Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Sci Rep ; 10(1): 18581, 2020 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-33122657

RESUMEN

Antipsychotic drugs (AP) are used to treat a multitude of psychiatric conditions including schizophrenia and bipolar disorder. However, APs also have metabolic side effects including increased food intake and body weight, but the underlying mechanisms remain unknown. We previously reported that minocycline (MINO) co-treatment abrogates olanzapine (OLZ)-induced hyperphagia and weight gain in mice. Using this model, we investigated the changes in the pharmacometabolome in the plasma and hypothalamus associated with OLZ-induced hyperphagia and weight gain. Female C57BL/6 mice were divided into groups and fed either i) control, CON (45% fat diet) ii) CON + MINO, iii) OLZ (45% fat diet with OLZ), iv) OLZ + MINO. We identified one hypothalamic metabolite indoxylsulfuric acid and 389 plasma metabolites (including 19 known metabolites) that were specifically associated with AP-induced hyperphagia and weight gain in mice. We found that plasma citrulline, tricosenoic acid, docosadienoic acid and palmitoleic acid were increased while serine, asparagine and arachidonic acid and its derivatives were decreased in response to OLZ. These changes were specifically blocked by co-treatment with MINO. These pharmacometabolomic profiles associated with AP-induced hyperphagia and weight gain provide candidate biomarkers and mechanistic insights related to the metabolic side effects of these widely used drugs.


Asunto(s)
Ingestión de Alimentos/efectos de los fármacos , Hiperfagia/metabolismo , Metaboloma/efectos de los fármacos , Minociclina/farmacología , Olanzapina/toxicidad , Aumento de Peso , Animales , Antibacterianos/farmacología , Antipsicóticos/toxicidad , Femenino , Hiperfagia/inducido químicamente , Hiperfagia/tratamiento farmacológico , Hiperfagia/patología , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Ratones , Ratones Endogámicos C57BL
2.
Horm Behav ; 120: 104690, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31954709

RESUMEN

Changes to neonatal nutrition result in long-lasting impairments in energy balance, which may be described as metabolic programing. Astrocytes, which are interconnected by gap junctions, have emerged as important players in the hypothalamic control of food intake. In order to study the effects of nutritional programming on glial morphology and protein expression, cross-fostered male Wistar rats at postnatal day 3 were assigned to three groups based on litter size: small litter (3 pups per dam, SL), normal litter (10 pups per dam, NL), and large litter (16 pups per dam, LL). Rats from the SL group exhibited higher body weight throughout the study and hyperphagia after weaning. LL animals exhibited hyperphagia, high energy efficiency and catch-up of body weight after weaning. Both the SL and LL groups at postnatal day 60 (PN60) exhibited increased levels of plasma leptin, the Lee index (as an index of obesity), adiposity content, immunoreactivity toward T-cell protein tyrosine phosphatase (TCPTP), and glial fibrillary acidic protein (GFAP) in the arcuate nucleus (ARC) of the hypothalamus. Astrocyte morphology was altered in the ARC of SL and LL animals, and this effect occurred in parallel with a reduction in immunoreactivity toward connexin 30 (CX30). The data obtained demonstrate that both neonatal over- and underfeeding promote not only alterations in the metabolic status but also morphological changes in glial cells in parallel with increasing TCPTP and changes in connexin expression.


Asunto(s)
Fenómenos Fisiológicos Nutricionales de los Animales , Conexinas/genética , Gliosis/etiología , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Adiposidad/fisiología , Animales , Animales Recién Nacidos , Conexinas/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Gliosis/genética , Gliosis/metabolismo , Hiperfagia/complicaciones , Hiperfagia/genética , Hiperfagia/metabolismo , Hiperfagia/patología , Hipotálamo/metabolismo , Tamaño de la Camada/fisiología , Masculino , Obesidad/complicaciones , Obesidad/genética , Obesidad/metabolismo , Obesidad/patología , Embarazo , Proteína Tirosina Fosfatasa no Receptora Tipo 2/metabolismo , Ratas , Ratas Wistar , Factores Sexuales , Factores de Tiempo
3.
Acta Neurochir (Wien) ; 159(5): 865-871, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28224319

RESUMEN

BACKGROUND: Various metabolic diseases induced by eating disorders are some of the most serious and difficult problems for modern public healthcare. However, little is known about hyperphagia, partly because of the lack of a clear definition. Several basic studies have analyzed eating habits using endocrinological or neurophysiological approaches, which have suggested a controlled balance between the hunger and satiety centers in the central nervous system. However, more detailed neuro-radiologic evaluations have not been achieved for the hypothalamus, and evaluations were limited only to the floor of the third ventricles. METHODS: Fine structures of hypothalamic morphology were investigated using high-resolution magnetic resonance imaging in seven patients with hypothalamo-pituitary tumors, who suffered from preoperative hyperphagia-induced severe obesity and metabolic disorders. Body mass index (BMI) varied from 22.4 to 40.5 kg/m2 (mean 32.8 kg/m2). Clinical data were compared with the data of nine patients without hyperphagia and seven healthy volunteers. RESULTS: Morphological evaluation was possible in all patients and control subjects, and patients with hyperphagia had significantly shortened maximum distances between the ependymal layers of the lateral wall of the third ventricle and fornixes (hyperphagia group right side 0.30 mm, left side 0.23 mm vs. patients without hyperphagia group right side 1.60, left side 1.53 vs. healthy group right side 1.73 mm, left side 1.85 mm) (p < 0.01). Two patients achieved postoperative improvement in both clinical and neuro-radiological findings. CONCLUSION: Eating and metabolic disorders are related to strong dysfunction of the medial nuclei of the hypothalamus in patients with hypothalamo-pituitary tumors. We report the first case of dynamic improvement from hyperphagia, with both symptomatic and neuro-radiological findings.


Asunto(s)
Hiperfagia/diagnóstico por imagen , Hipotálamo/diagnóstico por imagen , Obesidad/patología , Adolescente , Adulto , Animales , Estudios de Casos y Controles , Femenino , Humanos , Hiperfagia/complicaciones , Hiperfagia/patología , Hipotálamo/patología , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Obesidad/etiología
4.
J Biochem Mol Toxicol ; 31(6)2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28186389

RESUMEN

Corticosterone plays an important role in feeding behavior. However, its mechanism remains unclear. Therefore, the present study aimed to investigate the effect of corticosterone on feeding behavior. In this study, cumulative food intake was increased by acute corticosterone administration in a dose-dependent manner. Administration of the 5-HT2c receptor agonist m-chlorophenylpiperazin (mCPP) reversed the effect of corticosterone on food intake. The anorectic effects of mCPP were also blocked by the 5-HT2c receptor antagonist RS102221 in corticosterone-treated mice. Both corticosterone and mCPP increased c-Fos expression in hypothalamic nuclei, but not the nucleus of the solitary tract. RS102221 inhibited c-Fos expression induced by mCPP, but not corticosterone. In addition, mCPP had little effect on TH and POMC levels in the hypothalamus. Furthermore, mCPP antagonized decreasing effect of the leptin produced by corticosterone. Taken together, our findings suggest that 5-HT2c receptors and leptin may be involved in the effects of corticosterone-induced hyperphagia.


Asunto(s)
Regulación del Apetito/efectos de los fármacos , Corticosterona/farmacología , Hipotálamo/efectos de los fármacos , Leptina/agonistas , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Receptor de Serotonina 5-HT2C/metabolismo , Animales , Depresores del Apetito/química , Depresores del Apetito/farmacología , Estimulantes del Apetito/administración & dosificación , Estimulantes del Apetito/agonistas , Estimulantes del Apetito/antagonistas & inhibidores , Estimulantes del Apetito/farmacología , Conducta Animal/efectos de los fármacos , Corticosterona/administración & dosificación , Corticosterona/agonistas , Corticosterona/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Ingestión de Energía/efectos de los fármacos , Hiperfagia/sangre , Hiperfagia/inducido químicamente , Hiperfagia/metabolismo , Hiperfagia/patología , Hipotálamo/metabolismo , Hipotálamo/patología , Leptina/antagonistas & inhibidores , Leptina/sangre , Leptina/metabolismo , Ratones Endogámicos ICR , Proteínas del Tejido Nervioso/agonistas , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Neuronas/patología , Especificidad de Órganos , Piperazinas/antagonistas & inhibidores , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-fos/agonistas , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptor de Serotonina 5-HT2C/química , Agonistas del Receptor de Serotonina 5-HT2/farmacología , Antagonistas del Receptor de Serotonina 5-HT2/farmacología , Compuestos de Espiro/farmacología , Sulfonamidas/farmacología , Regulación hacia Arriba/efectos de los fármacos
5.
Nat Commun ; 8: 14014, 2017 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-28072397

RESUMEN

Alcohol intake associates with overeating in humans. This overeating is a clinical concern, but its causes are puzzling, because alcohol (ethanol) is a calorie-dense nutrient, and calorie intake usually suppresses brain appetite signals. The biological factors necessary for ethanol-induced overeating remain unclear, and societal causes have been proposed. Here we show that core elements of the brain's feeding circuits-the hypothalamic Agrp neurons that are normally activated by starvation and evoke intense hunger-display electrical and biochemical hyperactivity on exposure to dietary doses of ethanol in brain slices. Furthermore, by circuit-specific chemogenetic interference in vivo, we find that the Agrp cell activity is essential for ethanol-induced overeating in the absence of societal factors, in single-housed mice. These data reveal how a widely consumed nutrient can paradoxically sustain brain starvation signals, and identify a biological factor required for appetite evoked by alcohol.


Asunto(s)
Proteína Relacionada con Agouti/metabolismo , Etanol/efectos adversos , Hiperfagia/inducido químicamente , Neuronas/metabolismo , Proteína Relacionada con Agouti/genética , Animales , Ingestión de Alimentos/efectos de los fármacos , Electrofisiología/métodos , Femenino , Hiperfagia/patología , Hipotálamo/citología , Hipotálamo/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/efectos de los fármacos , Análisis de la Célula Individual/métodos
6.
Nature ; 538(7624): 253-256, 2016 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-27698417

RESUMEN

Atypical food intake is a primary cause of obesity and other eating and metabolic disorders. Insight into the neural control of feeding has previously focused mainly on signalling mechanisms associated with the hypothalamus, the major centre in the brain that regulates body weight homeostasis. However, roles of non-canonical central nervous system signalling mechanisms in regulating feeding behaviour have been largely uncharacterized. Acetylcholine has long been proposed to influence feeding owing in part to the functional similarity between acetylcholine and nicotine, a known appetite suppressant. Nicotine is an exogenous agonist for acetylcholine receptors, suggesting that endogenous cholinergic signalling may play a part in normal physiological regulation of feeding. However, it remains unclear how cholinergic neurons in the brain regulate food intake. Here we report that cholinergic neurons of the mouse basal forebrain potently influence food intake and body weight. Impairment of cholinergic signalling increases food intake and results in severe obesity, whereas enhanced cholinergic signalling decreases food consumption. We found that cholinergic circuits modulate appetite suppression on downstream targets in the hypothalamus. Together our data reveal the cholinergic basal forebrain as a major modulatory centre underlying feeding behaviour.


Asunto(s)
Regulación del Apetito/fisiología , Prosencéfalo Basal/citología , Prosencéfalo Basal/fisiología , Neuronas Colinérgicas/metabolismo , Conducta Alimentaria/fisiología , Respuesta de Saciedad/fisiología , Acetilcolina/metabolismo , Animales , Peso Corporal/fisiología , Muerte Celular , Colina O-Acetiltransferasa/deficiencia , Agonistas Colinérgicos , Neuronas Colinérgicas/patología , Ingestión de Alimentos/fisiología , Ingestión de Alimentos/psicología , Conducta Alimentaria/psicología , Femenino , Homeostasis , Hiperfagia/enzimología , Hiperfagia/genética , Hiperfagia/patología , Hipotálamo/citología , Hipotálamo/fisiología , Masculino , Ratones , Ratones Noqueados , Modelos Neurológicos , Nicotina/metabolismo , Obesidad/enzimología , Obesidad/genética , Obesidad/patología , Receptores Colinérgicos/metabolismo
7.
Autophagy ; 12(11): 2009-2025, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27533078

RESUMEN

Hypothalamic AMP-activated protein kinase (AMPK) plays important roles in the regulation of food intake by altering the expression of orexigenic or anorexigenic neuropeptides. However, little is known about the mechanisms of this regulation. Here, we report that hypothalamic AMPK modulates the expression of NPY (neuropeptide Y), an orexigenic neuropeptide, and POMC (pro-opiomelanocortin-α), an anorexigenic neuropeptide, by regulating autophagic activity in vitro and in vivo. In hypothalamic cell lines subjected to low glucose availability such as 2-deoxy-d-glucose (2DG)-induced glucoprivation or glucose deprivation, autophagy was induced via the activation of AMPK, which regulates ULK1 and MTOR complex 1 followed by increased Npy and decreased Pomc expression. Pharmacological or genetic inhibition of autophagy diminished the effect of AMPK on neuropeptide expression in hypothalamic cell lines. Moreover, AMPK knockdown in the arcuate nucleus of the hypothalamus decreased autophagic activity and changed Npy and Pomc expression, leading to a reduction in food intake and body weight. AMPK knockdown abolished the orexigenic effects of intraperitoneal 2DG injection by decreasing autophagy and changing Npy and Pomc expression in mice fed a high-fat diet. We suggest that the induction of autophagy is a possible mechanism of AMPK-mediated regulation of neuropeptide expression and control of feeding in response to low glucose availability.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Autofagia , Ingestión de Alimentos , Regulación de la Expresión Génica , Hipotálamo/enzimología , Neuropéptido Y/genética , Proopiomelanocortina/genética , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Animales , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Núcleo Arqueado del Hipotálamo/enzimología , Autofagia/efectos de los fármacos , Autofagia/genética , Proteína 5 Relacionada con la Autofagia/metabolismo , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Peso Corporal/efectos de los fármacos , Línea Celular , Desoxiglucosa/farmacología , Regulación hacia Abajo/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/genética , Activación Enzimática/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Hiperfagia/patología , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones Endogámicos C57BL , Complejos Multiproteicos/metabolismo , Neuropéptido Y/metabolismo , Proopiomelanocortina/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Regulación hacia Arriba/efectos de los fármacos
8.
Cell Metab ; 23(5): 821-36, 2016 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-27133129

RESUMEN

Despite significant advances in our understanding of the biology determining systemic energy homeostasis, the treatment of obesity remains a medical challenge. Activation of AMP-activated protein kinase (AMPK) has been proposed as an attractive strategy for the treatment of obesity and its complications. AMPK is a conserved, ubiquitously expressed, heterotrimeric serine/threonine kinase whose short-term activation has multiple beneficial metabolic effects. Whether these translate into long-term benefits for obesity and its complications is unknown. Here, we observe that mice with chronic AMPK activation, resulting from mutation of the AMPK γ2 subunit, exhibit ghrelin signaling-dependent hyperphagia, obesity, and impaired pancreatic islet insulin secretion. Humans bearing the homologous mutation manifest a congruent phenotype. Our studies highlight that long-term AMPK activation throughout all tissues can have adverse metabolic consequences, with implications for pharmacological strategies seeking to chronically activate AMPK systemically to treat metabolic disease.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Células Secretoras de Insulina/enzimología , Células Secretoras de Insulina/patología , Obesidad/enzimología , Adiposidad/genética , Adulto , Envejecimiento/patología , Proteína Relacionada con Agouti/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Metabolismo Energético/genética , Activación Enzimática , Conducta Alimentaria , Femenino , Heterocigoto , Humanos , Hiperfagia/complicaciones , Hiperfagia/enzimología , Hiperfagia/genética , Hiperfagia/patología , Hipotálamo/metabolismo , Insulina/metabolismo , Masculino , Ratones , Mitocondrias/metabolismo , Mutación/genética , Neuronas/metabolismo , Obesidad/sangre , Obesidad/complicaciones , Obesidad/patología , Fosforilación Oxidativa , Receptores de Ghrelina/metabolismo , Ribosomas/metabolismo , Transducción de Señal/genética , Transcriptoma/genética , Regulación hacia Arriba/genética
9.
J Cell Sci ; 129(9): 1855-65, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-26985063

RESUMEN

Sustained endoplasmic reticulum (ER) stress disrupts normal cellular homeostasis and leads to the development of many types of human diseases, including metabolic disorders. TAK1 (also known as MAP3K7) is a member of the mitogen-activated protein kinase kinase kinase (MAP3K) family and is activated by a diverse set of inflammatory stimuli. Here, we demonstrate that TAK1 regulates ER stress and metabolic signaling through modulation of lipid biogenesis. We found that deletion of Tak1 increased ER volume and facilitated ER-stress tolerance in cultured cells, which was mediated by upregulation of sterol-regulatory-element-binding protein (SREBP)-dependent lipogenesis. In the in vivo setting, central nervous system (CNS)-specific Tak1 deletion upregulated SREBP-target lipogenic genes and blocked ER stress in the hypothalamus. Furthermore, CNS-specific Tak1 deletion prevented ER-stress-induced hypothalamic leptin resistance and hyperphagic obesity under a high-fat diet (HFD). Thus, TAK1 is a crucial regulator of ER stress in vivo, which could be a target for alleviation of ER stress and its associated disease conditions.


Asunto(s)
Estrés del Retículo Endoplásmico , Hipotálamo/metabolismo , Leptina/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Animales , Grasas de la Dieta/efectos adversos , Grasas de la Dieta/farmacología , Hiperfagia/inducido químicamente , Hiperfagia/genética , Hiperfagia/metabolismo , Hiperfagia/patología , Hipotálamo/patología , Leptina/genética , Quinasas Quinasa Quinasa PAM/genética , Ratones , Ratones Noqueados , Obesidad/inducido químicamente , Obesidad/genética , Obesidad/metabolismo , Obesidad/patología , Proteínas de Unión a los Elementos Reguladores de Esteroles/genética , Proteínas de Unión a los Elementos Reguladores de Esteroles/metabolismo
10.
Biofactors ; 42(2): 201-11, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26893251

RESUMEN

Cinnamaldehyde, a bioactive component of cinnamon, is increasingly gaining interest for its preventive and therapeutic effects against metabolic complications like type-2 diabetes. This study is an attempt to understand the effect of cinnamaldehyde in high-fat diet (HFD)-associated increase in fasting-induced hyperphagia and related hormone levels, adipose tissue lipolysis and inflammation, and selected cecal microbial count in mice. Cinnamaldehyde, at 40 µM dose, prevented lipid accumulation and altered gene expression toward lipolytic phenotype in 3T3-L1 preadipocyte cell lines. In vivo, cinnamaldehyde coadministration prevented HFD-induced body weight gain, decreased fasting-induced hyperphagia, as well as circulating leptin and leptin/ghrelin ratio. In addition to that, cinnamaldehyde altered serum biochemical parameters related to lipolysis, that is, glycerol and free fatty acid levels. At transcriptional level, cinnamaldehyde increased anorectic gene expression in hypothalamus and lipolytic gene expression in visceral white adipose tissue. Furthermore, cinnamaldehyde also decreased serum IL-1ß and inflammatory gene expression in visceral white adipose tissue. However, cinnamaldehyde did not modulate the population of selected gut microbial (Lactobacillus, Bifidibaceria, and Roseburia) count in cecal content. In conclusion, cinnamaldehyde increased adipose tissue lipolysis, decreased fasting-induced hyperphagia, normalized circulating levels of leptin/ghrelin ratio, and reduced inflammation in HFD-fed mice, which augurs well for its antiobesity role.


Asunto(s)
Acroleína/análogos & derivados , Suplementos Dietéticos , Hiperfagia/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Células 3T3-L1 , Acroleína/administración & dosificación , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/patología , Animales , Dieta Alta en Grasa , Ayuno/efectos adversos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Hiperfagia/metabolismo , Hiperfagia/patología , Inflamación/sangre , Inflamación/genética , Inflamación/patología , Interleucina-1beta/sangre , Metabolismo de los Lípidos/efectos de los fármacos , Lipólisis/efectos de los fármacos , Ratones , Aumento de Peso/efectos de los fármacos
11.
Cell Metab ; 22(1): 175-88, 2015 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-26073495

RESUMEN

Brain-derived neurotrophic factor (BDNF) is a key regulator of energy balance; however, its underlying mechanism remains unknown. By analyzing BDNF-expressing neurons in paraventricular hypothalamus (PVH), we have uncovered neural circuits that control energy balance. The Bdnf gene in the PVH was mostly expressed in previously undefined neurons, and its deletion caused hyperphagia, reduced locomotor activity, impaired thermogenesis, and severe obesity. Hyperphagia and reduced locomotor activity were associated with Bdnf deletion in anterior PVH, whereas BDNF neurons in medial and posterior PVH drive thermogenesis by projecting to spinal cord and forming polysynaptic connections to brown adipose tissues. Furthermore, BDNF expression in the PVH was increased in response to cold exposure, and its ablation caused atrophy of sympathetic preganglionic neurons. Thus, BDNF neurons in anterior PVH control energy intake and locomotor activity, whereas those in medial and posterior PVH promote thermogenesis by releasing BDNF into spinal cord to boost sympathetic outflow.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Metabolismo Energético , Conducta Alimentaria , Hipotálamo/citología , Hipotálamo/fisiología , Neuronas/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Femenino , Eliminación de Gen , Hiperfagia/genética , Hiperfagia/metabolismo , Hiperfagia/patología , Hipotálamo/patología , Locomoción , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/patología , Termogénesis
12.
Obesity (Silver Spring) ; 22(6): 1477-84, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24639082

RESUMEN

OBJECTIVE: Androgen excess in women is associated with visceral adiposity. However, little is known on the mechanism through which androgen promotes visceral fat accumulation. METHODS: To address this issue, female mice to chronic androgen excess using 5α-dihydrotestosterone (DHT) and studied the regulation of energy homeostasis was exposed. RESULTS: DHT induced a leptin failure to decrease body weight associated with visceral adiposity but without alterations in leptin anorectic action. This paralleled leptin's failure to upregulate brown adipose tissue expression of uncoupling protein-1, associated with decreased energy expenditure (EE). DHT decreased hypothalamic proopiomelanocortin (pomc) mRNA expression and increased POMC intensity in neuronal bodies of the arcuate nucleus while simultaneously decreasing the intensity of POMC projections to the dorsomedial hypothalamus (DMH). This was associated with a failure of the melanocortin 4 receptor agonist melanotan-II to suppress body weight. CONCLUSION: Taken together, these data indicate that androgen excess promotes visceral adiposity with reduced POMC neuronal innervation in the DMH, reduced EE but without hyperphagia.


Asunto(s)
Adiposidad/fisiología , Andrógenos/administración & dosificación , Andrógenos/sangre , Dihidrotestosterona/administración & dosificación , Dihidrotestosterona/sangre , Grasa Intraabdominal/efectos de los fármacos , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/metabolismo , Andrógenos/efectos adversos , Animales , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Núcleo Arqueado del Hipotálamo/metabolismo , Composición Corporal , Peso Corporal , Metabolismo Energético , Femenino , Hiperfagia/patología , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Grasa Intraabdominal/metabolismo , Canales Iónicos/genética , Canales Iónicos/metabolismo , Leptina/sangre , Ratones , Ratones Endogámicos C57BL , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Obesidad/metabolismo , Péptidos Cíclicos/metabolismo , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor de Melanocortina Tipo 4/genética , Receptor de Melanocortina Tipo 4/metabolismo , Proteína Desacopladora 1 , Regulación hacia Arriba , alfa-MSH/análogos & derivados , alfa-MSH/metabolismo
13.
Obesity (Silver Spring) ; 21(6): 1086-92, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23526718

RESUMEN

We sought to determine a role for NPY overexpression in the dorsomedial hypothalamus (DMH) in obesity etiology using the rat model of adeno-associated virus (AAV)-mediated expression of NPY (AAVNPY) in the DMH. Rats received bilateral DMH injections of AAVNPY or control vector and were fed on regular chow. Five-week postviral injection, half the rats from each group were switched to access to a high-fat diet for another 11 weeks. We examined variables including body weight, food intake, energy efficiency, meal patterns, glucose tolerance, fat mass, plasma insulin, plasma leptin, and hypothalamic gene expression. Rats with DMH NPY overexpression had increased food intake and body weight and lowered metabolic efficiency. The hyperphagia was mediated through increased meal size during the dark. Although these rats had normal blood glucose, their plasma insulin levels were increased in both basal and glucose challenge conditions. While high-fat diet induced hyperphagia, obesity, and hyperinsulinemia, these effects were amplified in rats with DMH NPY overexpression. Arcuate Npy, agouti-related protein and proopiomelanocortin expression was appropriately regulated in response to positive energy balance. These results indicate that DMH NPY overexpression can cause hyperphagia and obesity and DMH NPY may have actions in glucose homeostasis.


Asunto(s)
Hiperfagia/genética , Hipotálamo/metabolismo , Neuropéptido Y/genética , Obesidad/genética , Proteína Relacionada con Agouti/genética , Proteína Relacionada con Agouti/metabolismo , Animales , Peso Corporal , Dependovirus/genética , Dependovirus/metabolismo , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Metabolismo Energético , Regulación de la Expresión Génica , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Homeostasis , Hiperfagia/patología , Leptina/sangre , Masculino , Neuropéptido Y/metabolismo , Obesidad/patología , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo , Ratas , Ratas Sprague-Dawley
14.
Nat Commun ; 4: 1526, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23443554

RESUMEN

Brain-derived neurotrophic factor (BDNF) suppresses food intake by acting on neurons in the hypothalamus. Here we show that BDNF-producing haematopoietic cells control appetite and energy balance by migrating to the hypothalamic paraventricular nucleus. These haematopoietic-derived paraventricular nucleus cells produce microglial markers and make direct contacts with neurons in response to feeding status. Mice with congenital BDNF deficiency, specifically in haematopoietic cells, develop hyperphagia, obesity and insulin resistance. These abnormalities are ameliorated by bone marrow transplantation with wild-type bone marrow cells. Furthermore, when injected into the third ventricle, wild-type bone marrow mononuclear cells home to the paraventricular nucleus and reverse the hyperphagia of BDNF-deficient mice. Our results suggest a novel mechanism of feeding control based on the production of BDNF by haematopoietic cells and highlight a potential new therapeutic route for the treatment of obesity.


Asunto(s)
Apetito , Movimiento Celular , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Hipotálamo/metabolismo , Animales , Apetito/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Trasplante de Médula Ósea , Factor Neurotrófico Derivado del Encéfalo/administración & dosificación , Factor Neurotrófico Derivado del Encéfalo/deficiencia , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/farmacología , Movimiento Celular/efectos de los fármacos , Conducta de Ingestión de Líquido/efectos de los fármacos , Ayuno/metabolismo , Conducta Alimentaria/efectos de los fármacos , Eliminación de Gen , Células Madre Hematopoyéticas/efectos de los fármacos , Hiperfagia/complicaciones , Hiperfagia/patología , Hiperfagia/fisiopatología , Hipotálamo/efectos de los fármacos , Hipotálamo/patología , Hipotálamo/ultraestructura , Inyecciones Intraventriculares , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/efectos de los fármacos , Microglía/metabolismo , Microglía/patología , Obesidad/complicaciones , Obesidad/patología , Obesidad/fisiopatología , Especificidad de Órganos/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/metabolismo , Núcleo Hipotalámico Paraventricular/patología , Núcleo Hipotalámico Paraventricular/ultraestructura
15.
J Comp Neurol ; 521(8): 1891-914, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23172177

RESUMEN

The dorsomedial hypothalamus (DMH) has long been implicated in feeding behavior and thermogenesis. The DMH contains orexigenic neuropeptide Y (NPY) neurons, but the role of these neurons in the control of energy homeostasis is not well understood. NPY expression in the DMH is low under normal conditions in adult rodents but is significantly increased during chronic hyperphagic conditions such as lactation and diet-induced obesity (DIO). To understand better the role of DMH-NPY neurons, we characterized the efferent projections of DMH-NPY neurons using the anterograde tracer biotinylated dextran amine (BDA) in lactating rats and DIO mice. In both models, BDA- and NPY-colabeled fibers were limited mainly to the hypothalamus, including the paraventricular nucleus of the hypothalamus (PVH), lateral hypothalamus/perifornical area (LH/PFA), and anteroventral periventricular nucleus (AVPV). Specifically in lactating rats, BDA-and NPY-colabeled axonal swellings were in close apposition to cocaine- and amphetamine-regulated transcript (CART)-expressing neurons in the PVH and AVPV. Although the DMH neurons project to the rostral raphe pallidus (rRPa), these projections did not contain NPY immunoreactivity in either the lactating rat or the DIO mouse. Instead, the majority of BDA-labeled fibers in the rRPa were orexin positive. Furthermore, DMH-NPY projections were not observed within the nucleus of the solitary tract (NTS), another brainstem site critical for the regulation of sympathetic outflow. The present data suggest that NPY expression in the DMH during chronic hyperphagic conditions plays important roles in feeding behavior and thermogenesis by modulating neuronal functions within the hypothalamus, but not in the brainstem.


Asunto(s)
Vías Eferentes/metabolismo , Hiperfagia/patología , Hipotálamo/citología , Neuronas/metabolismo , Neuropéptido Y/metabolismo , Obesidad/patología , Factores de Edad , Animales , Animales Recién Nacidos , Biotina/análogos & derivados , Enfermedad Crónica , Dextranos , Modelos Animales de Enfermedad , Vías Eferentes/fisiología , Femenino , Hormona Liberadora de Gonadotropina/metabolismo , Hormonas Hipotalámicas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ácido Láctico/metabolismo , Masculino , Melaninas/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/metabolismo , Neuropéptidos/metabolismo , Obesidad/etiología , Orexinas , Fragmentos de Péptidos/metabolismo , Hormonas Hipofisarias/metabolismo , Embarazo , Ratas , Ratas Wistar , Triptófano Hidroxilasa/metabolismo
16.
J Clin Invest ; 122(11): 4203-12, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23093774

RESUMEN

Obesity is a chronic metabolic disorder affecting half a billion people worldwide. Major difficulties in managing obesity are the cessation of continued weight loss in patients after an initial period of responsiveness and rebound to pretreatment weight. It is conceivable that chronic weight gain unrelated to physiological needs induces an allostatic regulatory state that defends a supranormal adipose mass despite its maladaptive consequences. To challenge this hypothesis, we generated a reversible genetic mouse model of early-onset hyperphagia and severe obesity by selectively blocking the expression of the proopiomelanocortin gene (Pomc) in hypothalamic neurons. Eutopic reactivation of central POMC transmission at different stages of overweight progression normalized or greatly reduced food intake in these obesity-programmed mice. Hypothalamic Pomc rescue also attenuated comorbidities such as hyperglycemia, hyperinsulinemia, and hepatic steatosis and normalized locomotor activity. However, effectiveness of treatment to normalize body weight and adiposity declined progressively as the level of obesity at the time of Pomc induction increased. Thus, our study using a novel reversible monogenic obesity model reveals the critical importance of early intervention for the prevention of subsequent allostatic overload that auto-perpetuates obesity.


Asunto(s)
Tejido Adiposo/fisiopatología , Adiposidad , Ingestión de Alimentos , Hipotálamo/fisiopatología , Obesidad/prevención & control , Obesidad/fisiopatología , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Modelos Animales de Enfermedad , Hiperfagia/genética , Hiperfagia/metabolismo , Hiperfagia/patología , Hiperfagia/fisiopatología , Hiperfagia/prevención & control , Hipotálamo/metabolismo , Hipotálamo/patología , Ratones , Ratones Noqueados , Neuronas/metabolismo , Neuronas/patología , Obesidad/genética , Obesidad/metabolismo , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo
17.
PLoS One ; 7(7): e40764, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22815809

RESUMEN

G-protein coupled receptor 26 (GPR26) is a brain-specific orphan GPCR with high expression in the brain region that controls satiety. Depletion of GPR26 has been shown to increase fat storage in C. elegans, whereas GPR26 deficiency in the hypothalamus is associated with high genetic susceptibility to the onset of obesity in mice. However, the metabolic function of GPR26 in mammals remains elusive. Herein, we investigated a role of GPR26 in regulating energy homeostasis by generating mice with targeted deletion of the GPR26 gene. We show that GPR26 deficiency causes hyperphagia and hypometabolism, leading to early onset of diet-induced obesity. Accordingly, GPR26 deficiency also caused metabolic complications commonly associated with obesity, including glucose intolerance, hyperinsulinemia, and dyslipidemia. Moreover, consistent with hyperphagia in GPR26 null mice, GPR26 deficiency significantly increased hypothalamic activity of AMPK, a key signaling event that stimulates appetite. In further support of a regulatory role of GPR26 in satiety, GPR26 knockout mice also demonstrate hypersensitivity to treatment of rimonabant, an endocannabinoid receptor-1 antagonist commonly used to treat obesity by suppressing appetite in humans. Together, these findings identified a key role of GPR26 as a central regulator of energy homeostasis though modulation of hypothalamic AMPK activation.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Adiposidad/genética , Silenciador del Gen , Marcación de Gen , Hiperfagia/patología , Hipotálamo/patología , Receptores Acoplados a Proteínas G/genética , Adiposidad/efectos de los fármacos , Animales , Dieta , Dislipidemias/complicaciones , Dislipidemias/patología , Metabolismo Energético/genética , Activación Enzimática/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Intolerancia a la Glucosa/complicaciones , Intolerancia a la Glucosa/genética , Hiperinsulinismo/complicaciones , Hiperinsulinismo/patología , Hiperfagia/complicaciones , Hiperfagia/genética , Hipotálamo/efectos de los fármacos , Hipotálamo/enzimología , Ratones , Ratones Endogámicos C57BL , Obesidad/complicaciones , Obesidad/patología , Fosforilación/efectos de los fármacos , Piperidinas/farmacología , Pirazoles/farmacología , Receptor Cannabinoide CB1/antagonistas & inhibidores , Receptor Cannabinoide CB1/metabolismo , Receptores Acoplados a Proteínas G/deficiencia , Rimonabant , Pérdida de Peso/efectos de los fármacos
18.
PLoS One ; 7(4): e36453, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22558467

RESUMEN

Single-minded 1 (Sim1) is a transcription factor necessary for development of the paraventricular nucleus of the hypothalamus (PVH). This nucleus is a critical regulator of appetite, energy expenditure and body weight. Previously we showed that Sim1(+/-) mice and conditional postnatal Sim1(-/-) mice exhibit hyperphagia, obesity, increased linear growth and susceptibility to diet-induced obesity, but no decrease in energy expenditure. Bilateral ablation of the PVH causes obesity due to hyperphagia and reduced energy expenditure. It remains unknown whether Sim1 neurons regulate energy expenditure. In this study, Sim1cre mice were bred to homozygous inducible diphtheria toxin receptor (iDTR) mice to generate mice expressing the simian DTR in Sim1 cells. In these mice, Sim1 neuron ablation was performed by intracerebroventricular (ICV) injection of diphtheria toxin. Compared to controls, mice with Sim1 neuron ablation became obese (with increased fat mass) on a chow diet due to increased food intake and reduced energy expenditure. In post-injection mice, we observed a strong inverse correlation between the degree of obesity and hypothalamic Sim1 expression. The reduction in baseline energy expenditure observed in these mice was accompanied by a reduction in activity. This reduction in activity did not fully account for the reduced energy expenditure as these mice exhibited decreased resting energy expenditure, decreased body temperature, decreased brown adipose tissue temperature, and decreased UCP1 expression suggesting an impairment of thermogenesis. In injected mice, hypothalamic gene expression of Sim1, oxytocin (OXT) and thyrotropin releasing hormone (TRH) was reduced by about 50%. These results demonstrate that Sim1 neurons in adult mice regulate both food intake and energy expenditure. Based on the body of work in the field, feeding regulation by Sim1 neurons likely occurs in both the PVH and medial amygdala, in contrast to energy expenditure regulation by Sim1 neurons, which likely is localized to the PVH.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Metabolismo Energético , Hiperfagia/metabolismo , Hiperfagia/patología , Neuronas/metabolismo , Obesidad/metabolismo , Obesidad/patología , Proteínas Represoras/metabolismo , Animales , Peso Corporal/efectos de los fármacos , Toxina Diftérica/toxicidad , Metabolismo Energético/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Factor de Crecimiento Similar a EGF de Unión a Heparina , Hiperfagia/inducido químicamente , Hiperfagia/genética , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones , Ratones Transgénicos , Neuronas/efectos de los fármacos , Neuropéptidos/metabolismo , Obesidad/inducido químicamente , Obesidad/genética , Termogénesis/efectos de los fármacos
19.
Mol Cell Biochem ; 368(1-2): 37-45, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22638648

RESUMEN

The metabolic syndrome is recognized as a cluster of disturbances associated with obesity, type 2 diabetes and hypertension. Over the past two decades, the number of people with the metabolic syndrome has increased at an alarming rate. This increase is associated with the global epidemic of both obesity and diabetes. Cardiovascular mortality is increased among diabetics and obesity-related insulin-resistant patients, and obesity is currently recognized as independent risk factor for cardiovascular disease. We aimed to establish the effects of a short period of an altered diet on the heart using a rat model of hyperphagia-induced obesity (diet supplemented with sucrose and condensed milk for 8 weeks = DIO) compared to age-matched controls. Isolated, perfused hearts were subjected to global or regional ischaemia/reperfusion. Function on reperfusion was recorded and infarct size determined. A plasma lipid profile was established via HPLC-based methods and proteins involved in metabolic signalling determined either by western blotting or RT-PCR. 8 weeks of diet resulted in whole-body but not myocardial insulin resistance, increased plasma triglyceride and phospholipid levels as well as increased lipid peroxidation. Despite the similar baseline function, hearts from DIO animals showed significantly poorer postischaemic recovery than controls (41.9 % RPP recovery vs 57.9 %, P < 0.05, n = 7-11/group) but surprisingly, smaller infarct size (24.95 ± 1.97 vs 47.26 ± 4.05 % of the area at risk, P < 0.005, n = 8/group). Basal phosphorylation of PKB/Akt was elevated but IRS-1 and SERCA-2 expression severely downregulated. In conclusion, after only 8 weeks of a slight change in diet, the rat heart shows signs of metabolic remodelling. Some of these changes may be protective but others may be detrimental and eventually lead to maladaptation.


Asunto(s)
Dieta/efectos adversos , Resistencia a la Insulina , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Obesidad/metabolismo , Obesidad/fisiopatología , Animales , Hiperfagia/inducido químicamente , Hiperfagia/metabolismo , Hiperfagia/patología , Hiperfagia/fisiopatología , Proteínas Sustrato del Receptor de Insulina/metabolismo , Proteínas Musculares/metabolismo , Infarto del Miocardio/inducido químicamente , Infarto del Miocardio/mortalidad , Miocardio/metabolismo , Miocardio/patología , Obesidad/inducido químicamente , Obesidad/patología , Fosfolípidos/sangre , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Wistar , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Triglicéridos/sangre
20.
Diabetologia ; 55(3): 763-72, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22159884

RESUMEN

AIMS/HYPOTHESIS: The carcino-embryonic antigen-related cell adhesion molecule (CEACAM)2 is produced in many feeding control centres in the brain, but not in peripheral insulin-targeted tissues. Global Ceacam2 null mutation causes insulin resistance and obesity resulting from hyperphagia and hypometabolism in female Ceacam2 homozygous null mutant mice (Cc2 [also known as Ceacam2](-/-)) mice. Because male mice are not obese, the current study examined their metabolic phenotype. METHODS: The phenotype of male Cc2(-/-) mice was characterised by body fat composition, indirect calorimetry, hyperinsulinaemic-euglycaemic clamp analysis and direct recording of sympathetic nerve activity. RESULTS: Despite hyperphagia, total fat mass was reduced, owing to the hypermetabolic state in male Cc2(-/-) mice. In contrast to females, male mice also exhibited insulin sensitivity with elevated ß-oxidation in skeletal muscle, which is likely to offset the effects of increased food intake. Males and females had increased brown adipogenesis. However, only males had increased activation of sympathetic tone regulation of adipose tissue and increased spontaneous activity. The mechanisms underlying sexual dimorphism in energy balance with the loss of Ceacam2 remain unknown. CONCLUSIONS/INTERPRETATION: These studies identified a novel role for CEACAM2 in the regulation of metabolic rate and insulin sensitivity via effects on brown adipogenesis, sympathetic nervous outflow to brown adipose tissue, spontaneous activity and energy expenditure in skeletal muscle.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Metabolismo Energético , Glicoproteínas/metabolismo , Hiperfagia/metabolismo , Resistencia a la Insulina , Músculo Esquelético/metabolismo , Adipogénesis , Tejido Adiposo Pardo/inervación , Tejido Adiposo Pardo/patología , Adiposidad , Animales , Moléculas de Adhesión Celular , Femenino , Glicoproteínas/genética , Hiperfagia/genética , Hiperfagia/patología , Hiperfagia/fisiopatología , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , ARN Mensajero/metabolismo , Caracteres Sexuales , Sistema Nervioso Simpático/fisiopatología , Transmisión Sináptica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA