Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Int J Mol Sci ; 23(23)2022 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-36499327

RESUMEN

Non-alcoholic fatty liver disease (NAFLD) and hepatocellular carcinoma (HCC) show clear evidence of sexual dimorphism, with a significantly higher incidence in males. Among the determining factors that could explain this sex-based difference, the specific distribution of fat by sex has been suggested as a primary candidate, since obesity is a relevant risk factor. In this context, obesity, considered a low-grade chronic inflammatory pathology and responsible for the promotion of liver disease, could lead to sexual dimorphism in the expression profile of genes related to tumor development. When we compared the expression levels of genes associated with the early stages of carcinogenesis in the liver between male and female diet-induced obesity (DIO) rats, we observed that the expression pattern was similar in obese male and female animals. Interestingly, the SURVIVIN/BIRC5 oncogene showed a higher expression in male DIO rats than in female DIO and lean rats. This trend related to sexual dimorphism was observed in leukocytes from patients with obesity, although the difference was not statistically significant. In conclusion, this study evidenced a similar pattern in the expression of most carcinogenesis-related genes in the liver, except SUVIVIN/BIRC5, which could be a predictive biomarker of liver carcinogenesis predisposition in male patients with obesity.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Enfermedad del Hígado Graso no Alcohólico , Masculino , Femenino , Ratas , Animales , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Caracteres Sexuales , Hígado/metabolismo , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Carcinogénesis/metabolismo , Obesidad/complicaciones , Obesidad/genética , Obesidad/metabolismo , Expresión Génica , Dieta Alta en Grasa
2.
Int J Obes (Lond) ; 45(3): 502-514, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33139887

RESUMEN

BACKGROUND/OBJECTIVES: Liver-expressed antimicrobial peptide 2 (LEAP-2) was recently identified as an endogenous non-competitive allosteric antagonist of the growth hormone secretagogue receptor 1a (GHSR1a). LEAP-2 blunts ghrelin-induced feeding and its plasma levels are modulated in response to nutritional status in humans. Despite the relevant role of ghrelin in childhood, puberty, and childhood obesity, the potential implication of LEAP-2 in these aspects remains totally unknown. We aimed to investigate the regulation of circulating plasma LEAP-2 in childhood and adolescent either lean or obese. METHODS AND RESULTS: Plasma levels of LEAP-2 were analyzed in a cross-sectional study with lean and obese children and adolescents (n = 150). Circulating LEAP-2 levels were significantly higher in girls than in boys independently of whether they were obese or lean. In addition, LEAP-2 was significantly increased (p < 0.001) in pubertal than in prepubertal girls, while no changes were found in boys between both developmental stages. Moreover, in girls LEAP-2 was positively correlated with insulin, IGF-1, HOMA-IR and triglycerides and negatively with ghrelin. In boys, LEAP-2 was positively correlated with leptin and negatively with vitamin D levels. CONCLUSION: This study reveals a sexual dimorphism in LEAP-2 levels in children and adolescents. These changes and the higher levels during puberty imply that LEAP-2 may contribute to some of the biological adaptations occurring during pubertal development in terms of food intake, energy balance, growth rate, and puberty onset. Future studies assessing LEAP-2 levels in longitudinal studies and its implications in growth rate, puberty onset, and reproductive hormones will help to understand the relevance of this hormone in this stage of life.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/sangre , Pubertad/sangre , Adolescente , Proteínas Sanguíneas , Niño , Preescolar , Estudios Transversales , Femenino , Ghrelina/sangre , Humanos , Masculino , Obesidad Infantil/sangre , Obesidad Infantil/epidemiología
3.
J Cell Mol Med ; 24(18): 10958-10969, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32767737

RESUMEN

The modulation of acetylcholine (ACh) release by botulinum toxin injection into epicardial fat diminishes atrial fibrillation (AF) recurrence. These results suggest an interaction between autonomic imbalance and epicardial fat as risk factors of AF. Our aim was to study the inflammatory, lipidic and fibroblastic profile of epicardial stroma from patients who underwent open-heart surgery, their regulation by cholinergic activity and its association with AF. We performed in vitro and ex vivo assays from paired subcutaneous and epicardial stromal cells or explants from 33 patients. Acute ACh effects in inflammation and lipid-related genes were analysed by qPCR, in intracellular calcium mobilization were performed by Fluo-4 AM staining and in neutrophil migration by trans-well assays. Chronic ACh effects on lipid accumulation were visualized by AdipoRed. Plasma protein regulation by parasympathetic denervation was studied in vagotomized rats. Our results showed a higher pro-inflammatory profile in epicardial regarding subcutaneous stromal cells. Acute ACh treatment up-regulated monocyte chemoattractant protein 1 levels. Chronic ACh treatment improved lipid accumulation within epicardial stromal cells (60.50% [22.82-85.13] vs 13.85% [6.17-23.16], P < .001). Additionally, patients with AF had higher levels of fatty acid-binding protein 4 (1.54 ± 0.01 vs 1.47 ± 0.01, P = .005). Its plasma levels were pronouncedly declined in vagotomized rats (2.02 ± 0.21 ng/mL vs 0.65 ± 0.23 ng/mL, P < .001). Our findings support the characterization of acute or chronic cholinergic activity on epicardial stroma and its association with AF.


Asunto(s)
Acetilcolina/metabolismo , Fibrilación Atrial/metabolismo , Metabolismo de los Lípidos , Pericardio/patología , Células del Estroma/metabolismo , Acetilcolina/farmacología , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Anciano , Animales , Fibrilación Atrial/etiología , Señalización del Calcio , Procedimientos Quirúrgicos Cardíacos , Células Cultivadas , Quimiocina CCL2/biosíntesis , Quimiocina CCL2/genética , Quimiotaxis de Leucocito/efectos de los fármacos , Proteínas de Unión a Ácidos Grasos , Perfilación de la Expresión Génica , Células HL-60 , Humanos , Inflamación , Síndrome Metabólico/metabolismo , Persona de Mediana Edad , Neutrófilos/efectos de los fármacos , Obesidad/complicaciones , Obesidad/fisiopatología , Sistema Nervioso Parasimpático/fisiopatología , Ratas , Ratas Sprague-Dawley , Células del Estroma/efectos de los fármacos , Grasa Subcutánea/metabolismo , Vagotomía
4.
Neuroendocrinology ; 110(11-12): 1042-1054, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31945763

RESUMEN

Linaclotide is a synthetic peptide approved by the FDA for the treatment of constipation-predominant irritable bowel syndrome and chronic constipation. Linaclotide binds and activates the transmembrane receptor guanylate cyclase 2C (Gucy2c). Uroguanylin (UGN) is a 16 amino acid peptide that is mainly secreted by enterochromaffin cells in the duodenum and proximal small intestine. UGN is the endogenous ligand of Gucy2c and decreases body weight in diet-induced obese (DIO) mice via the activation of the thermogenic program in brown adipose tissue. Therefore, we wanted to evaluate whether oral linaclotide could also improve DIO mice metabolic phenotype. In this study, we have demonstrated that DIO mice orally treated with linaclotide exhibited a significant reduction of body weight without modifying food intake. Linaclotide exerts its actions through the central nervous system, and more specifically, via Gucy2c receptors located in the mediobasal hypothalamus, leading to the activation of the sympathetic nervous system to trigger the thermogenic activity of brown fat stimulating energy expenditure. These findings indicate for first time that, in addition to its effects at intestinal level to treat irritable bowel syndrome with constipation and chronic constipation, linaclotide also exerts a beneficial effect in whole body metabolism.


Asunto(s)
Tejido Adiposo Pardo/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Agonistas de la Guanilato Ciclasa C/farmacología , Hipotálamo/efectos de los fármacos , Obesidad/tratamiento farmacológico , Péptidos/farmacología , Receptores de Enterotoxina/efectos de los fármacos , Termogénesis/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Dieta Alta en Grasa , Masculino , Ratones , Ratones Endogámicos C57BL
5.
Hepatology ; 64(4): 1086-104, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27387967

RESUMEN

UNLABELLED: The opioid system is widely known to modulate the brain reward system and thus affect the behavior of humans and other animals, including feeding. We hypothesized that the hypothalamic opioid system might also control energy metabolism in peripheral tissues. Mice lacking the kappa opioid receptor (κOR) and adenoviral vectors overexpressing or silencing κOR were stereotaxically delivered in the lateral hypothalamic area (LHA) of rats. Vagal denervation was performed to assess its effect on liver metabolism. Endoplasmic reticulum (ER) stress was inhibited by pharmacological (tauroursodeoxycholic acid) and genetic (overexpression of the chaperone glucose-regulated protein 78 kDa) approaches. The peripheral effects on lipid metabolism were assessed by histological techniques and western blot. We show that in the LHA κOR directly controls hepatic lipid metabolism through the parasympathetic nervous system, independent of changes in food intake and body weight. κOR colocalizes with melanin concentrating hormone receptor 1 (MCH-R1) in the LHA, and genetic disruption of κOR reduced melanin concentrating hormone-induced liver steatosis. The functional relevance of these findings was given by the fact that silencing of κOR in the LHA attenuated both methionine choline-deficient, diet-induced and choline-deficient, high-fat diet-induced ER stress, inflammation, steatohepatitis, and fibrosis, whereas overexpression of κOR in this area promoted liver steatosis. Overexpression of glucose-regulated protein 78 kDa in the liver abolished hypothalamic κOR-induced steatosis by reducing hepatic ER stress. CONCLUSIONS: This study reveals a novel hypothalamic-parasympathetic circuit modulating hepatic function through inflammation and ER stress independent of changes in food intake or body weight; these findings might have implications for the clinical use of opioid receptor antagonists. (Hepatology 2016;64:1086-1104).


Asunto(s)
Dieta , Estrés del Retículo Endoplásmico , Hormonas Hipotalámicas/fisiología , Hipotálamo/fisiología , Hepatopatías/etiología , Melaninas/fisiología , Hormonas Hipofisarias/fisiología , Receptores Opioides kappa/fisiología , Animales , Inflamación/complicaciones , Inflamación/etiología , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley
6.
Int J Neuropsychopharmacol ; 18(9)2015 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-25655433

RESUMEN

BACKGROUND: Cumulative data indicate that the endocannabinoid system plays a major role in feeding behavior and energy balance. Genetic silencing of cannabinoid receptor type 1 (CB1) reduces body weight gain, independently of food intake. METHODS: In this work, we investigated whether the hypothalamic neuropeptide expression pattern supports the absence of the anorexigenic response observed under constitutive CB1 ablation, by using neuronal CB1 conditional null mice (CamK-CB1-KO) and whole body CB1 null mice (CB1-KO). RESULTS: Our data showed that both CB1 null models display a marked decrease in proopiomelanocortin (POMC) and cocaine-amphetamine-regulated transcript (CART) expression in the arcuate nucleus of the hypothalamus (ARC). CONCLUSIONS: This evidence suggests that a lack of hypophagia is associated with the suppression of ARC anorexigenic neuropeptides and that behavioral changes in food intake (or lack thereof) after constitutive CB1 ablation are likely mediated by impaired melanocortin and CART signaling in the hypothalamus.


Asunto(s)
Anorexia/metabolismo , Núcleo Arqueado del Hipotálamo/metabolismo , Conducta Alimentaria/fisiología , Proteínas del Tejido Nervioso/metabolismo , Proopiomelanocortina/metabolismo , Receptor Cannabinoide CB1/fisiología , Animales , Conducta Animal , Peso Corporal , Antagonistas de Receptores de Cannabinoides/farmacología , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor Cannabinoide CB1/antagonistas & inhibidores , Receptor Cannabinoide CB1/deficiencia , Receptor Cannabinoide CB1/genética
7.
Gastroenterology ; 144(3): 636-649.e6, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23142626

