Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Neurochem Int ; 140: 104847, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32927026

RESUMEN

Fragile X mental retardation protein (FMRP), strongly associated with fragile X syndrome, plays important roles by regulating gene expression via interacting with other RNA binding proteins in the brain. However, the role of FMRP in hypothalamus, a central part responsible for metabolic control, is poorly known. Our study shows that FMRP is primarily located in the hypothalamic arcuate nucleus (ARC). Using proteomic analysis, we identified 56 up-regulated and 22 down-regulated proteins in the hypothalamus of Map1b KO mice, with microtubule-associated protein 1 B (MAP1B) being the most outstanding increased protein (more than 10 folds). Immunofluorescent assays showed that MAP1B significantly increased in the Map1b-KO ARC, in which the number of agouti-related peptide (AgRP)-staining neurons significantly reduced, but not altered for pro-opiomelanocortin (POMC) neurons. We further showed an age-dependent reduces in food intake and body weight of the KO mice, along with the decreases of MAP1B and AgRP at the same time points. In hypothalamic GT1-7 cells, the AgRP expression decreased upon knockdown of FMRP or overexpression of MAP1B, and increased in response to overexpression of FMRP or knockdown of MAP1B. Co-knockdown or co-overexpression of FMRP and MAP1B led to a reverse expression of AgRP compared to overexpression of knockdown of FMRP alone, demonstrating that MAP1B is essential for the regulatory effect of FMRP on AgRP expression. Taken together, these data suggest that FMRP-deficiency-induced increase of hypothalamic MAP1B and decrease of AgRP might be associated with reduces in food intake and body weight.


Asunto(s)
Proteína Relacionada con Agouti/biosíntesis , Peso Corporal/fisiología , Ingestión de Alimentos/fisiología , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Hipotálamo/metabolismo , Proteínas Asociadas a Microtúbulos/biosíntesis , Proteína Relacionada con Agouti/antagonistas & inhibidores , Proteína Relacionada con Agouti/genética , Animales , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Expresión Génica , Masculino , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/genética , Regulación hacia Arriba/fisiología
2.
Physiol Behav ; 204: 112-120, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30633898

RESUMEN

Corticosterone (CORT) is a powerful regulator of energy metabolism, and chronically high CORT levels cause obesity and diabetes in mice. It is reported that a chronically high CORT level changes food preference, increasing the intake of comfort foods such as fatty foods. Previously, we demonstrated that unlike a high fat diet, voluntary ingestion of 100% pure corn oil increased energy expenditure and thermogenesis through the activation of the interscapular brown adipose tissue (IBAT). In the present study, we investigated whether chronically high CORT affected corn oil intake, energy expenditure, and body weight gain. We delivered CORT to mice via water bottles and placed corn oil in a separate drinking bottle in the home cage. Voluntary corn oil ingestion with CORT induced significant body weight gain, while corn oil ingestion or CORT alone had a modest effect. CORT increased corn oil intake without reducing chow intake, which further increased the total daily caloric intake. CORT suppressed mRNA related to thermogenesis in IBAT. In the hypothalamus, CORT upregulated mRNA expression of the orexigenic neuropeptide, agouti-related protein. These data suggest that chronically high CORT might increase the desire to consume dietary fat, suppressing BAT function, thereby causing obesity.


Asunto(s)
Aceite de Maíz/farmacología , Corticosterona/farmacología , Aumento de Peso/efectos de los fármacos , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/metabolismo , Proteína Relacionada con Agouti/biosíntesis , Proteína Relacionada con Agouti/genética , Animales , Análisis de los Gases de la Sangre , Corticosterona/sangre , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Energía/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Consumo de Oxígeno/efectos de los fármacos , Termogénesis/efectos de los fármacos
3.
Physiol Behav ; 190: 61-70, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29031552

RESUMEN

Arcuate hypothalamus-derived agouti-related protein (AgRP) and neuropeptide Y (NPY) are critical for maintaining energy homeostasis. Fasting markedly upregulates AgRP/NPY expression and circulating ghrelin, and exogenous ghrelin treatment robustly increases acute food foraging and food intake, and chronic food hoarding behaviors in Siberian hamsters. We previously demonstrated that 3rd ventricular AgRP injection robustly stimulates acute and chronic food hoarding, largely independent of food foraging and intake. By contrast, 3rd ventricular NPY injection increases food foraging, food intake, and food hoarding, but this effect is transient and gone by 24h post-injection. Because of this discrepancy in AgRP/NPY-induced ingestive behaviors, we tested whether selective knockdown of AgRP blocks fasting and ghrelin-induced increases in food hoarding. AgRP gene knockdown by a novel DICER small interfering RNA (AgRP-DsiRNA) blocked food-deprivation induced increases in AgRP expression, but had no effect on NPY expression. AgRP-DsiRNA attenuated acute (1day), and significantly decreased chronic (4-6days), food deprivation-induced increases in food hoarding. In addition, AgRP-DsiRNA treatment blocked exogenous ghrelin-induced increases in food hoarding through day 3, but had no effect on basal food foraging, food intake, or food hoarding prior to ghrelin treatment. Lastly, chronic AgRP knockdown had no effect on body mass, fat mass, or lean mass in either food deprived or ad libitum fed hamsters. These data collectively suggest that the prolonged increase in food hoarding behavior following energetic challenges, and food deprivation especially, is primarily regulated by downstream AgRP signaling.


Asunto(s)
Proteína Relacionada con Agouti/deficiencia , Conducta Alimentaria/fisiología , Proteína Relacionada con Agouti/biosíntesis , Animales , Conducta Apetitiva/efectos de los fármacos , Conducta Apetitiva/fisiología , Composición Corporal/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Cricetinae , Ingestión de Alimentos/efectos de los fármacos , Ayuno , Conducta Alimentaria/efectos de los fármacos , Ghrelina/antagonistas & inhibidores , Ghrelina/farmacología , Masculino , Neuropéptido Y/biosíntesis , Phodopus , ARN Interferente Pequeño/farmacología
4.
J Clin Invest ; 127(7): 2868-2880, 2017 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-28628036

RESUMEN

Obesity increases sympathetic nerve activity (SNA) via activation of proopiomelanocortin neurons in the arcuate nucleus (ArcN), and this action requires simultaneous withdrawal of tonic neuropeptide Y (NPY) sympathoinhibition. However, the sites and neurocircuitry by which NPY decreases SNA are unclear. Here, using designer receptors exclusively activated by designer drugs (DREADDs) to selectively activate or inhibit ArcN NPY neurons expressing agouti-related peptide (AgRP) in mice, we have demonstrated that this neuronal population tonically suppresses splanchnic SNA (SSNA), arterial pressure, and heart rate via projections to the paraventricular nucleus (PVN) and dorsomedial hypothalamus (DMH). First, we found that ArcN NPY/AgRP fibers closely appose PVN and DMH presympathetic neurons. Second, nanoinjections of NPY or an NPY receptor Y1 (NPY1R) antagonist into PVN or DMH decreased or increased SSNA, respectively. Third, blockade of DMH NPY1R reversed the sympathoinhibition elicited by selective, DREADD-mediated activation of ArcN NPY/AgRP neurons. Finally, stimulation of ArcN NPY/AgRP terminal fields in the PVN and DMH decreased SSNA. Considering that chronic obesity decreases ArcN NPY content, we propose that the ArcN NPY neuropathway to the PVN and DMH is pivotal in obesity-induced elevations in SNA.


Asunto(s)
Núcleo Arqueado del Hipotálamo/metabolismo , Presión Sanguínea , Neuropéptido Y/metabolismo , Sistema Nervioso Simpático/metabolismo , Sistema Nervioso Simpático/fisiopatología , Proteína Relacionada con Agouti/biosíntesis , Proteína Relacionada con Agouti/genética , Animales , Núcleo Arqueado del Hipotálamo/patología , Núcleo Arqueado del Hipotálamo/fisiopatología , Enfermedad Crónica , Regulación de la Expresión Génica , Frecuencia Cardíaca , Ratones , Ratones Transgénicos , Neuropéptido Y/genética , Obesidad/genética , Obesidad/metabolismo , Obesidad/patología , Obesidad/fisiopatología , Núcleo Hipotalámico Paraventricular/metabolismo , Núcleo Hipotalámico Paraventricular/patología , Núcleo Hipotalámico Paraventricular/fisiopatología , Receptores de Neuropéptido Y/genética , Receptores de Neuropéptido Y/metabolismo , Sistema Nervioso Simpático/patología
5.
Biochem Biophys Res Commun ; 471(4): 566-71, 2016 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-26879142

RESUMEN

The regulation of food intake is a promising way to combat obesity. It has been implicated that various fatty acids exert different effects on food intake and body weight. However, the underlying mechanism remains poorly understood. The aim of the present study was to investigate the effects of linoleic acid (LA) and stearic acid (SA) on agouti-related protein (AgRP) expression and secretion in immortalized mouse hypothalamic N38 cells and to explore the likely underlying mechanisms. Our results demonstrated that LA inhibited, while SA stimulated AgRP expression and secretion of N38 cells in a dose-dependent manner. In addition, LA suppressed the protein expression of toll-like receptor 4 (TLR4), phosphorylation levels of JNK and IKKα/ß, suggesting the inhibition of TLR4-dependent inflammation pathway. However, the above mentioned inhibitory effects of LA were eliminated by TLR4 agonist lipopolysaccharide (LPS). In contrast, SA promoted TLR4 protein expression and activated TLR4-dependent inflammation pathway, with elevated ratio of p-JNK/JNK. While TLR4 siRNA reversed the stimulatory effects of SA on AgRP expression and TLR4-dependent inflammation. Moreover, we found that TLR4 was also involved in LA-enhanced and SA-impaired leptin/insulin signal pathways in N38 cells. In conclusion, our findings indicated that LA elicited inhibitory while SA exerted stimulatory effects on AgRP expression and secretion via TLR4-dependent inflammation and leptin/insulin pathways in N38 cells. These data provided a better understanding of the mechanism underlying fatty acids-regulated food intake and suggested the potential role of long-chain unsaturated fatty acids such as LA in reducing food intake and treating obesity.


Asunto(s)
Proteína Relacionada con Agouti , Ingestión de Alimentos/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Ácido Linoleico/farmacología , Ácidos Esteáricos/farmacología , Receptor Toll-Like 4/metabolismo , Proteína Relacionada con Agouti/agonistas , Proteína Relacionada con Agouti/antagonistas & inhibidores , Proteína Relacionada con Agouti/biosíntesis , Animales , Hipotálamo/citología , Hipotálamo/metabolismo , Quinasa I-kappa B/metabolismo , Inflamación/metabolismo , Leptina/metabolismo , Lipopolisacáridos/farmacología , Ratones , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Fosforilación , ARN Interferente Pequeño/genética , Transducción de Señal , Receptor Toll-Like 4/genética
6.
Psychoneuroendocrinology ; 67: 171-81, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26907996

RESUMEN

We hypothesize that anorexia nervosa (AN) poses a physiological stress. Therefore, the way an individual copes with stress may affect AN vulnerability. Since prenatal stress (PNS) exposure alters stress responsivity in offspring this may increase their risk of developing AN. We tested this hypothesis using the activity based anorexia (ABA) rat model in control and PNS rats that were characterized by either proactive or passive stress-coping behavior. We found that PNS passively coping rats ate less and lost more weight during the ABA paradigm. Exposure to ABA resulted in higher baseline corticosterone and lower insulin levels in all groups. However, leptin levels were only decreased in rats with a proactive stress-coping style. Similarly, ghrelin levels were increased only in proactively coping ABA rats. Neuropeptide Y (Npy) expression was increased and proopiomelanocortin (Pomc) expression was decreased in all rats exposed to ABA. In contrast, agouti-related peptide (Agrp) and orexin (Hctr) expression were increased in all but the PNS passively coping ABA rats. Furthermore, DNA methylation of the orexin gene was increased after ABA in proactive coping rats and not in passive coping rats. Overall our study suggests that passive PNS rats have innate impairments in leptin and ghrelin in responses to starvation combined with prenatal stress associated impairments in Agrp and orexin expression in response to starvation. These impairments may underlie decreased food intake and associated heightened body weight loss during ABA in the passively coping PNS rats.


Asunto(s)
Adaptación Psicológica/fisiología , Proteína Relacionada con Agouti/biosíntesis , Anorexia/metabolismo , Anorexia/fisiopatología , Orexinas/biosíntesis , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Animales , Peso Corporal/fisiología , Metilación de ADN , Ingestión de Líquidos/fisiología , Ingestión de Alimentos/fisiología , Femenino , Ghrelina/biosíntesis , Leptina/biosíntesis , Masculino , Actividad Motora/fisiología , Neuropéptido Y/biosíntesis , Embarazo , Proopiomelanocortina/biosíntesis , Ratas , Regulación hacia Arriba
7.
Exp Dermatol ; 25(2): 120-3, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26513428

RESUMEN

We previously found that ultraviolet B (UVB) could stimulate the paraventricular nucleus (PVN) with activation the systemic hypothalamic-pituitary- adrenal (HPA) axis. To investigate whether UVB can also stimulate other hypothalamic nuclei, we tested its effect on the proopiomelanocortin (POMC) related signalling system in the arcuate nucleus (ARC) of female C57BL/6 and FVB albino mice. The shaved back skin of the mice was irradiated with either 100 or 400 mJ/cm2 of UVB. After 1, 3, 6 and 12 h, blood and hypothalamus were collected and processed for gene and protein expression, and measurement of α-MSH and ß-endorphin (ß-END) levels. An in situ immunohistochemical examination was performed for melanocortin receptor 4 (MC4R) and POMC-derived α-MSH. The expression of Pomc and MC4R mRNAs was stimulated, whereas that of AgRP was inhibited after exposure to UVB. It was accompanied by an increased number of both α-MSH- and MC4R-immunoreactive neurons in the ARC, and by increased levels of α-MSH and ß-END (both found in the hypothalamus and plasma). This surprising discovery of UVB stimulating the POMC system in the ARC, accompanied by the increased plasma levels of α-MSH and ß-END, paves the way for exciting areas of research on the communication between the skin and the brain, as well as is suggesting a new role for UVB in regulation of body metabolism.


Asunto(s)
Núcleo Arqueado del Hipotálamo/metabolismo , Proopiomelanocortina/biosíntesis , Piel/efectos de la radiación , Rayos Ultravioleta , Proteína Relacionada con Agouti/biosíntesis , Proteína Relacionada con Agouti/genética , Animales , Metabolismo Basal , Femenino , Regulación de la Expresión Génica/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Proopiomelanocortina/genética , ARN Mensajero/biosíntesis , Receptor de Melanocortina Tipo 4/biosíntesis , Receptor de Melanocortina Tipo 4/genética , Transducción de Señal/fisiología , Organismos Libres de Patógenos Específicos , alfa-MSH/biosíntesis , alfa-MSH/sangre , alfa-MSH/genética , betaendorfina/biosíntesis , betaendorfina/sangre , betaendorfina/genética
8.
J Neurosci ; 35(29): 10440-50, 2015 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-26203139

RESUMEN

The hypothalamus is critical for feeding and body weight regulation. Prevailing studies focus on hypothalamic neurons that are defined by selectively expressing transcription factors or neuropeptides including those expressing proopiomelanocortin (POMC) and agouti-related peptides (AgRP). The Cre expression driven by the pancreas-duodenum homeobox 1 promoter is abundant in several hypothalamic nuclei but not in AgRP or POMC neurons. Using this line, we generated mice with disruption of GABA release from a major subset of non-POMC, non-AgRP GABAergic neurons in the hypothalamus. These mice exhibited a reduction in postweaning feeding and growth, and disrupted hyperphagic responses to NPY. Disruption of GABA release severely diminished GABAergic input to the paraventricular hypothalamic nucleus (PVH). Furthermore, disruption of GABA-A receptor function in the PVH also reduced postweaning feeding and blunted NPY-induced hyperphagia. Given the limited knowledge on postweaning feeding, our results are significant in identifying GABA release from a major subset of less appreciated hypothalamic neurons as a key mediator for postweaning feeding and NPY hyperphagia, and the PVH as one major downstream site that contributes significantly to the GABA action. Significance statement: Prevalent studies on feeding in the hypothalamus focus on well characterized, selective groups neurons [e.g., proopiomelanocortin (POMC) and agouti-related peptide (AgRP) neurons], and as a result, the role of the majority of other hypothalamic neurons is largely neglected. Here, we demonstrated an important role for GABAergic projections from non-POMC non-AgRP neurons to the paraventricular hypothalamic nucleus in promoting postweaning (mainly nocturnal) feeding and mediating NPY-induced hyperphagia. Thus, these results signify an importance to study those yet to be defined hypothalamic neurons in the regulation of energy balance and reveal a neural basis for postweaning (nocturnal) feeding and NPY-mediated hyperphagia.


Asunto(s)
Conducta Alimentaria/fisiología , Neuronas GABAérgicas/fisiología , Hiperfagia/fisiopatología , Núcleo Hipotalámico Paraventricular/fisiología , Proteína Relacionada con Agouti/biosíntesis , Animales , Hipotálamo/citología , Hipotálamo/fisiología , Hibridación in Situ , Ratones , Ratones Mutantes , Neuropéptido Y/metabolismo , Técnicas de Cultivo de Órganos , Núcleo Hipotalámico Paraventricular/citología , Técnicas de Placa-Clamp , Proopiomelanocortina/biosíntesis
9.
J Neurosci ; 35(11): 4571-81, 2015 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-25788674

RESUMEN

Peroxisome proliferator-activated receptor γ (PPARγ) is clinically targeted for type II diabetes treatment; however, rosiglitazone (ROSI), a PPARγ agonist, increases food intake and body/fat mass as side-effects. Mechanisms for these effects and the role of PPARγ in feeding are not understood. Therefore, we tested this role in Siberian hamsters, a model of human energy balance, and C57BL/6 mice. We tested the following: (1) how ROSI and/or GW9662 (2-chloro-5-nitro-N-phenylbenzamide; PPARγ antagonist) injected intraperitoneally or into the third ventricle (3V) affected Siberian hamster feeding behaviors; (2) whether food deprivation (FD) co-increases agouti-related protein (AgRP) and PPARγ mRNA expression in Siberian hamsters and mice; (3) whether intraperitoneally administered ROSI increases AgRP and NPY in ad libitum-fed animals; (4) whether intraperitoneally administered PPARγ antagonism blocks FD-induced increases in AgRP and NPY; and finally, (5) whether intraperitoneally administered PPARγ modulation affects plasma ghrelin. Third ventricular and intraperitoneally administered ROSI increased food hoarding and intake for 7 d, an effect attenuated by 3V GW9662, and also prevented (intraperitoneal) FD-induced feeding. FD hamsters and mice increased AgRP within the arcuate hypothalamic nucleus with concomitant increases in PPARγ exclusively within AgRP/NPY neurons. ROSI increased AgRP and NPY similarly to FD, and GW9662 prevented FD-induced increases in AgRP and NPY in both species. Neither ROSI nor GW9662 affected plasma ghrelin. Thus, we demonstrated that PPARγ activation is sufficient to trigger food hoarding/intake, increase AgRP/NPY, and possibly is necessary for FD-induced increases in feeding and AgRP/NPY. These findings provide initial evidence that FD-induced increases in AgRP/NPY may be a direct PPARγ-dependent process that controls ingestive behaviors.


Asunto(s)
Proteína Relacionada con Agouti/biosíntesis , Núcleo Arqueado del Hipotálamo/metabolismo , Conducta Alimentaria/fisiología , Neuropéptido Y/biosíntesis , PPAR gamma/biosíntesis , ARN Mensajero/biosíntesis , Animales , Cricetinae , Conducta Alimentaria/psicología , Ratones , Ratones Endogámicos C57BL , Phodopus
10.
Biochem Biophys Res Commun ; 456(1): 116-21, 2015 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-25446108

RESUMEN

G protein-coupled receptors (GPCRs) represent the largest family of transmembrane signaling proteins, and they are considered major targets of approximately half of all therapeutic agents. Human melanocortin-4 receptor (hMC4R) plays an important role in the control of energy homeostasis, and its mutants are directly related to severe human obesity. Here, we describe optimized protocols for the high-yield expression and purification of hMC4R that will accelerate structural study. Truncations of the N- and C-termini of hMC4R with T4 lysozyme (T4L) insertion increase the solubility as well as stability of the protein. Strikingly, co-expression of human mini-agouti-related protein (mini-AgRP) in Spodoptera frugiperda (Sf9) cells enables excellent stability of hMC4R. The protein yield in the human mini-AgRP co-expression system is increased by about 3-4 times compared to that of hMC4R alone. Data from analytical size exclusion chromatography (aSEC) and thermostability assay show that hMC4R becomes homogeneous and stable with a melting temperature of 58°C in the presence of human mini-AgRP.


Asunto(s)
Proteína Relacionada con Agouti/biosíntesis , Regulación de la Expresión Génica , Receptor de Melanocortina Tipo 4/biosíntesis , Secuencia de Aminoácidos , Animales , Bacteriófago T4/enzimología , Línea Celular , Cromatografía , Cromatografía en Gel , Humanos , Insectos , Ligandos , Datos de Secuencia Molecular , Muramidasa/química , Mutación , Estructura Terciaria de Proteína , Proteínas Recombinantes/biosíntesis , Transducción de Señal , Solubilidad , Temperatura
11.
Am J Physiol Endocrinol Metab ; 306(11): E1284-91, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24735891

RESUMEN

CCK and leptin are anorectic hormones produced in the small intestine and white adipose tissue, respectively. Investigating how these hormones act together as an integrated anorectic signal is important for elucidating the mechanisms by which energy balance is maintained. We found here that coadministration of subthreshold CCK and leptin, which individually have no effect on feeding, dramatically reduced food intake in rats. Phosphorylation of AMP-activated protein kinase (AMPK) in the hypothalamus significantly decreased after coinjection of CCK and leptin. In addition, coadministration of these hormones significantly increased mRNA levels of anorectic cocaine- and amphetamine-regulated transcript (CART) and thyrotropin-releasing hormone (TRH) in the hypothalamus. The interactive effect of CCK and leptin on food intake was abolished by intracerebroventricular preadministration of the AMPK activator AICAR or anti-CART/anti-TRH antibodies. These findings indicate that coinjection of CCK and leptin reduces food intake via reduced AMPK phosphorylation and increased CART/TRH in the hypothalamus. Furthermore, by using midbrain-transected rats, we investigated the role of the neural pathway from the hindbrain to the hypothalamus in the interaction of CCK and leptin to reduce food intake. Food intake reduction induced by coinjection of CCK and leptin was blocked in midbrain-transected rats. Therefore, the neural pathway from hindbrain to hypothalamus plays an important role in transmitting the anorectic signals provided by coinjection of CCK and leptin. Our findings give further insight into the mechanisms of feeding and energy balance.


Asunto(s)
Proteínas Quinasas Activadas por AMP/biosíntesis , Colecistoquinina/farmacología , Ingestión de Alimentos/efectos de los fármacos , Hipotálamo/metabolismo , Leptina/farmacología , Proteínas del Tejido Nervioso/biosíntesis , Hormona Liberadora de Tirotropina/biosíntesis , Proteína Relacionada con Agouti/biosíntesis , Animales , Western Blotting , Hipotálamo/efectos de los fármacos , Hipotálamo/enzimología , Inyecciones Intraventriculares , Masculino , Mesencéfalo/fisiología , Neuropéptido Y/biosíntesis , Fosforilación , ARN Mensajero/biosíntesis , Ratas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Transcripción STAT3/biosíntesis
12.
Mol Cell Endocrinol ; 382(1): 262-270, 2014 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-24145125

RESUMEN

Glucose-sensing neurons play a role in energy homeostasis, yet how orexigenic neurons sense glucose remains unclear. As models of glucose-inhibited (GI) neurons, mHypoE-29/1 and mHypoA-NPY/GFP cells express the essential orexigenic neuropeptide AgRP and glucose sensing machinery. Exposure to increasing concentrations of glucose or the glucose analog 2-deoxyglucose (2-DG) results in a decrease in AgRP mRNA levels. Taste receptor, Tas1R2 mRNA expression was reduced by glucose, whereas 2-DG reduced Tas1R3 mRNA levels. Increasing glucose concentrations elicited a rise in Akt and neuronal nitric oxide synthase (nNOS) phosphorylation, CaMKKß levels, and a reduction of AMP-kinase alpha phosphorylation. Inhibitors of NOS and the cystic fibrosis transmembrane conductance regulator (CFTR) prevented a decrease in AgRP secretion with glucose, suggesting a pivotal role for nNOS and the CFTR in glucose-sensing. These models possess the hallmark characteristics of GI neurons, and can be used to disentangle the mechanisms by which orexigenic neurons sense glucose.


Asunto(s)
Proteína Relacionada con Agouti/biosíntesis , Proteína Relacionada con Agouti/metabolismo , Glucosa/farmacología , Hipotálamo/citología , Hipotálamo/metabolismo , Modelos Biológicos , Adenilato Quinasa/metabolismo , Proteína Relacionada con Agouti/genética , Animales , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Carnitina O-Palmitoiltransferasa/genética , Carnitina O-Palmitoiltransferasa/metabolismo , Línea Celular , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Desoxiglucosa/farmacología , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Gemfibrozilo/farmacología , Proteínas Fluorescentes Verdes/metabolismo , Hipotálamo/efectos de los fármacos , Hipotálamo/embriología , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuropéptido Y , Óxido Nítrico Sintasa de Tipo I/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ácido Pirúvico/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
13.
J Biol Chem ; 289(7): 4055-69, 2014 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-24366864

RESUMEN

CTRP4 is a unique member of the C1q family, possessing two tandem globular C1q domains. Its physiological function is poorly defined. Here, we show that CTRP4 is an evolutionarily conserved, ∼34-kDa secretory protein expressed in the brain. In human, mouse, and zebrafish brain, CTRP4 expression begins early in development and is widespread in the central nervous system. Neurons, but not astrocytes, express and secrete CTRP4, and secreted proteins form higher-order oligomeric complexes. CTRP4 is also produced by peripheral tissues and circulates in blood. Its serum levels are increased in leptin-deficient obese (ob/ob) mice. Functional studies suggest that CTRP4 acts centrally to modulate energy metabolism. Refeeding following an overnight fast induced the expression of CTRP4 in the hypothalamus. Central administration of recombinant protein suppressed food intake and altered the whole-body energy balance in both chow-fed and high-fat diet-fed mice. Suppression of food intake by CTRP4 is correlated with a decreased expression of orexigenic neuropeptide (Npy and Agrp) genes in the hypothalamus. These results establish CTRP4 as a novel nutrient-responsive central regulator of food intake and energy balance.


Asunto(s)
Adipoquinas/metabolismo , Peso Corporal/fisiología , Citocinas/metabolismo , Ingestión de Alimentos/fisiología , Metabolismo Energético/fisiología , Hipotálamo/metabolismo , Pez Cebra/metabolismo , Adipoquinas/genética , Proteína Relacionada con Agouti/biosíntesis , Proteína Relacionada con Agouti/genética , Animales , Citocinas/genética , Humanos , Masculino , Ratones , Neuropéptido Y/biosíntesis , Neuropéptido Y/genética , Estructura Terciaria de Proteína , Ratas , Pez Cebra/genética
14.
Am J Physiol Endocrinol Metab ; 304(9): E909-21, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23443924

RESUMEN

Animal domestication has resulted in changes in growth and size. It has been suggested that this may have involved selection for differences in appetite. Divergent growth between chickens selected for egg laying or meat production is one such example. The neurons expressing AGRP and POMC in the basal hypothalamus are important components of appetite regulation, as are the satiety feedback pathways that carry information from the intestine, including CCK and its receptor CCKAR (CCK1 receptor). Using 16 generations of a cross between a fast and a relatively slow growing strain of chicken has identified a region on chromosome 4 downstream of the CCKAR gene, which is responsible for up to a 19% difference in body weight at 12 wk of age. Animals possessing the high-growth haplotype at the locus have lower expression of mRNA and immunoreactive CCKAR in the brain, intestine, and exocrine organs, which is correlated with increased levels of orexigenic AGRP in the hypothalamus. Animals with the high-growth haplotype are resistant to the anorectic effect of exogenously administered CCK, suggesting that their satiety set point has been altered. Comparison with traditional breeds shows that the high-growth haplotype has been present in the founders of modern meat-type strains and may have been selected early in domestication. This is the first dissection of the physiological consequences of a genetic locus for a quantitative trait that alters appetite and gives us an insight into the domestication of animals. This will allow elucidation of how differences in appetite occur in birds and also mammals.


Asunto(s)
Animales Domésticos , Peso Corporal/genética , Peso Corporal/fisiología , Pollos/genética , Pollos/fisiología , Crecimiento/genética , Crecimiento/fisiología , Receptor de Colecistoquinina A/biosíntesis , Receptor de Colecistoquinina A/fisiología , Respuesta de Saciedad/fisiología , Proteína Relacionada con Agouti/biosíntesis , Proteína Relacionada con Agouti/genética , Alelos , Animales , Química Encefálica/fisiología , Cruzamientos Genéticos , Ingestión de Alimentos/genética , Ingestión de Alimentos/fisiología , Femenino , Genotipo , Inmunohistoquímica , Masculino , Polimorfismo de Nucleótido Simple/genética , ARN/biosíntesis , ARN/aislamiento & purificación , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor de Colecistoquinina A/genética , Distribución Tisular , Transcripción Genética
15.
Neurosignals ; 21(1-2): 28-41, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-22456226

RESUMEN

Activation of mammalian target of rapamycin 1 (mTORC1) by nutrients, insulin and leptin leads to appetite suppression (anorexia). Contrastingly, increased AMP-activated protein kinase (AMPK) activity by ghrelin promotes appetite (orexia). However, the interplay between these mechanisms remains poorly defined. The relationship between the anorexigenic hormones, insulin and leptin, and the orexigenic hormone, ghrelin, on mTORC1 signalling was examined using S6 kinase phosphorylation as a marker for changes in mTORC1 activity in mouse hypothalamic GT1-7 cells. Additionally, the contribution of AMPK and mTORC1 signalling in relation to insulin-, leptin- and ghrelin-driven alterations to mouse hypothalamic agouti-related protein (AgRP) mRNA levels was examined. Insulin and leptin increase mTORC1 activity in a phosphoinositide-3-kinase (PI3K)- and protein kinase B (PKB)-dependent manner, compared to vehicle controls, whereas increasing AMPK activity inhibits mTORC1 activity and blocks the actions of the anorexigenic hormones. Ghrelin mediates an AMPK-dependent decrease in mTORC1 activity and increases hypothalamic AgRP mRNA levels, the latter effect being prevented by insulin in an mTORC1-dependent manner. In conclusion, mTORC1 acts as an integration node in hypothalamic neurons for hormone-derived PI3K and AMPK signalling and mediates at least part of the assimilated output of anorexigenic and orexigenic hormone actions in the hypothalamus.


Asunto(s)
Proteína Relacionada con Agouti/biosíntesis , Apetito/fisiología , Hipotálamo/metabolismo , Complejos Multiproteicos/metabolismo , ARN Mensajero/biosíntesis , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Anorexia/metabolismo , Femenino , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Endogámicos C57BL
16.
Neurosci Lett ; 515(1): 87-91, 2012 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-22450045

RESUMEN

It is well established that under fasting conditions the expression of the orexigenic neuropeptide agouti-related peptide (AGRP) is up-regulated in the hypothalamic arcuate nucleus (ARC), while inconsistent data exist regarding fasting regulation of the anorexigenic neurohormone proopiomelanocortin (POMC). Inconsistencies might have methodological reasons, especially concerning neuromorphological and/or experimental (nutritional) specificity. We analyzed the expression of both neuropeptides in ARC neurons, using lasercapture microdissection (LMD) and real-time PCR in 12h fasted vs. fed Wistar rats as well as after a standardized glucose load, i.e., under clinically relevant conditions in terms of diagnosing glucose intolerance in the human. Under fasting conditions, clear up-regulation of AGRP was observed, with increasing magnitude in ARC single neurons (SNP) as compared to ARC cell layers (+125% vs. +23%, resp.), closely correlated to hypoinsulinemia and hypoleptinemia. Surprisingly, in the fasting state POMC was not found to be down-regulated, neither in ARC cell layers nor in ARC single neurons (+9% vs. +6%). However, glucose-refeeding under diagnostically relevant conditions led to strong neuronal up-regulation of POMC expression in ARC SNP (+128%), and AGRP down-regulation (-50%). In conclusion, experimentally, topographically, and analytically specific and standardized conditions confirmed AGRP in ARC neurons as being neuronally up- and down-regulated, resp., depending on the general nutritional state, while POMC was found to be (up-) regulated only after peripheral glucose load. Findings suggest that POMC in ARC neurons acts glucose-mediated as an "anti-orexigenic" neurohormone, specifically responding to hyperglycemia.


Asunto(s)
Proteína Relacionada con Agouti/biosíntesis , Ayuno/metabolismo , Glucosa/metabolismo , Hipotálamo/metabolismo , Captura por Microdisección con Láser/métodos , Proopiomelanocortina/biosíntesis , Animales , Ingestión de Alimentos/fisiología , Masculino , Ratas , Ratas Wistar , Factores de Tiempo
17.
PLoS Biol ; 9(1): e1000575, 2011 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-21264353

RESUMEN

Obesity is a growing epidemic characterized by excess fat storage in adipocytes. Although lipoprotein receptors play important roles in lipid uptake, their role in controlling food intake and obesity is not known. Here we show that the lipoprotein receptor LRP1 regulates leptin signaling and energy homeostasis. Conditional deletion of the Lrp1 gene in the brain resulted in an obese phenotype characterized by increased food intake, decreased energy consumption, and decreased leptin signaling. LRP1 directly binds to leptin and the leptin receptor complex and is required for leptin receptor phosphorylation and Stat3 activation. We further showed that deletion of the Lrp1 gene specifically in the hypothalamus by Cre lentivirus injection is sufficient to trigger accelerated weight gain. Together, our results demonstrate that the lipoprotein receptor LRP1, which is critical in lipid metabolism, also regulates food intake and energy homeostasis in the adult central nervous system.


Asunto(s)
Encéfalo/metabolismo , Metabolismo Energético , Leptina/fisiología , Receptores de LDL/deficiencia , Proteínas Supresoras de Tumor/deficiencia , Proteína Relacionada con Agouti/biosíntesis , Proteína Relacionada con Agouti/genética , Animales , Regulación del Apetito , Línea Celular , Femenino , Intolerancia a la Glucosa/genética , Intolerancia a la Glucosa/metabolismo , Homeostasis , Hiperlipidemias/genética , Hiperlipidemias/metabolismo , Hipotálamo/metabolismo , Resistencia a la Insulina , Metabolismo de los Lípidos/efectos de los fármacos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Masculino , Ratones , Ratones Noqueados , Neuropéptido Y/biosíntesis , Neuropéptido Y/genética , Obesidad/genética , Obesidad/metabolismo , Receptores de LDL/genética , Proteínas Supresoras de Tumor/genética , Regulación hacia Arriba
18.
Int J Cancer ; 128(9): 2215-23, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-20602340

RESUMEN

Parathyroid hormone-related protein (PTHrP) is a causative factor of humoral hypercalcemia in malignancy. However, it is difficult to explain the mechanism of anorexia/cachexia with PTHrP secretion in detail. Previously, we demonstrated that the expressions of orexigenic peptides increased and anorexigenic peptides decreased under cachectic conditions in rats carrying tumors secreting PTHrP. In this study, we investigated whether such changes in the expression of hypothalamic feeding-regulating peptides can be solely attributed to PTHrP or are a general response under cachectic conditions. Cachectic syndromes were induced in rats by: (i) inoculation of human lung cancer LC-6 cells that secreted PTHrP, (ii) inoculation of human melanoma SEKI cells that secrete not PTHrP but LIF1, (iii) injection of heat-killed Mycobacterium leading to arthritis (AA) and (iv) oral administration of a high dose of 1α,25(OH)(2)D(3) that resulted in hypercalcemia. The LC-6-bearing rats and AA rats were treated with or without anti-PTHrP antibody and indomethacin, respectively, and the expression of the hypothalamic feeding-regulating peptide mRNAs were examined by in situ hybridization histochemistry. The orexigenic peptide mRNAs, such as neuropeptide Y and agouti-related protein, were significantly increased, and that of anorexigenic peptide mRNAs, such as proopiomelanocortin, cocaine- and amphetamine-regulated transcript and corticotropin-releasing hormone were significantly decreased when they developed cachectic syndromes and AA. A high dose of 1α,25(OH)(2)D(3) caused hypercalcemia and body weight loss but did not affect the expression of hypothalamic feeding-regulating peptide mRNAs. The expressions of the hypothalamic feeding-regulating peptides change commonly in different chronic cachectic models without relating to serum calcium levels.


Asunto(s)
Artritis Experimental/metabolismo , Caquexia/metabolismo , Hipotálamo/metabolismo , Leptina/sangre , Factor Inhibidor de Leucemia/metabolismo , Neoplasias Experimentales/metabolismo , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Proteína Relacionada con Agouti/biosíntesis , Animales , Artritis Experimental/complicaciones , Línea Celular Tumoral , Hormona Liberadora de Corticotropina/biosíntesis , Humanos , Hipercalcemia/etiología , Hibridación in Situ , Péptidos y Proteínas de Señalización Intracelular , Masculino , Neoplasias Experimentales/complicaciones , Proteínas del Tejido Nervioso/biosíntesis , Neuropéptido Y/biosíntesis , Neuropéptidos/biosíntesis , Orexinas , Proopiomelanocortina/biosíntesis , ARN Mensajero/análisis , Ratas , Ratas Desnudas , Ratas Wistar
19.
J Neurosci ; 30(35): 11815-25, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20810901

RESUMEN

Sirt1 has been associated with various effects of calorie restriction, including an increase in lifespan. Here we show in mice that a central regulatory component in energy metabolism, the hypothalamic melanocortin system, is affected by Sirt1, which promotes the activity and connectivity of this system resulting in negative energy balance. In adult mice, the pharmacological inhibition of brain Sirt1 activity decreased Agrp neuronal activity and the inhibitory tone on the anorexigenic POMC neurons, as measured by the number of synaptic inputs to these neurons. When a Sirt1 inhibitor (EX-527) was injected either peripherally (i.p., 10 mg/kg) or directly into the brain (i.c.v., 1.5 nmol/mouse), it decreased both food intake during the dark cycle and ghrelin-induced food intake. This effect on feeding is mediated by upstream melanocortin receptors, because the MC4R antagonist, SHU9119, reversed Sirt1's effect on food intake. This action of Sirt1 required an appropriate shift in the mitochondrial redox state: in the absence of such an adaptation enabled by the mitochondrial protein, UCP2, Sirt1-induced cellular and behavioral responses were impaired. In accordance with the pharmacological results, the selective knock-out of Sirt1 in hypothalamic Agrp neurons through the use of Cre-Lox technology decreased electric responses of Agrp neurons to ghrelin and decreased food intake, leading to decreased lean mass, fat mass, and body weight. The present data indicate that Sirt1 has a central mode of action by acting on the NPY/Agrp neurons to affect body metabolism.


Asunto(s)
Proteína Relacionada con Agouti/fisiología , Metabolismo Energético/fisiología , Melanocortinas/fisiología , Plasticidad Neuronal/fisiología , Neuronas/fisiología , Sirtuina 1/fisiología , Sinapsis/fisiología , Potenciales Sinápticos/fisiología , Proteína Relacionada con Agouti/biosíntesis , Animales , Carbazoles/administración & dosificación , Combinación de Medicamentos , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Metabolismo Energético/efectos de los fármacos , Femenino , Masculino , Melanocortinas/metabolismo , Hormonas Estimuladoras de los Melanocitos/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mitocondrias/efectos de los fármacos , Mitocondrias/fisiología , Plasticidad Neuronal/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Oxidación-Reducción/efectos de los fármacos , Sirtuina 1/antagonistas & inhibidores , Sirtuina 1/deficiencia , Sinapsis/efectos de los fármacos , Potenciales Sinápticos/efectos de los fármacos
20.
BMC Neurosci ; 11: 94, 2010 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-20701764

RESUMEN

BACKGROUND: Multiple neuropeptides, sometimes with opposing functions, can be produced from one precursor gene. To study the roles of the different neuropeptides encoded by one large precursor we developed a method to overexpress minigenes and establish local secretion. RESULTS: We fused the signal peptide from the Von Willebrand Factor (VWF) to a furin site followed by a processed form of the Agouti related protein (AgRP), AgRP(83-132) or alpha-melanocyte stimulating hormone. In vitro, these minigenes were secreted and biologically active. Additionally, the proteins of the minigenes were not transported into projections of primary neurons, thereby ensuring local release. In vivo administration of VWF-AgRP(83-132), using an adeno-associated viral vector as a delivery vehicle, into the paraventricular hypothalamus increased body weight and food intake of these rats compared to rats which received a control vector. CONCLUSIONS: This study demonstrated that removal of the N-terminal part of full length AgRP and addition of a VWF signal peptide is a successful strategy to deliver neuropeptide minigenes to the brain and establish local neuropeptide secretion.


Asunto(s)
Neuropéptidos/metabolismo , Señales de Clasificación de Proteína/fisiología , Factor de von Willebrand/fisiología , Proteína Relacionada con Agouti/biosíntesis , Proteína Relacionada con Agouti/genética , Secuencia de Aminoácidos , Animales , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , ADN Complementario/biosíntesis , ADN Complementario/genética , Dependovirus/genética , Técnicas de Transferencia de Gen , Células HEK293 , Humanos , Immunoblotting , Inmunohistoquímica , Hibridación in Situ , Operón Lac , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Señales de Clasificación de Proteína/genética , Receptor de Melanocortina Tipo 4/biosíntesis , Receptor de Melanocortina Tipo 4/genética , alfa-MSH/biosíntesis , alfa-MSH/genética , Factor de von Willebrand/biosíntesis , Factor de von Willebrand/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA