Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 125
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 160(1-2): 88-104, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25594176

RESUMEN

The primary task of white adipose tissue (WAT) is the storage of lipids. However, "beige" adipocytes also exist in WAT. Beige adipocytes burn fat and dissipate the energy as heat, but their abundance is diminished in obesity. Stimulating beige adipocyte development, or WAT browning, increases energy expenditure and holds potential for combating metabolic disease and obesity. Here, we report that insulin and leptin act together on hypothalamic neurons to promote WAT browning and weight loss. Deletion of the phosphatases PTP1B and TCPTP enhanced insulin and leptin signaling in proopiomelanocortin neurons and prevented diet-induced obesity by increasing WAT browning and energy expenditure. The coinfusion of insulin plus leptin into the CNS or the activation of proopiomelanocortin neurons also increased WAT browning and decreased adiposity. Our findings identify a homeostatic mechanism for coordinating the status of energy stores, as relayed by insulin and leptin, with the central control of WAT browning.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Insulina/metabolismo , Leptina/metabolismo , Proopiomelanocortina/metabolismo , Adiposidad , Animales , Regulación de la Temperatura Corporal , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Obesidad/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 2/metabolismo
2.
Diabetes Obes Metab ; 25(5): 1213-1220, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36597795

RESUMEN

AIMS: To examine association of liver-expressed antimicrobial peptide 2 (LEAP2), an endogenous ghrelin antagonist with anorexiant effects, to key cardiometabolic risk factors in people with overweight and obesity. METHODS: In this cross-sectional study, we sought to identify associations between LEAP2 levels and cardiometabolic risk factors, including body composition (dual X-ray absorptiometry), insulin and glucose metabolism (oral and intravenous glucose tolerance tests and hyperinsulinaemic-euglycaemic clamps), plasma lipids and inflammation markers (ELISA and multiplex assays). RESULTS: In 65 participants with overweight or obesity (63.1% male, mean age 31.3 ± 8.5 years), LEAP2 levels were associated with total body fat, but not with body mass index or waist-hip ratio in both univariable and age- and sex-adjusted models (P < 0.05). Higher LEAP2 level was also positively associated with higher insulin secretion in univariable (P = 0.047) and multivariable models adjusted for age, sex and body fat (P = 0.03), but not with fasting glucose levels (P ≥ 0.05). Higher LEAP2 levels were associated insulin resistance (P = 0.07) after adjustment for age and sex, but the association disappeared after an additional adjustment for body fat (P = 0.2). There was an inverse association between LEAP2 levels and nuclear factor kappa-B (NFκB) activity in the peripheral blood mononuclear cells in age-, sex- and body fat-adjusted models (P = 0.04). There were no associations with cardiovascular risk factors (lipids, blood pressure) or other inflammation markers. CONCLUSIONS: These results provide important insights into the association between LEAP2 and cardiometabolic health in a high-risk population of individuals with overweight and obesity. This is a first report of an association between LEAP2 and insulin secretion, insulin sensitivity and NFκB activity. LEAP2 may represent an important potential therapeutic target to promote insulin secretion in people with type 2 diabetes and obesity.


Asunto(s)
Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Obesidad , Sobrepeso , Adulto , Femenino , Humanos , Masculino , Adulto Joven , Estudios Transversales , Diabetes Mellitus Tipo 2/complicaciones , Hepcidinas/metabolismo , Inflamación/complicaciones , Insulina/metabolismo , Secreción de Insulina , Leucocitos Mononucleares/metabolismo , Lípidos , Obesidad/complicaciones , Sobrepeso/complicaciones
3.
Int J Obes (Lond) ; 45(11): 2447-2454, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34341471

RESUMEN

BACKGROUND/OBJECTIVES: Obesity has been ascribed to corticostriatal regions taking control over homeostatic areas. To test this assumption, we applied an effective connectivity approach to reveal the direction of information flow between brain regions and the valence of connections (excitatory versus inhibitory) as a function of increased BMI and homeostatic state. SUBJECTS/METHODS: Forty-one participants (21 overweight/obese) underwent two resting-state fMRI scans: after overnight fasting (hunger) and following a standardised meal (satiety). We used spectral dynamic causal modelling to unravel hunger and increased BMI-related changes in directed connectivity between cortical, insular, striatal and hypothalamic regions. RESULTS: During hunger, as compared to satiety, we found increased excitation of the ventromedial prefrontal cortex over the ventral striatum and hypothalamus, suggesting enhanced top-down modulation compensating energy depletion. Increased BMI was associated with increased excitation of the anterior insula over the hypothalamus across the hunger and satiety conditions. The interaction of hunger and increased BMI yielded decreased intra-cortical excitation from the dorso-lateral to the ventromedial prefrontal cortex. CONCLUSIONS: Our findings suggest that excess weight and obesity is associated with persistent top-down excitation of the hypothalamus, regardless of homeostatic state, and hunger-related reductions of dorso-lateral to ventromedial prefrontal inputs. These findings are compatible with eating without hunger and reduced self-regulation views of obesity.


Asunto(s)
Índice de Masa Corporal , Hipotálamo/fisiopatología , Red Nerviosa/anomalías , Corteza Prefrontal/fisiopatología , Adulto , Femenino , Humanos , Hipotálamo/anomalías , Imagen por Resonancia Magnética/métodos , Imagen por Resonancia Magnética/estadística & datos numéricos , Masculino , Red Nerviosa/fisiopatología , Corteza Prefrontal/anomalías
4.
FASEB J ; : fj201800634R, 2018 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-29932868

RESUMEN

Hunger-sensing agouti-related peptide (AgRP) neurons ensure survival by adapting metabolism and behavior to low caloric environments. This adaption is accomplished by consolidating food intake, suppressing energy expenditure, and maximizing fat storage (nutrient partitioning) for energy preservation. The intracellular mechanisms responsible are unknown. Here we report that AgRP carnitine acetyltransferase (Crat) knockout (KO) mice exhibited increased fatty acid utilization and greater fat loss after 9 d of calorie restriction (CR). No differences were seen in mice with ad libitum food intake. Eleven days ad libitum feeding after CR resulted in greater food intake, rebound weight gain, and adiposity in AgRP Crat KO mice compared with wild-type controls, as KO mice act to restore pre-CR fat mass. Collectively, this study highlights the importance of Crat in AgRP neurons to regulate nutrient partitioning and fat mass during chronically reduced caloric intake. The increased food intake, body weight gain, and adiposity in KO mice after CR also highlights the detrimental and persistent metabolic consequence of impaired substrate utilization associated with CR. This finding may have significant implications for postdieting weight management in patients with metabolic diseases.-Reichenbach, A., Stark, R., Mequinion, M., Lockie, S. H., Lemus, M. B., Mynatt, R. L., Luquet, S., Andrews, Z. B. Carnitine acetyltransferase (Crat) in hunger-sensing AgRP neurons permits adaptation to calorie restriction.

5.
J Neurochem ; 147(6): 715-729, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29704424

RESUMEN

Emerging evidence from human imaging studies suggests that obese individuals have altered connectivity between the hypothalamus, the key brain region controlling energy homeostasis, and cortical regions involved in decision-making and reward processing. Historically, animal studies have demonstrated that the lateral hypothalamus is the key hypothalamic region involved in feeding and reward. The lateral hypothalamus is a heterogeneous structure comprised of several distinct types of neurons which are scattered throughout. In addition, the lateral hypothalamus receives inputs from a number of cortical brain regions suggesting that it is uniquely positioned to be a key integrator of cortical information and metabolic feedback. In this review, we summarize how human brain imaging can inform detailed animal studies to investigate neural pathways connecting cortical regions and the hypothalamus. Here, we discuss key cortical brain regions that are reciprocally connected to the lateral hypothalamus and are implicated in decision-making processes surrounding food.


Asunto(s)
Corteza Cerebral/fisiología , Cuerpo Estriado/fisiología , Conducta Alimentaria/fisiología , Hipotálamo/fisiología , Red Nerviosa/fisiología , Obesidad/fisiopatología , Animales , Corteza Cerebral/fisiopatología , Cuerpo Estriado/fisiopatología , Humanos , Hipotálamo/fisiopatología , Red Nerviosa/fisiopatología
6.
Neuroendocrinology ; 107(4): 340-354, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30208370

RESUMEN

BACKGROUND/AIMS: Abuse of toluene products (e.g., glue-sniffing) primarily occurs during adolescence and has been associated with appetite suppression and weight impairments. However, the metabolic phenotype arising from adolescent inhalant abuse has never been fully characterised, and its persistence during abstinence and underlying mechanisms remain unknown. METHODS: Adolescent male Wistar rats (post-natal day 27) were exposed to inhaled toluene (10,000 ppm) (n = 32) or air (n = 48) for 1 h/day, 3 days/week for 4 weeks, followed by 4 weeks of abstinence. Twenty air rats were pair-fed to the toluene group, to differentiate the direct effects of toluene from under-nutrition. Food intake, weight, and growth were monitored. Metabolic hormones were measured after exposure and abstinence periods. Energy expenditure was measured using indirect calorimetry. Adrenal function was assessed using adrenal histology and hormone testing. RESULTS: Inhalant abuse suppressed appetite and increased energy expenditure. Reduced weight gain and growth were observed in both the toluene and pair-fed groups. Compared to the pair-fed group, and despite normalisation of food intake, the suppression of weight and growth for toluene-exposed rats persisted during abstinence. After exposure, toluene-exposed rats had low fasting blood glucose and insulin compared to the air and pair-fed groups. Consistent with adrenal insufficiency, adrenal hypertrophy and increased basal adrenocorticotropic hormone were observed in the toluene-exposed rats, despite normal basal corticosterone levels. CONCLUSIONS: Inhalant abuse results in negative energy balance, persistent growth impairment, and endocrine changes suggestive of adrenal insufficiency. We conclude that adrenal insufficiency contributes to the negative energy balance phenotype, potentially presenting a significant additional health risk for inhalant users.


Asunto(s)
Enfermedades de las Glándulas Suprarrenales/inducido químicamente , Trastornos del Crecimiento/inducido químicamente , Abuso de Inhalantes/complicaciones , Enfermedades Metabólicas/inducido químicamente , Maduración Sexual , Adolescente , Conducta del Adolescente/efectos de los fármacos , Conducta del Adolescente/fisiología , Desarrollo del Adolescente/efectos de los fármacos , Enfermedades de las Glándulas Suprarrenales/metabolismo , Enfermedades de las Glándulas Suprarrenales/fisiopatología , Glándulas Suprarrenales/fisiopatología , Animales , Apetito/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Modelos Animales de Enfermedad , Ingestión de Alimentos/efectos de los fármacos , Trastornos del Crecimiento/metabolismo , Trastornos del Crecimiento/fisiopatología , Humanos , Abuso de Inhalantes/metabolismo , Abuso de Inhalantes/patología , Abuso de Inhalantes/fisiopatología , Masculino , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/fisiopatología , Actividad Motora/efectos de los fármacos , Fenotipo , Ratas , Ratas Wistar , Maduración Sexual/efectos de los fármacos , Maduración Sexual/fisiología , Tolueno/toxicidad
7.
J Neurosci ; 36(10): 3049-63, 2016 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-26961958

RESUMEN

Calorie restriction (CR) is neuroprotective in Parkinson's disease (PD) although the mechanisms are unknown. In this study we hypothesized that elevated ghrelin, a gut hormone with neuroprotective properties, during CR prevents neurodegeneration in an 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model of PD. CR attenuated the MPTP-induced loss of substantia nigra (SN) dopamine neurons and striatal dopamine turnover in ghrelin WT but not KO mice, demonstrating that ghrelin mediates CR's neuroprotective effect. CR elevated phosphorylated AMPK and ACC levels in the striatum of WT but not KO mice suggesting that AMPK is a target for ghrelin-induced neuroprotection. Indeed, exogenous ghrelin significantly increased pAMPK in the SN. Genetic deletion of AMPKß1 and 2 subunits only in dopamine neurons prevented ghrelin-induced AMPK phosphorylation and neuroprotection. Hence, ghrelin signaling through AMPK in SN dopamine neurons mediates CR's neuroprotective effects. We consider targeting AMPK in dopamine neurons may recapitulate neuroprotective effects of CR without requiring dietary intervention.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Restricción Calórica , Ghrelina/metabolismo , Intoxicación por MPTP/patología , Intoxicación por MPTP/prevención & control , Enfermedad de Parkinson/fisiopatología , Transducción de Señal/fisiología , Proteínas Quinasas Activadas por AMP/genética , Animales , Proteínas de Unión al Calcio/metabolismo , Recuento de Células , Cuerpo Estriado/patología , Modelos Animales de Enfermedad , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/genética , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Ghrelina/genética , Ghrelina/farmacología , Proteína Ácida Fibrilar de la Glía/metabolismo , Intoxicación por MPTP/inducido químicamente , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Microfilamentos/metabolismo , Actividad Motora/efectos de los fármacos , Actividad Motora/genética , Neuronas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Tirosina 3-Monooxigenasa/metabolismo
8.
Annu Rev Nutr ; 36: 105-28, 2016 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-27296500

RESUMEN

There is a growing view that certain foods, particularly those high in refined sugars and fats, are addictive and that some forms of obesity can usefully be treated as a food addiction. This perspective is supported by a growing body of neuroscience research demonstrating that the chronic consumption of energy-dense foods causes changes in the brain's reward pathway that are central to the development and maintenance of drug addiction. Obese and overweight individuals also display patterns of eating behavior that resemble the ways in which addicted individuals consume drugs. We critically review the evidence that some forms of obesity or overeating could be considered a food addiction and argue that the use of food addiction as a diagnostic category is premature. We also examine some of the potential positive and negative clinical, social, and public policy implications of describing obesity as a food addiction that require further investigation.


Asunto(s)
Conducta Adictiva/psicología , Medicina Basada en la Evidencia , Política de Salud , Modelos Neurológicos , Obesidad/psicología , Sobrepeso/psicología , Animales , Conducta Adictiva/fisiopatología , Conducta Adictiva/terapia , Investigación Biomédica/tendencias , Dieta Saludable , Ingestión de Energía/efectos de los fármacos , Humanos , Neurobiología/métodos , Neurobiología/tendencias , Trastornos Neurocognitivos/fisiopatología , Trastornos Neurocognitivos/prevención & control , Trastornos Neurocognitivos/psicología , Trastornos Neurocognitivos/terapia , Política Nutricional , Obesidad/fisiopatología , Obesidad/prevención & control , Obesidad/terapia , Sobrepeso/fisiopatología , Sobrepeso/prevención & control , Sobrepeso/terapia , Recompensa
9.
Int J Mol Sci ; 18(3)2017 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-28273852

RESUMEN

Parkinson's disease (PD) is a neurodegenerative disorder, characterized by a loss of dopamine (DA) neurons in the substantia nigra pars compacta (SNc). Caloric restriction (CR) has been shown to exert ghrelin-dependent neuroprotective effects in the 1-methyl-4-phenyl-1,2,3,6-tetrathydropyridine (MPTP)-based animal model for PD. We here investigated whether CR is neuroprotective in the lactacystin (LAC) mouse model for PD, in which proteasome disruption leads to the destruction of the DA neurons of the SNc, and whether this effect is mediated via the ghrelin receptor. Adult male ghrelin receptor wildtype (WT) and knockout (KO) mice were maintained on an ad libitum (AL) diet or on a 30% CR regimen. After 3 weeks, LAC was injected unilaterally into the SNc, and the degree of DA neuron degeneration was evaluated 1 week later. In AL mice, LAC injection significanty reduced the number of DA neurons and striatal DA concentrations. CR protected against DA neuron degeneration following LAC injection. However, no differences were observed between ghrelin receptor WT and KO mice. These results indicate that CR can protect the nigral DA neurons from toxicity related to proteasome disruption; however, the ghrelin receptor is not involved in this effect.


Asunto(s)
Acetilcisteína/análogos & derivados , Restricción Calórica , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Fármacos Neuroprotectores , Receptores de Ghrelina/metabolismo , Acetilcisteína/administración & dosificación , Acetilcisteína/farmacología , Factores de Edad , Animales , Recuento de Células , Masculino , Ratones , Ratones Noqueados , Receptores de Ghrelina/genética , Sustancia Negra/efectos de los fármacos , Sustancia Negra/metabolismo , Sustancia Negra/patología
10.
J Neurochem ; 137(3): 460-71, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26872221

RESUMEN

The gut hormone ghrelin is widely beneficial in many disease states. However, ghrelin exists in two distinctive isoforms, each with its own metabolic profile. In Parkinson's Disease (PD) acylated ghrelin administration is neuroprotective, however, the role of des-acylated ghrelin remains unknown. In this study, we wanted to identify the relative contribution each isoform plays using the MPTP model of PD. Chronic administration of acylated ghrelin in mice lacking both isoforms of ghrelin (Ghrelin KO) attenuated the MPTP-induced loss on tyrosine hydroxylase (TH) neuronal number and volume and TH protein expression in the nigrostriatal pathway. Moreover, acylated ghrelin reduced the increase in glial fibrillary acidic protein and Ionized calcium binding adaptor molecule 1 microglia in the substantia nigra. However, injection of acylated ghrelin also elevated plasma des-acylated ghrelin, indicating in vivo deacetylation. Next, we chronically administered des-acylated ghrelin to Ghrelin KO mice and observed no neuroprotective effects in terms of TH cell number, TH protein expression, glial fibrillary acidic protein and ionized calcium binding adaptor molecule 1 cell number. The lack of a protective effect was mirrored in ghrelin-O-acyltransferase KO mice, which lack the ability to acylate ghrelin and consequently these mice have chronically increased plasma des-acyl ghrelin. Plasma corticosterone was elevated in ghrelin-O-acyltransferase KO mice and with des-acylated ghrelin administration. Overall, our studies suggest that acylated ghrelin is the isoform responsible for in vivo neuroprotection and that pharmacological approaches preventing plasma conversion from acyl ghrelin to des-acyl ghrelin may have clinical efficacy to help slow or prevent the debilitating effects of PD. Ghrelin exists in the plasma as acyl and des-acyl ghrelin. We determined the form responsible for in vivo neuroprotection in a mouse model of Parkinson's disease. Although exogenous acyl ghrelin is deacylated in situ to des-acyl, only acyl ghrelin was neuroprotective by attenuating dopamine cell loss and glial activation. Acyl ghrelin is a therapeutic option to reduce Parkinson's Disease progression. Cover Image for this issue: doi: 10.1111/jnc.13316.


Asunto(s)
Ghrelina/análogos & derivados , Ghrelina/farmacología , Intoxicación por MPTP/prevención & control , Fármacos Neuroprotectores/farmacología , Enfermedad de Parkinson Secundaria/prevención & control , Acilación , Aciltransferasas/metabolismo , Animales , Proteínas de Unión al Calcio/metabolismo , Recuento de Células , Ghrelina/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteínas de la Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/metabolismo , Vías Nerviosas/patología , Neuronas/patología , Fármacos Neuroprotectores/química , Tirosina 3-Monooxigenasa/metabolismo
11.
Clin Sci (Lond) ; 130(17): 1545-58, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27303049

RESUMEN

The major ghrelin forms, acylated ghrelin and des-acylated ghrelin, are novel gastrointestinal hormones. Moreover, emerging evidence indicates that these peptides may have other functions including neuro- and vaso-protection. Here, we investigated whether post-stroke treatment with acylated ghrelin or des-acylated ghrelin could improve functional and histological endpoints of stroke outcome in mice after transient middle cerebral artery occlusion (tMCAo). We found that des-acylated ghrelin (1 mg/kg) improved neurological and functional performance, reduced infarct and swelling, and decreased apoptosis. In addition, it reduced blood-brain barrier (BBB) disruption in vivo and attenuated the hyper-permeability of mouse cerebral microvascular endothelial cells after oxygen glucose deprivation and reoxygenation (OGD + RO). By contrast, acylated ghrelin (1 mg/kg or 5 mg/kg) had no significant effect on these endpoints of stroke outcome. Next we found that des-acylated ghrelin's vasoprotective actions were associated with increased expression of tight junction proteins (occludin and claudin-5), and decreased cell death. Moreover, it attenuated superoxide production, Nox activity and expression of 3-nitrotyrosine. Collectively, these results demonstrate that post-stroke treatment with des-acylated ghrelin, but not acylated ghrelin, protects against ischaemia/reperfusion-induced brain injury and swelling, and BBB disruption, by reducing oxidative and/or nitrosative damage.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Lesiones Encefálicas/metabolismo , Ghrelina/metabolismo , Sustancias Protectoras/metabolismo , Accidente Cerebrovascular/complicaciones , Acilación , Animales , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/etiología , Células Endoteliales/metabolismo , Ghrelina/administración & dosificación , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Oxígeno/metabolismo , Sustancias Protectoras/administración & dosificación
12.
Neuroendocrinology ; 103(3-4): 223-9, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26088805

RESUMEN

BACKGROUND/AIMS: Ciliary neurotrophic factor (CNTF) exerts powerful anorectic effects and has been suggested to regulate long-term energy balance by inducing adult neurogenesis in the arcuate nucleus of the hypothalamus. METHODS: The CNTF analogue, Axokine, was infused into the lateral ventricle of high-fat-fed mice for 1 week. Food intake, energy expenditure, body mass, glucose metabolism, and neurogenesis in the arcuate nucleus (ARC) of the hypothalamus were assessed 3 weeks after cessation of Axokine treatment. RESULTS: Short-term administration of Axokine induced an anorexic response but did not promote sustained weight loss. Instead, a rapid rebound in food intake and body mass occurred immediately after cessation of Axokine treatment, and this tended to reduce insulin sensitivity. Immunolabeling of 5-bromo-2'-deoxyuridine revealed limited neurogenesis in the ARC 3 weeks after Axokine treatment. CONCLUSION: These findings suggest that Axokine/CNTF does not induce substantial or sustained ARC neurogenesis or contribute to the long-term regulation of energy balance in mice.


Asunto(s)
Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Factor Neurotrófico Ciliar/farmacología , Metabolismo Energético/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Análisis de Varianza , Animales , Peso Corporal/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Glucosa/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Inyecciones Intraventriculares , Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuropéptido Y/genética , Neuropéptido Y/metabolismo , Consumo de Oxígeno/efectos de los fármacos , Factores de Tiempo
13.
Clin Exp Pharmacol Physiol ; 43(4): 468-75, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26800483

RESUMEN

The ghrelin gene is expressed in the stomach where it ultimately encodes up to three peptides, namely, acylated ghrelin, des-acylated ghrelin and obestatin, which all have neuroendocrine roles. Recently, the authors' reported that these peptides have important physiological roles in positively regulating vasodilator nitric oxide (NO) production in the cerebral circulation, and may normally suppress superoxide production by the pro-oxidant enzyme, Nox2-NADPH oxidase. To date, the majority of studies using exogenous peptides infer that they may have similar roles in the systemic circulation. Therefore, this study examined whether exogenous and endogenous ghrelin-related peptides modulate NO production and superoxide levels in mouse mesenteric arteries and/or thoracic aorta. Using wire myography, it was found that application of exogenous acylated ghrelin, des-acylated ghrelin or obestatin to mouse thoracic aorta or mesenteric arteries failed to elicit a vasorelaxation response, whereas all three peptides elicited vasorelaxation responses of rat thoracic aorta. Also, none of the peptides modulated mouse aortic superoxide levels as measured by L-012-enhanced chemiluminescence. Next, it was found that NO bioactivity and superoxide levels were unaffected in the thoracic aorta from ghrelin-deficient mice when compared with wild-type mice. Lastly, using novel GHSR-eGFP reporter mice in combination with double-labelled immunofluorescence, no evidence was found for the growth hormone secretagogue receptor (GHSR1a) in the throracic aorta, which is the only functional ghrelin receptor identified to date. Collectively these findings demonstrate that, in contrast to systemic vessels of other species (e.g. rat and human) and mouse cerebral vessels, ghrelin-related peptides do not modulate vasodilator NO production or superoxide levels in mouse systemic arteries.


Asunto(s)
Aorta Torácica/efectos de los fármacos , Ghrelina/farmacología , Arterias Mesentéricas/efectos de los fármacos , Óxido Nítrico/biosíntesis , Superóxidos/metabolismo , Vasodilatación/efectos de los fármacos , Animales , Aorta Torácica/metabolismo , Aorta Torácica/fisiología , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Arterias Mesentéricas/metabolismo , Arterias Mesentéricas/fisiología , Ratones , Óxido Nítrico/metabolismo , Ratas , Receptores de Ghrelina/metabolismo
14.
J Neurosci ; 34(49): 16309-19, 2014 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-25471570

RESUMEN

The hypothalamic NPY system plays an important role in regulating food intake and energy expenditure. Different biological actions of NPY are assigned to NPY receptor subtypes. Recent studies demonstrated a close relationship between food intake and growth hormone (GH) secretion; however, the mechanism through which endogenous NPY modulates GH release remains unknown. Moreover, conclusive evidence demonstrating a role for NPY and Y-receptors in regulating the endogenous pulsatile release of GH does not exist. We used genetically modified mice (germline Npy, Y1, and Y2 receptor knock-out mice) to assess pulsatile GH secretion under both fed and fasting conditions. Deletion of NPY did not impact fed GH release; however, it reversed the fasting-induced suppression of pulsatile GH secretion. The recovery of GH secretion was associated with a reduction in hypothalamic somatotropin release inhibiting factor (Srif; somatostatin) mRNA expression. Moreover, observations revealed a differential role for Y1 and Y2 receptors, wherein the postsynaptic Y1 receptor suppresses GH secretion in fasting. In contrast, the presynaptic Y2 receptor maintains normal GH output under long-term ad libitum-fed conditions. These data demonstrate an integrated neural circuit that modulates GH release relative to food intake, and provide essential information to address the differential roles of Y1 and Y2 receptors in regulating the release of GH under fed and fasting states.


Asunto(s)
Ayuno/fisiología , Hormona del Crecimiento/metabolismo , Neuropéptido Y/fisiología , Receptores de Neuropéptido Y/fisiología , Animales , Glucemia , Hormona Liberadora de Hormona del Crecimiento/biosíntesis , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Noqueados , Neuropéptido Y/sangre , Neuropéptido Y/genética , Péptido YY/sangre , Receptores de Neuropéptido Y/genética , Somatostatina/biosíntesis
15.
FASEB J ; 28(1): 35-44, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24022403

RESUMEN

Subjects characterized as cortisol high responders (HRs) consume more calories after stress, but it is unknown whether cortisol responsiveness predicts a propensity for obesity. Female sheep with either high or low cortisol responses to adrenocorticotropin (ACTH) were identified. Body composition was similar in HRs and cortisol low responders (LRs), but the HRs had greater (P<0.01) adiposity than did the LRs (40.5±0.7 vs. 35.8±1.4%) after high-energy feeding, despite comparable food intake. Postprandial thermogenesis in muscle temperature was 0.8 ± 0.08°C higher in the LRs than in the HRs (P<0.01), whereas feeding-induced changes in fat temperature were similar. Leptin and insulin sensitivity were similar in the HRs and LRs. Feeding lowered (P<0.001) the respiratory control ratio in muscle (HRs 9.2±0.8-5.2±1.2; LRs 8.4±0.5-5.2±0.7), indicative of increased uncoupled respiration. Also in muscle, the feeding-induced increases in uncoupling protein (UCP)-3 (fold increase: HRs, 2.4; LRs, 2.0), ryanodine 1 receptor (RyR1; fold increase: HRs 3.1; LRs 2.1), and sarcoendoplasmic reticulum Ca(2+)-dependent ATPase (fold increase: HRs 1.5; LRs 1.6) were equivalent in the HRs and LRs. Sequencing of mitochondrial DNA revealed no haplotypic differences between the 2 groups. We conclude that predisposition to obesity can be predicted by cortisol responsiveness to an ACTH challenge and that the response is due to innate differences in muscle thermogenesis.


Asunto(s)
Hidrocortisona/farmacología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Obesidad/metabolismo , Termogénesis/efectos de los fármacos , Animales , Western Blotting , Composición Corporal/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Femenino , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/metabolismo , Leptina/farmacología , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Sistema Hipófiso-Suprarrenal/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Ovinos
16.
Breast Cancer Res Treat ; 147(1): 193-201, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25056185

RESUMEN

Aromatase converts androgens into estrogens and its expression within adipose stromal cells (ASCs) is believed to be the major driver of estrogen-dependent cancers in older women. Ghrelin is a gut-hormone that is involved in the regulation of appetite and known to bind to and activate the cognate ghrelin receptor, GHSR1a. The unacylated form of ghrelin, des-acyl ghrelin, binds weakly to GHSR1a but has been shown to play an important role in regulating a number of physiological processes. The aim of this study was to determine the effect of ghrelin and des-acyl ghrelin on aromatase in primary human ASCs. Primary human ASCs were isolated from adipose tissue of women undergoing cosmetic surgery. Real-time PCR and tritiated water-release assays were performed to examine the effect of treatment on aromatase transcript expression and aromatase activity, respectively. Treatments included ghrelin, des-acyl ghrelin, obestatin, and capromorelin (GHSR1a agonist). GHSR1a protein expression was assessed by Western blot and effects of treatment on Ca(2+) and cAMP second messenger systems were examined using the Flexstation assay and the Lance Ultra cAMP kit, respectively. Results demonstrate that pM concentrations of ghrelin and des-acyl ghrelin inhibit aromatase transcript expression and activity in ASCs under basal conditions and in PGE2-stimulated cells. Moreover, the effects of ghrelin and des-acyl ghrelin are mediated via effects on aromatase promoter PII-specific transcripts. Neither the GHSR1a-specific agonist capromorelin nor obestatin had any effect on aromatase transcript expression or activity. Moreover, GHSR1a protein was undetectable by Western blot and neither ghrelin nor capromorelin elicited a calcium response in ASCs. Finally, ghrelin caused a significant decrease in basal and forskolin-stimulated cAMP in ASC. These findings suggest that ghrelin acts at alternate receptors in ASCs by decreasing intracellular cAMP levels. Ghrelin mimetics may be useful in the treatment of estrogen-dependent breast cancer.


Asunto(s)
Tejido Adiposo/enzimología , Aromatasa/química , Mama/enzimología , AMP Cíclico/metabolismo , Ghrelina/farmacología , Células del Estroma/enzimología , Tejido Adiposo/citología , Tejido Adiposo/efectos de los fármacos , Aromatasa/genética , Aromatasa/metabolismo , Western Blotting , Mama/citología , Mama/efectos de los fármacos , Calcio/metabolismo , Células Cultivadas , Femenino , Humanos , Piperidinas/farmacología , Pirazoles/farmacología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Ghrelina/agonistas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células del Estroma/citología , Células del Estroma/efectos de los fármacos
17.
Nature ; 454(7206): 846-51, 2008 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-18668043

RESUMEN

The gut-derived hormone ghrelin exerts its effect on the brain by regulating neuronal activity. Ghrelin-induced feeding behaviour is controlled by arcuate nucleus neurons that co-express neuropeptide Y and agouti-related protein (NPY/AgRP neurons). However, the intracellular mechanisms triggered by ghrelin to alter NPY/AgRP neuronal activity are poorly understood. Here we show that ghrelin initiates robust changes in hypothalamic mitochondrial respiration in mice that are dependent on uncoupling protein 2 (UCP2). Activation of this mitochondrial mechanism is critical for ghrelin-induced mitochondrial proliferation and electric activation of NPY/AgRP neurons, for ghrelin-triggered synaptic plasticity of pro-opiomelanocortin-expressing neurons, and for ghrelin-induced food intake. The UCP2-dependent action of ghrelin on NPY/AgRP neurons is driven by a hypothalamic fatty acid oxidation pathway involving AMPK, CPT1 and free radicals that are scavenged by UCP2. These results reveal a signalling modality connecting mitochondria-mediated effects of G-protein-coupled receptors on neuronal function and associated behaviour.


Asunto(s)
Proteína Relacionada con Agouti/metabolismo , Ghrelina/metabolismo , Canales Iónicos/metabolismo , Proteínas Mitocondriales/metabolismo , Neuronas/metabolismo , Neuropéptido Y/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteína Relacionada con Agouti/genética , Animales , Carnitina O-Palmitoiltransferasa/metabolismo , Ácidos Grasos/metabolismo , Conducta Alimentaria/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Ghrelina/farmacología , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Canales Iónicos/genética , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Potencial de la Membrana Mitocondrial/fisiología , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/fisiología , Proteínas Mitocondriales/genética , Neuronas/efectos de los fármacos , Neuropéptido Y/genética , Fosforilación/efectos de los fármacos , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Proteína Desacopladora 2
18.
J Neuroendocrinol ; 36(5): e13389, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38599683

RESUMEN

Hunger increases the motivation for calorie consumption, often at the expense of low-taste appeal. However, the neural mechanisms integrating calorie-sensing with increased motivation for calorie consumption remain unknown. Agouti-related peptide (AgRP) neurons in the arcuate nucleus of the hypothalamus sense hunger, and the ingestion of caloric solutions promotes dopamine release in the absence of sweet taste perception. Therefore, we hypothesised that metabolic-sensing of hunger by AgRP neurons would be essential to promote dopamine release in the nucleus accumbens in response to caloric, but not non-caloric solutions. Moreover, we examined whether metabolic sensing in AgRP neurons affected taste preference for bitter solutions under conditions of energy need. Here we show that impaired metabolic sensing in AgRP neurons attenuated nucleus accumbens dopamine release in response to sucrose, but not saccharin, consumption. Furthermore, metabolic sensing in AgRP neurons was essential to distinguish nucleus accumbens dopamine response to sucrose consumption when compared with saccharin. Under conditions of hunger, metabolic sensing in AgRP neurons increased the preference for sucrose solutions laced with the bitter tastant, quinine, to ensure calorie consumption, whereas mice with impaired metabolic sensing in AgRP neurons maintained a strong aversion to sucrose/quinine solutions despite ongoing hunger. In conclusion, we demonstrate normal metabolic sensing in AgRP neurons drives the preference for calorie consumption, primarily when needed, by engaging dopamine release in the nucleus accumbens.


Asunto(s)
Proteína Relacionada con Agouti , Dopamina , Núcleo Accumbens , Sacarosa , Núcleo Accumbens/metabolismo , Animales , Dopamina/metabolismo , Proteína Relacionada con Agouti/metabolismo , Ratones , Masculino , Preferencias Alimentarias/fisiología , Ratones Endogámicos C57BL , Neuronas/metabolismo , Hambre/fisiología , Percepción del Gusto/fisiología
19.
Endocrinology ; 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38980913

RESUMEN

The resurgence of interest in psychedelics as treatments for psychiatric disorders necessitates a better understanding of potential sex differences in response to these substances. Sex as a biological variable (SABV) has been historically neglected in medical research, posing limits to our understanding of treatment efficacy. Human studies have provided insights into the efficacy of psychedelics across various diagnoses and aspects of cognition, yet sex-specific effects remain unclear, making it difficult to draw strong conclusions about sex-dependent differences in response to psychedelic treatments. Compounding this further, animal studies used to understand biological mechanisms of psychedelics predominantly use one sex and present mixed neurobiological and behavioural outcomes. Studies that do include both sexes often do not investigate sex differences further, which may hinder the translation of findings to the clinic. In reviewing sex differences in responses to psychedelics, we will highlight the direct interaction between estrogen (the most extensively studied steroid hormone) and the serotonin system (central to the mechanism of action of psychedelics), and the potential that estrogen-serotonin interactions may influence the efficacy of psychedelics in female subjects. Estrogen influences serotonin neurotransmission by affecting its synthesis and release, as well as modulating the sensitivity and responsiveness of serotonin receptor subtypes in the brain. This could potentially influence the efficacy of psychedelics in females by modifying their therapeutic efficacy across menstrual cycles and developmental stages. Investigating this interaction in the context of psychedelic research could aid in the advancement of therapeutic outcomes, especially for conditions with sex-specific prevalence.

20.
Am J Physiol Regul Integr Comp Physiol ; 304(11): R980-90, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23576609

RESUMEN

Ghrelin is a gastrointestinal hormone with a well-characterized role in feeding and metabolism. Recent evidence suggests that ghrelin may also be neuroprotective after injury in animal models of cerebral ischemia. Thus exogenous ghrelin treatment can improve cell survival, reduce infarct size, and rescue memory deficits in focal ischemia models, doing so by suppressing inflammation and apoptosis. Endogenous ghrelin plays a key a role in a number of physiological processes, including feeding, metabolism, stress, and anxiety. However, no study has examined whether endogenous ghrelin also contributes to neuroprotection after cerebral ischemia. Here, we aimed to determine whether endogenous ghrelin normally protects against neuronal cell death and cognitive impairments after global cerebral ischemia and whether such changes are linked with inflammation or apoptosis. We used a two-vessel occlusion (2VO) model of global cerebral ischemia in wild-type (wt) and ghrelin knockout (ghr-/-) C57/Bl6J mice. ghr-/- mice had improved cell survival in the Cornu Ammonis(CA)-2/3 region of the hippocampus-a region of significant growth hormone secretagogue receptor expression. They also displayed less cellular degeneration than wt mice after the 2VO (Fluoro-Jade) and had less cognitive impairment in the novel object-recognition test. These outcomes were despite evidence of more neuroinflammation and apoptosis in the ghr-/- and less of a postsurgery hypothermia. Finally, we found that mortality in the week following the 2VO was reduced more in ghr-/- mice than in wt. Overall, these experiments point to a neurodegenerative but antiapoptotic effect of endogenous ghrelin in this model of global ischemia, highlighting that further research is essential before we can apply ghrelin treatments to neurodegenerative insults in the clinic.


Asunto(s)
Isquemia Encefálica/patología , Ghrelina/fisiología , Hipocampo/patología , Accidente Cerebrovascular/patología , Animales , Apoptosis , Astrocitos/patología , Conducta Animal/fisiología , Isquemia Encefálica/genética , Región CA2 Hipocampal/patología , Región CA3 Hipocampal/patología , Cognición/fisiología , Cartilla de ADN , Encefalitis/patología , Ghrelina/genética , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/fisiología , Neuronas/patología , Desempeño Psicomotor/fisiología , Receptores de Ghrelina/fisiología , Reconocimiento en Psicología/fisiología , Accidente Cerebrovascular/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA