes, among many others, sheep-derived thyroid extracts, mitochondrial uncouplers, amphetamines, serotonergics, lipase inhibitors, and a variety of hormones produced and secreted by the gastrointestinal tract or adipose tissue. Unfortunately, when used as a single hormone therapy, most of these drugs are underwhelming in their efficacy or safety, and placebo-subtracted weight loss attributed to such therapy is typically not more than 10%. In 2009, the generation of a single molecule with agonism at the receptors for glucagon and the glucagon-like peptide 1 broke new ground in obesity pharmacology. This molecule combined the beneficial anorectic and glycemic effects of glucagon-like peptide 1 with the thermogenic effect of glucagon into a single molecule with enhanced potency and sustained action. Several other unimolecular dual agonists have subsequently been developed, and, based on their preclinical success, these molecules illuminate the path to a new and more fruitful era in obesity pharmacology. In this review, we focus on the historical pharmacological approaches to treat obesity and glucose intolerance and describe how the knowledge obtained by these studies led to the discovery of unimolecular polypharmacology.


Asunto(s)
Obesidad/tratamiento farmacológico , Animales , Femenino , Humanos , Masculino , Pérdida de Peso
6.
J Endocrinol ; 238(2): R109-R119, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29848610

RESUMEN

Obesity is a worldwide pandemic, which can be fatal for the most extremely affected individuals. Lifestyle interventions such as diet and exercise are largely ineffective and current anti-obesity medications offer little in the way of significant or sustained weight loss. Bariatric surgery is effective, but largely restricted to only a small subset of extremely obese patients. While the hormonal factors mediating sustained weight loss and remission of diabetes by bariatric surgery remain elusive, a new class of polypharmacological drugs shows potential to shrink the gap in efficacy between a surgery and pharmacology. In essence, this new class of drugs combines the beneficial effects of several independent hormones into a single entity, thereby combining their metabolic efficacy to improve systems metabolism. Such unimolecular drugs include single molecules with agonism at the receptors for glucagon, glucagon-like peptide 1 and the glucose-dependent insulinotropic polypeptide. In preclinical studies, these specially tailored multiagonists outperform both their mono-agonist components and current best in class anti-obesity medications. While clinical trials and vigorous safety analyses are ongoing, these drugs are poised to have a transformative effect in anti-obesity therapy and might hopefully lead the way to a new era in weight-loss pharmacology.


Asunto(s)
Fármacos Antiobesidad/uso terapéutico , Obesidad/tratamiento farmacológico , Péptidos/uso terapéutico , Animales , Fármacos Antiobesidad/síntesis química , Fármacos Antiobesidad/clasificación , Cirugía Bariátrica , Descubrimiento de Drogas , Metabolismo Energético/efectos de los fármacos , Terapia por Ejercicio , Humanos , Estilo de Vida , Obesidad/metabolismo , Obesidad/terapia , Péptidos/química , Programas de Reducción de Peso/métodos
7.
Int J Obes (Lond) ; 42(3): 507-517, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28901330

RESUMEN

BACKGROUND/OBJECTIVES: Dieting is a popular yet often ineffective way to lower body weight, as the majority of people regain most of their pre-dieting weights in a relatively short time. The underlying molecular mechanisms driving weight regain and the increased risk for metabolic disease are still incompletely understood. Here we investigate the molecular alterations inherited from a history of obesity. METHODS: In our model, male high-fat diet (HFD)-fed obese C57BL/6J mice were switched to a low caloric chow diet, resulting in a decline of body weight to that of lean mice. We measured body composition, as well as metrics of glucose, insulin and lipid homeostasis. This was accompanied by histological and gene expression analysis of adipose tissue and liver to assess adipose tissue inflammation and hepatosteatosis. Moreover, acute hypothalamic response to (re-) exposure to HFD was assessed by qPCR. RESULTS & CONCLUSIONS: Within 7 weeks after diet switch, most obesity-associated phenotypes, such as body mass, glucose intolerance and blood metabolite levels were reversed. However, hepatic inflammation, hepatic steatosis as well as hypertrophy and inflammation of perigonadal, but not subcutaneous, adipocytes persisted in formerly obese mice. Transcriptional profiling of liver and perigonadal fat revealed an upregulation of pathways associated with immune function and cellularity. Thus, we show that weight reduction leaves signs of inflammation in liver and perigonadal fat, indicating that persisting proinflammatory signals in liver and adipose tissue could contribute to an increased risk of formerly obese subjects to develop the metabolic syndrome upon recurring weight gain.


Asunto(s)
Tejido Adiposo/metabolismo , Inflamación/metabolismo , Hígado/metabolismo , Obesidad/metabolismo , Pérdida de Peso/fisiología , Tejido Adiposo/química , Animales , Biomarcadores/análisis , Restricción Calórica , Hígado Graso/metabolismo , Hígado/química , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/dietoterapia
8.
Physiol Rev ; 97(2): 721-766, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28275047

RESUMEN

In the last two decades we have witnessed sizable progress in defining the role of gastrointestinal signals in the control of glucose and energy homeostasis. Specifically, the molecular basis of the huge metabolic benefits in bariatric surgery is emerging while novel incretin-based medicines based on endogenous hormones such as glucagon-like peptide 1 and pancreas-derived amylin are improving diabetes management. These and related developments have fostered the discovery of novel insights into endocrine control of systemic metabolism, and in particular a deeper understanding of the importance of communication across vital organs, and specifically the gut-brain-pancreas-liver network. Paradoxically, the pancreatic peptide glucagon has reemerged in this period among a plethora of newly identified metabolic macromolecules, and new data complement and challenge its historical position as a gut hormone involved in metabolic control. The synthesis of glucagon analogs that are biophysically stable and soluble in aqueous solutions has promoted biological study that has enriched our understanding of glucagon biology and ironically recruited glucagon agonism as a central element to lower body weight in the treatment of metabolic disease. This review summarizes the extensive historical record and the more recent provocative direction that integrates the prominent role of glucagon in glucose elevation with its under-acknowledged effects on lipids, body weight, and vascular health that have implications for the pathophysiology of metabolic diseases, and the emergence of precision medicines to treat metabolic diseases.


Asunto(s)
Encéfalo/metabolismo , Tracto Gastrointestinal/metabolismo , Glucagón/metabolismo , Hígado/metabolismo , Enfermedades Metabólicas/metabolismo , Páncreas/metabolismo , Animales , Encéfalo/efectos de los fármacos , Tracto Gastrointestinal/efectos de los fármacos , Glucagón/farmacología , Homeostasis/fisiología , Humanos , Hígado/efectos de los fármacos , Páncreas/efectos de los fármacos
9.
Mol Psychiatry ; 22(2): 192-201, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27184124

RESUMEN

The maintenance of normal body weight is disrupted in patients with anorexia nervosa (AN) for prolonged periods of time. Prior to the onset of AN, premorbid body mass index (BMI) spans the entire range from underweight to obese. After recovery, patients have reduced rates of overweight and obesity. As such, loci involved in body weight regulation may also be relevant for AN and vice versa. Our primary analysis comprised a cross-trait analysis of the 1000 single-nucleotide polymorphisms (SNPs) with the lowest P-values in a genome-wide association meta-analysis (GWAMA) of AN (GCAN) for evidence of association in the largest published GWAMA for BMI (GIANT). Subsequently we performed sex-stratified analyses for these 1000 SNPs. Functional ex vivo studies on four genes ensued. Lastly, a look-up of GWAMA-derived BMI-related loci was performed in the AN GWAMA. We detected significant associations (P-values <5 × 10-5, Bonferroni-corrected P<0.05) for nine SNP alleles at three independent loci. Interestingly, all AN susceptibility alleles were consistently associated with increased BMI. None of the genes (chr. 10: CTBP2, chr. 19: CCNE1, chr. 2: CARF and NBEAL1; the latter is a region with high linkage disequilibrium) nearest to these SNPs has previously been associated with AN or obesity. Sex-stratified analyses revealed that the strongest BMI signal originated predominantly from females (chr. 10 rs1561589; Poverall: 2.47 × 10-06/Pfemales: 3.45 × 10-07/Pmales: 0.043). Functional ex vivo studies in mice revealed reduced hypothalamic expression of Ctbp2 and Nbeal1 after fasting. Hypothalamic expression of Ctbp2 was increased in diet-induced obese (DIO) mice as compared with age-matched lean controls. We observed no evidence for associations for the look-up of BMI-related loci in the AN GWAMA. A cross-trait analysis of AN and BMI loci revealed variants at three chromosomal loci with potential joint impact. The chromosome 10 locus is particularly promising given that the association with obesity was primarily driven by females. In addition, the detected altered hypothalamic expression patterns of Ctbp2 and Nbeal1 as a result of fasting and DIO implicate these genes in weight regulation.


Asunto(s)
Anorexia Nerviosa/genética , Alelos , Índice de Masa Corporal , Peso Corporal/genética , Bases de Datos Genéticas , Femenino , Frecuencia de los Genes/genética , Sitios Genéticos , Predisposición Genética a la Enfermedad/genética , Variación Genética , Estudio de Asociación del Genoma Completo , Humanos , Desequilibrio de Ligamiento/genética , Masculino , Obesidad/genética , Polimorfismo de Nucleótido Simple/genética , Factores de Riesgo
11.
Drug Res (Stuttg) ; 66(S 01): S10, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27806391
12.
Neuroimage ; 142: 465-473, 2016 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-27402596

RESUMEN

Ghrelin regulates energy homeostasis in various species and enhances memory in rodent models. In humans, the role of ghrelin in cognitive processes has yet to be characterized. Here we show in a double-blind randomized crossover design that acute administration of ghrelin alters encoding-related brain activity, however does not enhance memory formation in humans. Twenty-one healthy young male participants had to memorize food- and non-food-related words presented on a background of a virtual navigational route while undergoing fMRI recordings. After acute ghrelin administration, we observed decreased post-encoding resting state fMRI connectivity between the caudate nucleus and the insula, amygdala, and orbitofrontal cortex. In addition, brain activity related to subsequent memory performance was modulated by ghrelin. On the next day, however, no differences were found in free word recall or cued location-word association recall between conditions; and ghrelin's effects on brain activity or functional connectivity were unrelated to memory performance. Further, ghrelin had no effect on a cognitive test battery comprising tests for working memory, fluid reasoning, creativity, mental speed, and attention. In conclusion, in contrast to studies with animal models, we did not find any evidence for the potential of ghrelin acting as a short-term cognitive enhancer in humans.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/fisiología , Cognición/fisiología , Conectoma/métodos , Ghrelina/farmacología , Memoria a Largo Plazo/fisiología , Recuerdo Mental/fisiología , Adulto , Encéfalo/diagnóstico por imagen , Cognición/efectos de los fármacos , Estudios Cruzados , Método Doble Ciego , Ghrelina/administración & dosificación , Humanos , Imagen por Resonancia Magnética , Masculino , Memoria a Largo Plazo/efectos de los fármacos , Recuerdo Mental/efectos de los fármacos , Adulto Joven
13.
J Neuroendocrinol ; 27(12): 872-86, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26442444

RESUMEN

Ghrelin, a gut hormone originating from the post-translational cleavage of preproghrelin, is the endogenous ligand of growth hormone secretagogue receptor 1a (GHS-R1a). Within the growth hormone (GH) axis, the biological activity of ghrelin requires octanoylation by ghrelin-O-acyltransferase (GOAT), conferring selective binding to the GHS-R1a receptor via acylated ghrelin. Complete loss of preproghrelin-derived signalling (through deletion of the Ghrl gene) contributes to a decline in peak GH release; however, the selective contribution of endogenous acyl-ghrelin to pulsatile GH release remains to be established. We assessed the pulsatile release of GH in ad lib. fed male germline goat(-/-) mice, extending measures to include mRNA for key hypothalamic regulators of GH release, and peripheral factors that are modulated relative to GH release. The amount of GH released was reduced in young goat(-/-) mice compared to age-matched wild-type mice, whereas pulse frequency and irregularity increased. Altered GH release did not coincide with alterations in hypothalamic Ghrh, Srif, Npy or Ghsr mRNA expression, or pituitary GH content, suggesting that loss of Goat does not compromise canonical mechanisms that contribute to pituitary GH production and release. Although loss of Goat resulted in an irregular pattern of GH release (characterised by an increase in the number of GH pulses observed during extended secretory events), this did not contribute to a change in the expression of sexually dimorphic GH-dependent liver genes. Of interest, circulating levels of insulin-like growth factor (IGF)-1 were elevated in goat(-/-) mice. This rise in circulating levels of IGF-1 was correlated with an increase in GH pulse frequency, suggesting that sustained or increased IGF-1 release in goat(-/-) mice may occur in response to altered GH release patterning. Our observations demonstrate that germline loss of Goat alters GH release and patterning. Although the biological relevance of altered GH secretory patterning remains unclear, we propose that this may contribute to sustained IGF-1 release and growth in goat(-/-) mice.


Asunto(s)
Aciltransferasas/deficiencia , Aciltransferasas/fisiología , Hormona del Crecimiento/metabolismo , Aciltransferasas/genética , Animales , Hormona Liberadora de Hormona del Crecimiento/biosíntesis , Hipotálamo/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Proteínas de la Membrana , Ratones , Ratones Noqueados , Neuropéptido Y/biosíntesis , Receptores de Ghrelina/biosíntesis , Somatostatina/biosíntesis
14.
Mol Psychiatry ; 19(10): 1095-105, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24732670

RESUMEN

Circulating triglycerides (TGs) normally increase after a meal but are altered in pathophysiological conditions, such as obesity. Although TG metabolism in the brain remains poorly understood, several brain structures express enzymes that process TG-enriched particles, including mesolimbic structures. For this reason, and because consumption of high-fat diet alters dopamine signaling, we tested the hypothesis that TG might directly target mesolimbic reward circuits to control reward-seeking behaviors. We found that the delivery of small amounts of TG to the brain through the carotid artery rapidly reduced both spontaneous and amphetamine-induced locomotion, abolished preference for palatable food and reduced the motivation to engage in food-seeking behavior. Conversely, targeted disruption of the TG-hydrolyzing enzyme lipoprotein lipase specifically in the nucleus accumbens increased palatable food preference and food-seeking behavior. Finally, prolonged TG perfusion resulted in a return to normal palatable food preference despite continued locomotor suppression, suggesting that adaptive mechanisms occur. These findings reveal new mechanisms by which dietary fat may alter mesolimbic circuit function and reward seeking.


Asunto(s)
Encéfalo/metabolismo , Conducta Alimentaria/fisiología , Motivación/fisiología , Recompensa , Triglicéridos/sangre , Anfetamina/farmacología , Animales , Arterias Carótidas/metabolismo , Estimulantes del Sistema Nervioso Central/farmacología , Lipoproteína Lipasa/metabolismo , Masculino , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología
15.
Rev Endocr Metab Disord ; 14(4): 331-8, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24009071

RESUMEN

Obesity, and its associated comorbidities such as type 2 diabetes, cardiovascular diseases, and certain cancers, represent major health challenges. Importantly, there is a sexual dimorphism with respect to the prevalence of obesity and its associated metabolic diseases, implicating a role for gonadal hormones. Specifically, estrogens have been demonstrated to regulate metabolism perhaps by acting as a leptin mimetic in the central nervous system (CNS). CNS estrogen receptors (ERs) include ER alpha (ERα) and ER beta (ERß), which are found in nuclear, cytoplasmic and membrane sites throughout the brain. Additionally, estrogens can bind to and activate a G protein-coupled estrogen receptor (GPER), which is a membrane-associated ER. ERs are expressed on neurons as well as glia, which are known to play a major role in providing nutrient supply for neurons and have recently received increasing attention for their potentially important involvement in the CNS regulation of systemic metabolism and energy balance. This brief overview summarizes data focusing on the potential role of astrocytic estrogen action as a key component of estrogenic modulation responsible for mediating the sexual dimorphism in body weight regulation and obesity.


Asunto(s)
Astrocitos/fisiología , Estrógenos/fisiología , Metabolismo , Sistemas Neurosecretores/fisiología , Animales , Humanos , Hipotálamo/citología , Hipotálamo/metabolismo , Obesidad/etiología , Caracteres Sexuales
16.
J Endocrinol Invest ; 36(3): 180-4, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22526011

RESUMEN

BACKGROUND: Ghrelin is a gastrointestinal peptide that promotes a positive energy balance. The enzyme ghrelin O-acyltransferase (GOAT) esterifies an n-octanoic acid to the peptide, thereby enabling ghrelin to bind and activate the ghrelin receptor. Although ghrelin has previously been implicated in the control and maintenance of body core temperature (BCT), the role that this acylation may play in thermoregulation has not been examined. AIM: We aimed to investigate the endogenous role of ghrelin acylation in thermoregulation. METHODS: In this study, we exposed mice lacking the enzyme GOAT as well as wild-type (WT) control mice to cold temperatures under ad libitum and fasting conditions. Additionally, we investigated the role of GOAT in metabolic adaptation to cold temperatures by analyzing BCT and energy metabolism in mice with and without GOAT that were progressively exposed to low ambient temperatures (31-7 C). RESULTS: We find that regardless of nutritional status, mice lacking GOAT maintain a similar BCT as their WT counterparts during an 8 h cold exposure. Furthermore, mice lacking GOAT maintain a similar BCT and metabolic adaptation asWT controls during acclimatization to low ambient temperatures. CONCLUSIONS: We conclude that the absence of the enzyme GOAT does not play a significant role in maintenance of BCT or metabolic adaptation during exposure to low external temperatures.


Asunto(s)
Aciltransferasas/fisiología , Regulación de la Temperatura Corporal/genética , Aclimatación/genética , Aciltransferasas/genética , Animales , Frío , Ingestión de Alimentos/fisiología , Metabolismo Energético/genética , Ayuno/sangre , Ayuno/metabolismo , Ayuno/fisiología , Proteínas de la Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Tiempo
17.
Diabetologia ; 54(12): 3121-31, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21987346

RESUMEN

AIMS/HYPOTHESIS: We examined the physiological mechanisms by which cannabinoid receptor 1 (CB1) antagonism improves glucose metabolism and insulin sensitivity independent of its anorectic and weight-reducing effects, as well as the effects of CB1 antagonism on brown adipose tissue (BAT) function. METHODS: Three groups of diet-induced obese mice received for 1 month: vehicle; the selective CB1 antagonist SR141716; or vehicle/pair-feeding. After measurements of body composition and energy expenditure, mice underwent euglycaemic-hyperinsulinaemic clamp studies to assess in vivo insulin action. In separate cohorts, we assessed insulin action in weight-reduced mice with diet-induced obesity (DIO), and the effect of CB1 antagonism on BAT thermogenesis. Surgical denervation of interscapular BAT (iBAT) was carried out in order to study the requirement for the sympathetic nervous system in mediating the effects of CB1 antagonism on BAT function. RESULTS: Weight loss associated with chronic CB1 antagonism was accompanied by increased energy expenditure, enhanced insulin-stimulated glucose utilisation, and marked activation of BAT thermogenesis. Insulin-dependent glucose uptake was significantly increased in white adipose tissue and BAT, whereas glycogen synthesis was increased in liver, fat and muscle. Despite marked weight loss in the mice, SR141716 treatment did not improve insulin-mediated suppression of hepatic glucose production nor increase skeletal muscle glucose uptake. Denervation of iBAT blunted the effect of SR141716 on iBAT differentiation and insulin-mediated glucose uptake. CONCLUSIONS/INTERPRETATION: Chronic CB1 antagonism markedly enhances insulin-mediated glucose utilisation in DIO mice, independent of its anorectic and weight-reducing effects. The potent effect on insulin-stimulated BAT glucose uptake reveals a novel role for CB1 receptors as regulators of glucose metabolism.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Glucosa/metabolismo , Piperidinas/administración & dosificación , Pirazoles/administración & dosificación , Receptor Cannabinoide CB1/antagonistas & inhibidores , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/inervación , Tejido Adiposo Pardo/cirugía , Animales , Composición Corporal/efectos de los fármacos , Dieta Alta en Grasa , Metabolismo Energético/efectos de los fármacos , Gluconeogénesis/efectos de los fármacos , Glucógeno/biosíntesis , Insulina/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Rimonabant , Termogénesis/efectos de los fármacos , Pérdida de Peso/efectos de los fármacos
18.
Peptides ; 32(11): 2301-8, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21893140

RESUMEN

Since its discovery, many physiologic functions have been ascribed to ghrelin, a gut derived hormone. The presence of a median fatty acid side chain on the ghrelin peptide is required for the binding and activation of the classical ghrelin receptor, the growth hormone secretagogue receptor (GHSR)-1a. Ghrelin O-acyl transferase (GOAT) was recently discovered as the enzyme responsible for this acylation process. GOAT is expressed in all tissues that have been found to express ghrelin and has demonstrated actions on several complex endocrine organ systems such as the hypothalamus-pituitary-gonadal, insular and adrenal axis as well as the gastrointestinal (GI) tract, bone and gustatory system. Ghrelin acylation is dependent on the function of GOAT and the availability of substrates such as proghrelin and short- to medium-chain fatty acids (MCFAs). This process is governed by GOAT activity and has been shown to be modified by dietary lipids. In this review, we provided evidence that support an important role of GOAT in the regulation of energy homeostasis and glucose metabolism by modulating acyl ghrelin (AG) production. The relevance of GOAT and AG during periods of starvation remains to be defined. In addition, we summarized the recent literature on the metabolic effects of GOAT specific inhibitors and shared our view on the potential of targeting GOAT for the treatment of metabolic disorders such as obesity and type 2 diabetes.


Asunto(s)
Aciltransferasas , Ghrelina/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Péptidos/farmacología , Receptores de Ghrelina/metabolismo , Transducción de Señal , Acilación , Aciltransferasas/antagonistas & inhibidores , Aciltransferasas/metabolismo , Animales , Regulación del Apetito/efectos de los fármacos , Regulación del Apetito/fisiología , Glucemia/metabolismo , Peso Corporal , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/fisiología , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Ácidos Grasos/metabolismo , Mucosa Gástrica/metabolismo , Expresión Génica , Ghrelina/genética , Humanos , Factor I del Crecimiento Similar a la Insulina/genética , Ratones , Ratones Noqueados , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Péptidos/uso terapéutico , Receptores de Ghrelina/genética
19.
Physiol Behav ; 105(1): 52-61, 2011 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-21554896

RESUMEN

Recent studies suggest that spontaneous physical activity (SPA) may be under the non-conscious control of neuroendocrine circuits that are known to control food intake. To further elucidate endocrine gut-brain communication as a component of such circuitry, we here analyzed long-term and acute effects of the gastrointestinal hormones ghrelin and PYY 3-36 as well as their hypothalamic neuropeptide targets NPY, AgRP and POMC (alpha-MSH), on locomotor activity and home cage behaviors in rats. For the analysis of SPA, we used an automated infrared beam break activity measuring system, combined with a novel automated video-based behavior analysis system (HomeCageScan (HCS)). Chronic (one-month) peripheral infusion of ghrelin potently increased body weight and fat mass in rats. Such positive energy balance was intriguingly not due to an overall increased caloric ingestion, but was predominantly associated with a decrease in SPA. Chronic intracerebroventricular infusion (7 days) of ghrelin corroborated the decrease in SPA and suggested a centrally mediated mechanism. Central administration of AgRP and NPY increased food intake as expected. AgRP administration led to a delayed decrease in SPA, while NPY acutely (but transiently) increased SPA. Behavioral dissection using HCS corroborated the observed acute and transient increases of food intake and SPA by central NPY infusion. Acute central administration of alpha-MSH rapidly decreased food intake but did not change SPA. Central administration of the NPY receptor agonist PYY 3-36 transiently increased SPA. Our data suggest that the control of spontaneous physical activity by gut hormones or their neuropeptide targets may represent an important mechanistic component of energy balance regulation.


Asunto(s)
Ingestión de Alimentos/efectos de los fármacos , Ghrelina/farmacología , Hipotálamo/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Péptido YY/farmacología , Proteína Relacionada con Agouti/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Composición Corporal/efectos de los fármacos , Composición Corporal/fisiología , Ingestión de Alimentos/fisiología , Hipotálamo/fisiología , Masculino , Actividad Motora/fisiología , Neuropéptido Y/metabolismo , Proopiomelanocortina/metabolismo , Ratas , Ratas Long-Evans , Ratas Sprague-Dawley
20.
Diabetologia ; 54(4): 900-9, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21181395

RESUMEN

AIMS/HYPOTHESIS: Obesity and type 2 diabetes are among the most serious health pathologies worldwide. Stress has been proposed as a factor contributing to the development of these health risk factors; however, the underlying mechanisms that link stress to obesity and diabetes need to be further clarified. Here, we study in mice how chronic stress affects dietary consumption and how that relationship contributes to obesity and diabetes. METHODS: C57BL/6J mice were subjected to chronic variable stress (CVS) for 15 days and subsequently fed with a standard chow or high-fat diet. Food intake, body weight, respiratory quotient, energy expenditure and spontaneous physical activity were measured with a customised calorimetric system and body composition was measured with nuclear magnetic resonance. A glucose tolerance test was also applied and blood glucose levels were measured with a glucometer. Plasma levels of adiponectin and resistin were measured using Lincoplex kits. RESULTS: Mice under CVS and fed with a high-fat diet showed impaired glucose tolerance associated with low plasma adiponectin:resistin ratios. CONCLUSIONS/INTERPRETATION: This study demonstrates, in a novel mouse model, how post-traumatic stress disorder enhances vulnerability for impaired glucose metabolism in an energy-rich environment and proposes a potential adipokine-based mechanism.


Asunto(s)
Estrés Fisiológico/fisiología , Adiponectina/sangre , Animales , Composición Corporal/fisiología , Modelos Animales de Enfermedad , Metabolismo Energético/fisiología , Prueba de Tolerancia a la Glucosa , Masculino , Ratones , Ratones Endogámicos C57BL , Resistina/sangre , Estrés Fisiológico/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...