RESUMEN

BACKGROUND & AIMS: Specific neuronal circuits modulate autonomic outflow to liver and white adipose tissue. Melanin-concentrating hormone (MCH)-deficient mice are hypophagic, lean, and do not develop hepatosteatosis when fed a high-fat diet. Herein, we sought to investigate the role of MCH, an orexigenic neuropeptide specifically expressed in the lateral hypothalamic area, on hepatic and adipocyte metabolism. METHODS: Chronic central administration of MCH and adenoviral vectors increasing MCH signaling were performed in rats and mice. Vagal denervation was performed to assess its effect on liver metabolism. The peripheral effects on lipid metabolism were assessed by real-time polymerase chain reaction and Western blot. RESULTS: We showed that the activation of MCH receptors promotes nonalcoholic fatty liver disease through the parasympathetic nervous system, whereas it increases fat deposition in white adipose tissue via the suppression of sympathetic traffic. These metabolic actions are independent of parallel changes in food intake and energy expenditure. In the liver, MCH triggers lipid accumulation and lipid uptake, with c-Jun N-terminal kinase being an essential player, whereas in adipocytes MCH induces metabolic pathways that promote lipid storage and decreases lipid mobilization. Genetic activation of MCH receptors or infusion of MCH specifically in the lateral hypothalamic area modulated hepatic lipid metabolism, whereas the specific activation of this receptor in the arcuate nucleus affected adipocyte metabolism. CONCLUSIONS: Our findings show that central MCH directly controls hepatic and adipocyte metabolism through different pathways.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Adiposidad/fisiología , Área Hipotalámica Lateral/fisiología , Hormonas Hipotalámicas/fisiología , Hígado/metabolismo , Melaninas/fisiología , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Hormonas Hipofisarias/fisiología , Adipocitos/efectos de los fármacos , Tejido Adiposo/efectos de los fármacos , Animales , Ingestión de Alimentos , Ácidos Grasos/metabolismo , Hígado Graso/metabolismo , Hígado Graso/fisiopatología , Área Hipotalámica Lateral/efectos de los fármacos , Hormonas Hipotalámicas/administración & dosificación , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/fisiología , Lipogénesis/efectos de los fármacos , Lipogénesis/fisiología , Hígado/efectos de los fármacos , Masculino , Melaninas/administración & dosificación , Ratones , Enfermedad del Hígado Graso no Alcohólico , Hormonas Hipofisarias/administración & dosificación , Ratas , Ratas Sprague-Dawley , Receptores de la Hormona Hipofisaria/agonistas , Receptores de la Hormona Hipofisaria/fisiología , Nervio Vago/efectos de los fármacos , Nervio Vago/fisiología , Nervio Vago/fisiopatología
8.
FASEB J ; 27(4): 1561-71, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23292070

RESUMEN

Growth hormone (GH) is a major anabolic hormone and the primary regulator of organism growth. Its transcription is triggered by GH-releasing hormone (GHRH) through the transcription factor cAMP response element-binding protein (CREB) and by caloric intake. In contrast, the deacetylase Sirt1 is activated by caloric restriction. Therefore, the present study investigates how Sirt1 affects CREB function and GH synthesis. Sirt1 pharmacological activation with resveratrol (IC50=87 µM) suppressed GHRH-induced GH secretion from rat anterior pituitary cells in vivo and in vitro, while vehicle controls showed no effect. Resveratrol's effects were abolished after knocking down Sirt1 with RNA interference, but not in control scrambled siRNA-transfected rat somatotrophs, confirming the Sirt1 specificity. Sirt1 activation and overexpression suppressed forskolin-induced CREB-Ser(133) phosphorylation, but no effect was seen with vehicle and empty plasmid controls. The deacetylase-dead mutant Sirt1 retained CREB-Ser(133) phosphorylation by keeping protein phosphatase protein phosphatase 1 activity low. Sirt1 activation suppressed glycogen synthase kinase 3 ß acetylation, and a mutation on the GSK3ß-Lys(205) residue mimicking a hypoacetylated form revealed increased activity. In summary, this is a novel mechanism through which Sirt1 intercepts the cAMP pathway by suppressing CREB transcriptional activation, resulting in decreased GH synthesis.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Hormona del Crecimiento/biosíntesis , Sirtuina 1/metabolismo , Animales , Colforsina/farmacología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Hormona del Crecimiento/genética , Hormona Liberadora de Hormona del Crecimiento/genética , Hormona Liberadora de Hormona del Crecimiento/metabolismo , Masculino , Adenohipófisis/efectos de los fármacos , Adenohipófisis/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Hormona Reguladora de Hormona Hipofisaria/genética , Receptores de Hormona Reguladora de Hormona Hipofisaria/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Activación Transcripcional/fisiología
9.
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
10.
J Biol Chem ; 287(45): 38379-89, 2012 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-22992743

RESUMEN

The maintenance and repair of skeletal muscle are attributable to an elaborate interaction between extrinsic and intrinsic regulatory signals that regulate the myogenic process. In the present work, we showed that obestatin, a 23-amino acid peptide encoded by the ghrelin gene, and the GPR39 receptor are expressed in rat skeletal muscle and are up-regulated upon experimental injury. To define their roles in muscle regeneration, L6E9 cells were used to perform in vitro assays. For the in vivo assays, skeletal muscle tissue was obtained from male rats and maintained under continuous subcutaneous infusion of obestatin. In differentiating L6E9 cells, preproghrelin expression and correspondingly obestatin increased during myogenesis being sustained throughout terminal differentiation. Autocrine action was demonstrated by neutralization of the endogenous obestatin secreted by differentiating L6E9 cells using a specific anti-obestatin antibody. Knockdown experiments by preproghrelin siRNA confirmed the contribution of obestatin to the myogenic program. Furthermore, GPR39 siRNA reduced obestatin action and myogenic differentiation. Exogenous obestatin stimulation was also shown to regulate myoblast migration and proliferation. Furthermore, the addition of obestatin to the differentiation medium increased myogenic differentiation of L6E9 cells. The relevance of the actions of obestatin was confirmed in vivo by the up-regulation of Pax-7, MyoD, Myf5, Myf6, myogenin, and myosin heavy chain (MHC) in obestatin-infused rats when compared with saline-infused rats. These data elucidate a novel mechanism whereby the obestatin/GPR39 system is coordinately regulated as part of the myogenic program and operates as an autocrine signal regulating skeletal myogenesis.


Asunto(s)
Ghrelina/metabolismo , Músculo Esquelético/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Regulación hacia Arriba , Animales , Comunicación Autocrina , Cardiotoxinas/toxicidad , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Línea Celular , Expresión Génica/efectos de los fármacos , Ghrelina/genética , Ghrelina/farmacología , Immunoblotting , Inmunohistoquímica , Masculino , Músculo Esquelético/lesiones , Músculo Esquelético/fisiopatología , Enfermedades Musculares/inducido químicamente , Enfermedades Musculares/patología , Enfermedades Musculares/fisiopatología , Proteína MioD/genética , Proteína MioD/metabolismo , Mioblastos Esqueléticos/citología , Mioblastos Esqueléticos/efectos de los fármacos , Mioblastos Esqueléticos/metabolismo , Miogenina/genética , Miogenina/metabolismo , Interferencia de ARN , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/genética , Regeneración , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
11.
Nat Metab ; 4(7): 901-917, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35879461

RESUMEN

Early-life determinants are thought to be a major factor in the rapid increase of obesity. However, while maternal nutrition has been extensively studied, the effects of breastfeeding by the infant on the reprogramming of energy balance in childhood and throughout adulthood remain largely unknown. Here we show that delayed weaning in rat pups protects them against diet-induced obesity in adulthood, through enhanced brown adipose tissue thermogenesis and energy expenditure. In-depth metabolic phenotyping in this rat model as well as in transgenic mice reveals that the effects of prolonged suckling are mediated by increased hepatic fibroblast growth factor 21 (FGF21) production and tanycyte-controlled access to the hypothalamus in adulthood. Specifically, FGF21 activates GABA-containing neurons expressing dopamine receptor 2 in the lateral hypothalamic area and zona incerta. Prolonged breastfeeding thus constitutes a protective mechanism against obesity by affecting long-lasting physiological changes in liver-to-hypothalamus communication and hypothalamic metabolic regulation.


Asunto(s)
Lactancia Materna , Obesidad , Animales , Femenino , Factores de Crecimiento de Fibroblastos , Humanos , Hipotálamo/metabolismo , Hígado/metabolismo , Ratones , Obesidad/metabolismo , Obesidad/prevención & control , Ratas
12.
Clin Nutr ; 40(9): 5141-5155, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34461588

RESUMEN

Chronic heart failure (CHF) is frequently associated with the involuntary loss of body weight and muscle wasting, which can determine the course of the disease and its prognosis. While there is no gold standard malnutrition screening tool for their detection in the CHF population, several bioelectrical and imaging methods have been used to assess body composition in these patients (such as Dual Energy X-Ray Absorptiometry and muscle ultrasound, among other techniques). In addition, numerous nutritional biomarkers have been found to be useful in the determination of the nutritional status. Nutritional considerations include the slow and progressive supply of nutrients, avoiding high volumes, which could ultimately lead to refeeding syndrome and worsen the clinical picture. If oral feeding is insufficient, hypercaloric and hyperproteic supplementation should be considered. ß-Hydroxy-ß-methylbutyrate and omega-3 polyunsaturated fatty acid administration prove to be beneficial in certain patients with CHF, and several interventional studies with micronutrient supplementation have also described their possible role in these subjects. Taking into account that CHF is sometimes associated with gastrointestinal dysfunction, parenteral nutritional support may be required in selected cases. In addition, potential therapeutic options regarding nutritional state and muscle wasting have also been tested in clinical studies. This review summarises the scientific evidence that demonstrates the necessity to carry out a careful nutritional evaluation and nutritional treatment to prevent or improve cardiac cachexia and sarcopenia in CHF, as well as improve its course.


Asunto(s)
Caquexia/diagnóstico , Insuficiencia Cardíaca/complicaciones , Evaluación Nutricional , Apoyo Nutricional/métodos , Sarcopenia/diagnóstico , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores/análisis , Composición Corporal , Caquexia/etiología , Caquexia/terapia , Suplementos Dietéticos , Femenino , Insuficiencia Cardíaca/fisiopatología , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Sarcopenia/etiología , Sarcopenia/terapia
13.
Biochem Biophys Res Commun ; 390(4): 1377-81, 2009 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-19895783

RESUMEN

Obestatin, the ghrelin-associated peptide, showed to activate MAPK signaling with no effect on Akt nor cell proliferating activity in rat tumor somatotroph cells (growth cells, GC). A sequential analysis of the obestatin transmembrane signaling pathway indicated a route involving the consecutive activation of G(i), PI3k, novel PKCepsilon, and Src for ERK1/2 activation. Furthermore, obestatin treatment triggers growth hormone (GH) release in the first 30min, being more acute at 15min. At 1h, obestatin treated cells showed the same levels in GH secretion than controls. Added to this functionality, obestatin was secreted by GC cells. Based on the capacity to stimulate GH release from somatotroph cells, obestatin may act directly in the pituitary through an autocrine/paracrine mechanism.


Asunto(s)
Ghrelina/farmacología , Hormona del Crecimiento/metabolismo , Somatotrofos/efectos de los fármacos , Animales , Línea Celular Tumoral , Activación Enzimática , Ratones , Proteína Quinasa 3 Activada por Mitógenos/biosíntesis , Fosfatidilinositol 3-Quinasas/biosíntesis , Proteína Quinasa C-epsilon/biosíntesis , Ratas , Somatotrofos/enzimología , Somatotrofos/metabolismo , Familia-src Quinasas/biosíntesis
14.
Nutrients ; 11(4)2019 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-30935076

RESUMEN

The gastrointestinal-brain axis is a key mediator of the body weight and energy homeostasis regulation. Uroguanylin (UGN) has been recently proposed to be a part of this gut-brain axis regulating food intake, body weight and energy expenditure. Expression of UGN is regulated by the nutritional status and dependent on leptin levels. However, the exact molecular mechanisms underlying this UGN-leptin metabolic regulation at a hypothalamic level still remains unclear. Using leptin resistant diet-induced obese (DIO) mice, we aimed to determine whether UGN could improve hypothalamic leptin sensitivity. The present work demonstrates that the central co-administration of UGN and leptin potentiates leptin's ability to decrease the food intake and body weight in DIO mice, and that UGN activates the hypothalamic signal transducer and activator of transcription 3 (STAT3) and phosphatidylinositide 3-kinases (PI3K) pathways. At a functional level, the blockade of PI3K, but not STAT3, blunted UGN-mediated leptin responsiveness in DIO mice. Overall, these findings indicate that UGN improves leptin sensitivity in DIO mice.


Asunto(s)
Ingestión de Alimentos/efectos de los fármacos , Leptina/metabolismo , Péptidos Natriuréticos/metabolismo , Obesidad/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Dieta/efectos adversos , Hipotálamo/metabolismo , Ratones , Ratones Obesos , Obesidad/etiología , Fosfatidilinositol 3-Quinasa/metabolismo , Factor de Transcripción STAT3/metabolismo
15.
J Exp Med ; 216(5): 1108-1119, 2019 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-30944152

RESUMEN

Hepatocellular carcinoma (HCC) is the sixth most common cancer type and the fourth leading cause of cancer-related death. This cancer appears with higher incidence in men and during obesity; however, the specific mechanisms underlying this correlation are unknown. Adipose tissue, a key organ in metabolic syndrome, shows evident gender disparities in the production of adipokines. Levels of the important adipokine adiponectin decrease in men during puberty, as well as in the obese state. Here, we show that this decrease in adiponectin levels is responsible for the increased liver cancer risk in males. We found that testosterone activates the protein JNK in mouse and human adipocytes. JNK-mediated inhibition of adiponectin secretion increases liver cancer cell proliferation, since adiponectin protects against liver cancer development through the activation of AMP-activated protein kinase (AMPK) and p38α. This study provides insight into adipose tissue to liver crosstalk and its gender relation during cancer development, having the potential to guide strategies for new cancer therapeutics.


Asunto(s)
Adiponectina/sangre , Carcinoma Hepatocelular/epidemiología , Cálculos Biliares/sangre , Neoplasias Hepáticas/epidemiología , Proteínas Quinasas Activadas por AMP/metabolismo , Adipocitos/metabolismo , Adiponectina/genética , Tejido Adiposo/metabolismo , Animales , Estudios de Cohortes , Femenino , Cálculos Biliares/cirugía , Humanos , Incidencia , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Obesidad/metabolismo , Factores Sexuales
16.
Nutrients ; 11(6)2019 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-31207920

RESUMEN

Angiopoietin-like protein 4 (ANGPTL-4) regulates lipidic metabolism and affects energy homeostasis. However, its function in children with obesity remains unknown. We investigated plasma ANGPTL-4 levels in children and its relationship with body mass index (BMI) and different lipidic parameters such as free fatty acids (FFA). Plasma ANGPTL-4 levels were analyzed in two different cohorts. In the first cohort (n = 150, age 3-17 years), which included children with normal weight or obesity, we performed a cross-sectional study. In the second cohort, which included only children with obesity (n = 20, age 5-18 years) followed up for two years after an intervention for weight loss, in which we performed a longitudinal study measuring ANGPTL-4 before and after BMI-loss. In the cross-sectional study, circulating ANGPTL-4 levels were lower in children with obesity than in those with normal weight. Moreover, ANGPTL-4 presented a negative correlation with BMI, waist circumference, weight, insulin, homeostasis model assessment of insulin resistance index (HOMA index), triglycerides, and leptin, and a positive correlation with FFA and vitamin-D. In the longitudinal study, the percent change in plasma ANGPTL-4 was correlated with the percent change in FFA, total-cholesterol and high-density lipoprotein cholesterol. This study reveals a significant association of ANGPTL-4 with pediatric obesity and plasma lipid profile.


Asunto(s)
Proteína 4 Similar a la Angiopoyetina/sangre , Lípidos/sangre , Obesidad/sangre , Obesidad/epidemiología , Adolescente , Índice de Masa Corporal , Niño , Preescolar , Estudios Transversales , Humanos , Peso Corporal Ideal/fisiología , Estudios Longitudinales
17.
Nat Metab ; 1(8): 811-829, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31579887

RESUMEN

Dopamine signaling is a crucial part of the brain reward system and can affect feeding behavior. Dopamine receptors are also expressed in the hypothalamus, which is known to control energy metabolism in peripheral tissues. Here we show that pharmacological or chemogenetic stimulation of dopamine receptor 2 (D2R) expressing cells in the lateral hypothalamic area (LHA) and the zona incerta (ZI) decreases body weight and stimulates brown fat activity in rodents in a feeding-independent manner. LHA/ZI D2R stimulation requires an intact sympathetic nervous system and orexin system to exert its action and involves inhibition of PI3K in the LHA/ZI. We further demonstrate that, as early as 3 months after onset of treatment, patients treated with the D2R agonist cabergoline experience an increase in energy expenditure that persists for one year, leading to total body weight and fat loss through a prolactin-independent mechanism. Our results may provide a mechanistic explanation for how clinically used D2R agonists act in the CNS to regulate energy balance.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Dopamina/metabolismo , Hipotálamo/metabolismo , Transducción de Señal , Termogénesis/fisiología , Animales , Bromocriptina/administración & dosificación , Bromocriptina/farmacología , Femenino , Humanos , Hipotálamo/efectos de los fármacos , Inyecciones Intraventriculares , Masculino , Ratas
18.
J Physiol Biochem ; 74(1): 111-118, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28744831

RESUMEN

Vitamin D deficiency is associated with wide range of pathologies. Some evidences have shown that low vitamin D circulating levels in children and adolescent are related to fat mass and obesity. The objectives of the present study were to characterize vitamin D status in children and adolescents and to determine if serum 25-hydroxyvitamin D (25(OH)D) concentration is related to adiposity assessed by body mass index (BMI). Serum 25(OH)D levels were measured by LIAISON method in 471 children and adolescents (2 to 18 years age) and analyzed according to gender, pubertal period, age, and BMI. An overall prevalence of 25(OH)D insufficiency and deficiency was present in the 67.1%. Lower 25(OH)D levels were found in females (25.56 ± 14.03 vs 29.71 ± 17.10 ng ml-1; P = 0.004) and pubertal children (25.52 ± 13.97 vs 29.21 ± 16.83 ng ml-1; P = 0.011). In addition, an inverse relation of BMI and age on 25(OH)D concentrations was observed in children. In conclusion, low vitamin D status was highly prevalent among children and adolescents. Of note, a non-lineal regression model showed that 39.6% of vitamin D levels variability was explained by BMI. These results indicate that adiposity assessed by BMI impacts vitamin D status.


Asunto(s)
25-Hidroxivitamina D 2/sangre , Adiposidad , Calcifediol/sangre , Fenómenos Fisiológicos Nutricionales Infantiles , Sobrepeso/etiología , Obesidad Infantil/etiología , Deficiencia de Vitamina D/fisiopatología , Adolescente , Índice de Masa Corporal , Niño , Desarrollo Infantil , Preescolar , Estudios Transversales , Femenino , Transición de la Salud , Hospitales Universitarios , Humanos , Masculino , Estado Nutricional , Sobrepeso/sangre , Sobrepeso/epidemiología , Sobrepeso/fisiopatología , Obesidad Infantil/sangre , Obesidad Infantil/epidemiología , Obesidad Infantil/fisiopatología , Prevalencia , Factores de Riesgo , Índice de Severidad de la Enfermedad , Factores Sexuales , España/epidemiología , Deficiencia de Vitamina D/sangre , Deficiencia de Vitamina D/epidemiología , Deficiencia de Vitamina D/etiología
19.
Sci Rep ; 8(1): 14541, 2018 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-30266914

RESUMEN

Uroguanylin is a 16 amino acid peptide that constitutes a key component of the gut- brain axis with special relevance in body weight regulation. In childhood and adolescence, periods of life with notable metabolic changes; limited data exist, with measurements of pro-uroguanylin in adolescence but not in prepubertal children. This study investigates pro-uroguanylin circulating levels in children with obesity and its relationship with obesity, sex and pubertal development. We analyzed circulating prouroguanylin levels in 117 children (62) and adolescents (55), including 73 with obesity and 44 with normal weight. The pro-uroguanylin concentration is higher in lean girls during pre-puberty versus lean boys (1111 vs 635, p < 0.001). During puberty, pro-uroguanylin levels are higher in lean males with respect to lean females (1060 vs 698, p < 0.01). In girls, a negative correlation exists between pro-uroguanylin and age, Tanner stage, weight, height, BMI (body mass index), waist circumference and plasma levels of leptin and testosterone; a positive correlation was found between pro-uroguanylin and free triiodothyronine. In boys, a positive correlation was found between pro-uroguanylin and BMI and waist circumference and a negative correlation was found with high density lipoprotein-cholesterol. We conclude that a sexual dimorphism exists in circulating pro-uroguanylin levels with respect to BMI. Uroguanylin presents also an opposed circulating pattern during puberty in both sexes.


Asunto(s)
Péptidos Natriuréticos/sangre , Obesidad/sangre , Pubertad/sangre , Adolescente , Índice de Masa Corporal , Niño , Femenino , Humanos , Masculino , Caracteres Sexuales , Maduración Sexual
20.
Mol Nutr Food Res ; 62(10): e1800060, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29536615

RESUMEN

SCOPE: Angiopoietin-like protein 4 (ANGPTL-4) regulates plasma lipoprotein levels, but its relevance in human obesity and type 2 diabetes (T2D) is largely unknown. We aim to investigate the regulation of circulating ANGPTL-4 levels in obesity, T2D, and after changes in body weight. METHODS AND RESULTS: Circulating ANGPTL-4 levels were measured in two different cohorts. First, in a cross-sectional study, we evaluated ANGPTL-4 levels in lean and obese patients with normoglycemia or with altered glucose tolerance (AGT; n = 282). Second, in a longitudinal intervention study, 51 obese participants were evaluated. A hypocaloric diet was prescribed, with follow-up 2 years later. ANGPTL-4 levels were significantly increased in obese patients with AGT compared to lean participants. Moreover, ANGPTL-4 was positively correlated with BMI, waist circumference, fat mass, HbA1c, HOMA-IR, fasting triglycerides, and with inflammatory markers. Participants gaining weight after the follow-up showed increased ANGPTL-4 levels in parallel to increased BMI, fat mass, and fasting glucose, while ANGPTL-4 levels were reduced in participants losing weight. CONCLUSION: Our data support a relevant role of ANGPTL-4 in human obesity and its involvement in long-term body weight changes.


Asunto(s)
Proteína 4 Similar a la Angiopoyetina/sangre , Diabetes Mellitus Tipo 2/sangre , Obesidad/sangre , Adulto , Peso Corporal , Estudios Transversales , Femenino , Intolerancia a la Glucosa , Humanos , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Obesidad/dietoterapia , Circunferencia de la Cintura , Pérdida de Peso
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda