Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Peptides ; 136: 170444, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33245952

RESUMEN

VGF is a peptide precursor expressed in neuroendocrine cells that is suggested to play a role in the regulation of energy homeostasis. VGF is proteolytically cleaved to yield multiple bioactive peptides. However, the specific actions of VGF-derived peptides on energy homeostasis remain unclear. The aim of the present work was to investigate the role of VGF-derived peptides in energy homeostasis and explore the pharmacological actions of VGF-derived peptides on body weight in preclinical animal models. VGF-derived peptides (NERP-1, NERP-2, PGH-NH2, PGH-OH, NERP-4, TLQP-21, TLQP-30, TLQP-62, HHPD-41, AQEE-30, and LQEQ-19) were synthesized and screened for their ability to affect neuronal activity in vitro on hypothalamic brain slices and modulate food intake and energy expenditure after acute central administration in vivo. In addition, the effects of NERP-1, NERP-2, PGH-NH2, TLQP-21, TLQP-62, and HHPD-41 on energy homeostasis were studied after chronic central infusion. NERP-1, PGH-NH2, HHPD-41, and TLQP-62 increased the functional activity of hypothalamic neuronal networks. However, none of the peptides altered energy homeostasis after either acute or chronic ICV administration. The present data do not support the potential use of the tested VGF-derived peptides as novel anti-obesity drug candidates.


Asunto(s)
Fármacos Antiobesidad/farmacología , Neuropéptidos/genética , Neuropéptidos/farmacología , Obesidad/tratamiento farmacológico , Animales , Peso Corporal/efectos de los fármacos , Modelos Animales de Enfermedad , Metabolismo Energético/efectos de los fármacos , Humanos , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Hipotálamo/patología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Obesidad/genética , Obesidad/patología , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/farmacología , Ratas
2.
Nat Commun ; 11(1): 4458, 2020 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-32895383

RESUMEN

In rodent models of type 2 diabetes (T2D), sustained remission of hyperglycemia can be induced by a single intracerebroventricular (icv) injection of fibroblast growth factor 1 (FGF1), and the mediobasal hypothalamus (MBH) was recently implicated as the brain area responsible for this effect. To better understand the cellular response to FGF1 in the MBH, we sequenced >79,000 single-cell transcriptomes from the hypothalamus of diabetic Lepob/ob mice obtained on Days 1 and 5 after icv injection of either FGF1 or vehicle. A wide range of transcriptional responses to FGF1 was observed across diverse hypothalamic cell types, with glial cell types responding much more robustly than neurons at both time points. Tanycytes and ependymal cells were the most FGF1-responsive cell type at Day 1, but astrocytes and oligodendrocyte lineage cells subsequently became more responsive. Based on histochemical and ultrastructural evidence of enhanced cell-cell interactions between astrocytes and Agrp neurons (key components of the melanocortin system), we performed a series of studies showing that intact melanocortin signaling is required for the sustained antidiabetic action of FGF1. These data collectively suggest that hypothalamic glial cells are leading targets for the effects of FGF1 and that sustained diabetes remission is dependent on intact melanocortin signaling.


Asunto(s)
Diabetes Mellitus Experimental/dietoterapia , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Factor 1 de Crecimiento de Fibroblastos/administración & dosificación , Hipoglucemiantes/administración & dosificación , Hipotálamo/efectos de los fármacos , Proteínas Recombinantes/administración & dosificación , Proteína Relacionada con Agouti/metabolismo , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Glucemia/análisis , Comunicación Celular , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/etiología , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/etiología , Diabetes Mellitus Tipo 2/patología , Dieta Alta en Grasa/efectos adversos , Sacarosa en la Dieta/administración & dosificación , Sacarosa en la Dieta/efectos adversos , Humanos , Hipotálamo/citología , Hipotálamo/patología , Inyecciones Intraventriculares , Leptina/genética , Masculino , Melanocortinas/metabolismo , Hormonas Estimuladoras de los Melanocitos/administración & dosificación , Ratones , Ratones Noqueados , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , RNA-Seq , Receptor de Melanocortina Tipo 4/genética , Receptores de Melanocortina/antagonistas & inhibidores , Receptores de Melanocortina/metabolismo , Inducción de Remisión/métodos , Transducción de Señal/efectos de los fármacos , Análisis de la Célula Individual , Técnicas Estereotáxicas , Transcriptoma/efectos de los fármacos
3.
Am J Physiol Regul Integr Comp Physiol ; 313(2): R169-R179, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28404581

RESUMEN

Maternal high-fat-diet (HFD) consumption during pregnancy decreased fetal body weight and impacted development of hypothalamic melanocortin neural circuitry in nonhuman primate offspring. We investigated whether these impairments during gestation persisted in juvenile offspring and examined the interaction between maternal and early postnatal HFD consumption. Adult dams consumed either a control diet (CTR; 15% calories from fat) or a high-saturated-fat diet (HFD; 37% calories from fat) during pregnancy. Offspring were weaned onto a CTR or HFD at ~8 mo of age. Offspring from HFD-fed dams displayed early catch-up growth and elevated body weight at 6 and 13 mo of age. Maternal and postnatal HFD exposure reduced the amount of agouti-related peptide fibers in the paraventricular nucleus of the hypothalamus. Postnatal HFD consumption also decreased the amount of agouti-related peptide fibers in the arcuate nucleus of the hypothalamus. Postnatal HFD was associated with decreased food intake and increased activity. These results support and extend our previous findings of maternal diet effects on fetal development and reveal, for the first time in a nonhuman primate model, that maternal HFD-induced disturbances in offspring body weight regulation extended past gestation into the juvenile period. Maternal HFD consumption increases the risk for offspring developing obesity, with the developmental timing of HFD exposure differentially impacting the melanocortin system and energy balance regulation. The present findings provide translational insight into human clinical populations, suggesting that profound health consequences may await individuals later in life following intrauterine and postnatal HFD exposure.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Ingestión de Alimentos , Hipotálamo/fisiopatología , Melanocortinas/metabolismo , Obesidad/fisiopatología , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Animales , Metabolismo Energético , Conducta Alimentaria , Femenino , Desarrollo Fetal , Humanos , Macaca , Masculino , Obesidad/etiología , Embarazo , Preñez , Transducción de Señal
4.
J Clin Invest ; 124(10): 4473-88, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25202980

RESUMEN

Liraglutide is a glucagon-like peptide-1 (GLP-1) analog marketed for the treatment of type 2 diabetes. Besides lowering blood glucose, liraglutide also reduces body weight. It is not fully understood how liraglutide induces weight loss or to what degree liraglutide acts directly in the brain. Here, we determined that liraglutide does not activate GLP-1-producing neurons in the hindbrain, and liraglutide-dependent body weight reduction in rats was independent of GLP-1 receptors (GLP-1Rs) in the vagus nerve, area postrema, and paraventricular nucleus. Peripheral injection of fluorescently labeled liraglutide in mice revealed the presence of the drug in the circumventricular organs. Moreover, labeled liraglutide bound neurons within the arcuate nucleus (ARC) and other discrete sites in the hypothalamus. GLP-1R was necessary for liraglutide uptake in the brain, as liraglutide binding was not seen in Glp1r(-/-) mice. In the ARC, liraglutide was internalized in neurons expressing proopiomelanocortin (POMC) and cocaine- and amphetamine-regulated transcript (CART). Electrophysiological measurements of murine brain slices revealed that GLP-1 directly stimulates POMC/CART neurons and indirectly inhibits neurotransmission in neurons expressing neuropeptide Y (NPY) and agouti-related peptide (AgRP) via GABA-dependent signaling. Collectively, our findings indicate that the GLP-1R on POMC/CART-expressing ARC neurons likely mediates liraglutide-induced weight loss.


Asunto(s)
Péptido 1 Similar al Glucagón/análogos & derivados , Receptores de Glucagón/metabolismo , Pérdida de Peso/efectos de los fármacos , Animales , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Modelos Animales de Enfermedad , Electrofisiología , Péptido 1 Similar al Glucagón/farmacología , Receptor del Péptido 1 Similar al Glucagón , Hipotálamo/metabolismo , Liraglutida , Masculino , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Proopiomelanocortina/metabolismo , Ratas , Ratas Sprague-Dawley , Nervio Vago/metabolismo
5.
FASEB J ; 28(6): 2466-77, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24563374

RESUMEN

Resveratrol has been proposed as a potential therapeutic to improve metabolic health during pregnancy, yet little is known about the fetal effects of this maternal dietary supplement. We hypothesized that when administered to pregnant nonhuman primates (NHPs), resveratrol would increase uterine blood flow and mitigate the harmful consequences of maternal Western-style diet (WSD) consumption. NHPs were fed a WSD (36% fat) supplemented with 0.37% resveratrol throughout pregnancy. Outcomes were compared with cohorts fed WSD alone and control chow (14% fat) to distinguish between WSD and resveratrol-specific effects in these animals. In the early third trimester, uterine blood flow was measured by Doppler ultrasound before fetal delivery and tissue collection. Resveratrol resulted in 30% maternal weight loss and improved glucose tolerance, increased uterine artery volume blood flow, and decreased placental inflammation and liver triglyceride deposition. In addition, fetal pancreatic mass was enlarged by 42%, with a 12-fold increase in proliferation by Ki67 immunohistochemistry. These results demonstrate that resveratrol use during pregnancy yields improvements in maternal and placental phenotype with beneficial effects in the fetal liver but an unexplained and concerning alteration in fetal pancreatic development, which strongly cautions against the use of resveratrol by pregnant women.


Asunto(s)
Desarrollo Fetal/efectos de los fármacos , Estilbenos/efectos adversos , Estilbenos/farmacología , Animales , Contraindicaciones , Dieta/efectos adversos , Suplementos Dietéticos/efectos adversos , Femenino , Feto , Hígado/efectos de los fármacos , Hígado/embriología , Macaca , Páncreas/efectos de los fármacos , Páncreas/embriología , Circulación Placentaria/efectos de los fármacos , Embarazo , Flujo Sanguíneo Regional/efectos de los fármacos , Resveratrol , Estilbenos/sangre , Triglicéridos/sangre , Útero/irrigación sanguínea
6.
J Neurosci ; 33(38): 15306-17, 2013 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-24048859

RESUMEN

Neuropeptide Y (NPY) neurons in both the arcuate nucleus of the hypothalamus (ARH) and the dorsomedial hypothalamus (DMH) have been implicated in food intake and obesity. However, while ARH NPY is highly expressed in the lean animal, DMH NPY mRNA expression is observed only after diet-induced obesity (DIO). Furthermore, while ARH NPY neurons are inhibited by leptin, the effect of this adipokine on DMH NPY neurons is unknown. In this study we show that in contrast to the consistent expression in the ARH, DMH NPY mRNA expression was undetectable until after 10 weeks in mice fed a high-fat diet, and peaked at 20 weeks. Surprisingly, electrophysiological experiments demonstrated that leptin directly depolarized and increased the firing rate of DMH NPY neurons in DIO mice. To further differentiate the regulation of DMH and ARH NPY populations, fasting decreased expression of DMH NPY expression, while it increased ARH NPY expression. However, treatment with a leptin receptor antagonist failed to alter DMH NPY expression, indicating that leptin may not be the critical factor regulating mRNA expression. Importantly, we also demonstrated that DMH NPY neurons coexpress cocaine amphetamine-regulated transcript (CART); however, CART mRNA expression in the DMH peaked earlier in the progression of DIO. This study demonstrates novel and important findings. First, NPY and CART are coexpressed in the same neurons within the DMH, and second, leptin stimulates DMH NPY neurons. These studies suggest that during the progression of DIO, there is an unknown signal that drives the expression of the orexigenic NPY signal within the DMH, and that the chronic hyperleptinemia increases the activity of these NPY/CART neurons.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Leptina/farmacología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Neuropéptido Y/metabolismo , Obesidad/patología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/genética , Análisis de Varianza , Animales , Dieta/efectos adversos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Hipotálamo/patología , Técnicas In Vitro , Insulina/sangre , Leptina/antagonistas & inhibidores , Leptina/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Neuronas/efectos de los fármacos , Neuropéptido Y/genética , Obesidad/sangre , Obesidad/etiología , Técnicas de Placa-Clamp , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , ARN Mensajero/metabolismo , Radioinmunoensayo , Factor de Transcripción STAT3/metabolismo , Factores de Tiempo
7.
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
8.
Mol Endocrinol ; 26(12): 2071-80, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23015752

RESUMEN

Thyroid hormone (TH) is an essential regulator of both fetal development and energy homeostasis. Although the association between subclinical hypothyroidism and obesity has been well studied, a causal relationship has yet to be established. Using our well-characterized nonhuman primate model of excess nutrition, we sought to investigate whether maternal high-fat diet (HFD)-induced changes in TH homeostasis may underlie later in life development of metabolic disorders and obesity. Here, we show that in utero exposure to a maternal HFD is associated with alterations of the fetal thyroid axis. At the beginning of the third trimester, fetal free T(4) levels are significantly decreased with HFD exposure compared with those of control diet-exposed offspring. Furthermore, transcription of the deiodinase, iodothyronine (DIO) genes, which help maintain thyroid homeostasis, are significantly (P < 0.05) disrupted in the fetal liver, thyroid, and hypothalamus. Genes involved in TH production are decreased (TRH, TSHR, TG, TPO, and SLC5A5) in hypothalamus and thyroid gland. In experiments designed to investigate the molecular underpinnings of these observations, we observe that the TH nuclear receptors and their downstream regulators are disrupted with maternal HFD exposure. In fetal liver, the expression of TH receptor ß (THRB) is increased 1.9-fold (P = 0.012). Thorough analysis of the THRB promoter reveals a maternal diet-induced alteration in the fetal THRB histone code, alongside differential promoter occupancy of corepressors and coactivators. We speculate that maternal HFD exposure in utero may set the stage for later in life obesity through epigenomic modifications to the histone code, which modulates the fetal thyroid axis.


Asunto(s)
Dieta Alta en Grasa , Fenómenos Fisiologicos Nutricionales Maternos , Efectos Tardíos de la Exposición Prenatal , Glándula Tiroides/embriología , Receptores beta de Hormona Tiroidea/genética , Animales , Grasas de la Dieta/metabolismo , Femenino , Expresión Génica , Hipotálamo/embriología , Hipotiroidismo , Yoduro Peroxidasa/genética , Hígado/embriología , Macaca/embriología , Obesidad , Embarazo , Regiones Promotoras Genéticas , Glándula Tiroides/metabolismo , Receptores beta de Hormona Tiroidea/biosíntesis , Receptores beta de Hormona Tiroidea/metabolismo , Hormonas Tiroideas/genética , Hormonas Tiroideas/metabolismo
9.
PLoS One ; 6(2): e17261, 2011 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-21364873

RESUMEN

To begin to understand the contributions of maternal obesity and over-nutrition to human development and the early origins of obesity, we utilized a non-human primate model to investigate the effects of maternal high-fat feeding and obesity on breast milk, maternal and fetal plasma fatty acid composition and fetal hepatic development. While the high-fat diet (HFD) contained equivalent levels of n-3 fatty acids (FA's) and higher levels of n-6 FA's than the control diet (CTR), we found significant decreases in docosahexaenoic acid (DHA) and total n-3 FA's in HFD maternal and fetal plasma. Furthermore, the HFD fetal plasma n-6:n-3 ratio was elevated and was significantly correlated to the maternal plasma n-6:n-3 ratio and maternal hyperinsulinemia. Hepatic apoptosis was also increased in the HFD fetal liver. Switching HFD females to a CTR diet during a subsequent pregnancy normalized fetal DHA, n-3 FA's and fetal hepatic apoptosis to CTR levels. Breast milk from HFD dams contained lower levels of eicosopentanoic acid (EPA) and DHA and lower levels of total protein than CTR breast milk. This study links chronic maternal consumption of a HFD with fetal hepatic apoptosis and suggests that a potentially pathological maternal fatty acid milieu is replicated in the developing fetal circulation in the nonhuman primate.


Asunto(s)
Apoptosis/efectos de los fármacos , Grasas de la Dieta/efectos adversos , Ácidos Grasos Omega-3/sangre , Feto/efectos de los fármacos , Hígado/efectos de los fármacos , Fenómenos Fisiologicos Nutricionales Maternos , Animales , Apoptosis/fisiología , Dieta Aterogénica , Ácidos Grasos Omega-3/análisis , Ácidos Grasos Omega-3/metabolismo , Femenino , Sangre Fetal/química , Sangre Fetal/metabolismo , Feto/metabolismo , Feto/patología , Humanos , Hígado/embriología , Hígado/patología , Embarazo , Efectos Tardíos de la Exposición Prenatal/patología , Primates , Distribución Aleatoria
10.
PLoS One ; 3(5): e2202, 2008 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-18493584

RESUMEN

BACKGROUND: Cannabinoids, the active components of marijuana, stimulate appetite, and cannabinoid receptor-1 (CB1-R) antagonists suppress appetite and promote weight loss. Little is known about how CB1-R antagonists affect the central neurocircuitry, specifically the melanocortin system that regulates energy balance. METHODOLOGY/PRINCIPAL FINDINGS: Here, we show that peripherally administered CB1-R antagonist (AM251) or agonist equally suppressed or stimulated feeding respectively in A(y) , which lack a functional melanocortin system, and wildtype mice, demonstrating that cannabinoid effects on feeding do not require melanocortin circuitry. CB1-R antagonist or agonist administered into the ventral tegmental area (VTA) equally suppressed or stimulated feeding respectively, in both genotypes. In addition, peripheral and central cannabinoid administration similarly induced c-Fos activation in brain sites suggesting mediation via motivational dopaminergic circuitry. Amperometry-detected increases in evoked dopamine (DA) release by the CB1-R antagonist in nucleus accumbens slices indicates that AM251 modulates DA release from VTA terminals. CONCLUSIONS/SIGNIFICANCE: Our results demonstrate that the effects of cannabinoids on energy balance are independent of hypothalamic melanocortin circuitry and is primarily driven by the reward system.


Asunto(s)
Cannabinoides/farmacología , Conducta Alimentaria/efectos de los fármacos , Melanocortinas/fisiología , Animales , Hipotálamo/efectos de los fármacos , Hipotálamo/fisiología , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley
11.
Endocrinology ; 148(7): 3279-87, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17412803

RESUMEN

The hypothalamic neurocircuitry that regulates energy homeostasis in adult rats is not fully developed until the third postnatal week. In particular, fibers from the hypothalamic arcuate nucleus, including both neuropeptide Y (NPY) and alpha-MSH fibers, do not begin to innervate downstream hypothalamic targets until the second postnatal week. However, alpha-MSH fibers from the brainstem and melanocortin receptors are present in the hypothalamus at birth. The present study investigated the melanocortin system in the early postnatal period by examining effects of the melanocortin receptor agonist melanotan II (MTII) on body weight, energy expenditure, and hypothalamic NPY expression. Rat pups were injected ip with MTII (3 mg/kg body weight) or saline on postnatal day (P) 5 to P6, P10-P11, or P15-P16 at 1700 and 0900 h and then killed at 1300 h. Stomach weight and brown adipose tissue uncoupling protein 1 mRNA were determined. In addition, we assessed central c-Fos activation 90 min after MTII administration and hypothalamic NPY mRNA after twice daily MTII administration from P5-P10 or P10-P15. MTII induced hypothalamic c-Fos activation as well as attenuating body weight gain in rat pups. Stomach weight was significantly decreased and uncoupling protein 1 mRNA was increased at all ages, indicating decreased food intake and increased energy expenditure, respectively. However, MTII had no effect on NPY mRNA levels in any hypothalamic region. These findings demonstrate that MTII can inhibit food intake and stimulate energy expenditure before the full development of hypothalamic feeding neurocircuitry. These effects do not appear to be mediated by changes in NPY expression.


Asunto(s)
Ingestión de Alimentos/efectos de los fármacos , Canales Iónicos/genética , Proteínas Mitocondriales/genética , Péptidos Cíclicos/farmacología , Receptores de Corticotropina/metabolismo , alfa-MSH/análogos & derivados , Animales , Animales Lactantes , Relación Dosis-Respuesta a Droga , Femenino , Mucosa Gástrica/metabolismo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Hipotálamo/crecimiento & desarrollo , Hipotálamo/metabolismo , Inmunohistoquímica , Hibridación in Situ , Masculino , Neuropéptido Y/genética , Tamaño de los Órganos/efectos de los fármacos , Péptidos Cíclicos/administración & dosificación , Proteínas Proto-Oncogénicas c-fos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Receptores de Corticotropina/agonistas , Estómago/efectos de los fármacos , Estómago/crecimiento & desarrollo , Proteína Desacopladora 1 , alfa-MSH/administración & dosificación , alfa-MSH/farmacología
12.
Cell Metab ; 5(3): 181-94, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17339026

RESUMEN

Despite high leptin levels, most obese humans and rodents lack responsiveness to its appetite-suppressing effects. We demonstrate that leptin modulates NPY/AgRP and alpha-MSH secretion from the ARH of lean mice. High-fat diet-induced obese (DIO) mice have normal ObRb levels and increased SOCS-3 levels, but leptin fails to modulate peptide secretion and any element of the leptin signaling cascade. Despite this leptin resistance, the melanocortin system downstream of the ARH in DIO mice is over-responsive to melanocortin agonists, probably due to upregulation of MC4R. Lastly, we show that by decreasing the fat content of the mouse's diet, leptin responsiveness of NPY/AgRP and POMC neurons recovered simultaneously, with mice regaining normal leptin sensitivity and glycemic control. These results highlight the physiological importance of leptin sensing in the melanocortin circuits and show that their loss of leptin sensing likely contributes to the pathology of leptin resistance.


Asunto(s)
Núcleo Arqueado del Hipotálamo/metabolismo , Leptina/farmacología , Neuronas/metabolismo , Obesidad/metabolismo , Proteína Relacionada con Agouti , Animales , Núcleo Arqueado del Hipotálamo/citología , Composición Corporal , Dieta , Grasas de la Dieta/administración & dosificación , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/efectos de los fármacos , Hipotálamo/metabolismo , Técnicas In Vitro , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Leptina/administración & dosificación , Masculino , Melanocortinas/metabolismo , Ratones , Ratones Endogámicos C57BL , Neuropéptido Y/metabolismo , Proopiomelanocortina/metabolismo , ARN Mensajero , Transducción de Señal , Pérdida de Peso , alfa-MSH/metabolismo
13.
Cell Metab ; 4(6): 421-2, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17141625

RESUMEN

Signals from the hypothalamus govern food intake and energy balance. A new study describes nesfatin-1, a hypothalamic and brainstem peptide whose expression decreases during fasting. Although central treatment with nesfatin-1 inhibited food intake and nesfatin-1 blockade increased food intake, the role and mechanism of nesfatin in energy balance remains unclear.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión al ADN/metabolismo , Ingestión de Alimentos , Metabolismo Energético , Hipotálamo/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Transducción de Señal , Animales , Proteínas de Unión al Calcio/antagonistas & inhibidores , Proteínas de Unión al ADN/antagonistas & inhibidores , Metabolismo Energético/efectos de los fármacos , Ayuno/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/farmacología , Nucleobindinas , Ratas , Transducción de Señal/efectos de los fármacos
14.
J Clin Invest ; 115(12): 3393-7, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16322785

RESUMEN

Ghrelin, produced in the stomach, acts on growth hormone secretagogue receptors (GHSRs) in hypothalamic neurons to potently increase food intake. However, male mice with deletions of ghrelin (Ghrl-/- mice) or GHSR (Ghsr-/- mice) display normal growth and regulation of food intake. Furthermore, adult Ghrl-/- mice display a normal sensitivity to high-fat diet-induced obesity. These findings from early studies raised the question as to whether the ghrelin system is an essential component for the regulation of food intake and body weight homeostasis. However, recent studies by Wortley et al. and Zigman et al. demonstrate that Ghrl-/- and Ghsr-/- mice are resistant to diet-induced obesity when fed a high-fat diet during the early post-weaning period. This commentary highlights 3 key issues raised by these 2 reports: (a) the impact of ghrelin on the development of metabolic systems; (b) the constitutive activity of GHSR; and (c) gender differences in the sensitivity to deletion of the ghrelin signaling system.


Asunto(s)
Hormonas Peptídicas/fisiología , Alimentación Animal , Animales , Peso Corporal , Dieta , Femenino , Alimentos , Eliminación de Gen , Ghrelina , Hipotálamo/metabolismo , Leptina/metabolismo , Masculino , Ratones , Ratones Transgénicos , Modelos Biológicos , Neuronas/metabolismo , Obesidad/genética , Obesidad/metabolismo , Obesidad/terapia , Hormonas Peptídicas/antagonistas & inhibidores , Hormonas Peptídicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Ghrelina , Factores Sexuales , Transducción de Señal , Esteroides/metabolismo
15.
Endocrinology ; 145(11): 5344-54, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15256488

RESUMEN

Lactation and fasting are two physiological models characterized by negative energy balance. Our previous studies demonstrated that uncoupling protein (UCP) 3 expression in skeletal muscle was down-regulated during lactation and up-regulated during fasting. The present studies used cDNA microarray and real-time PCR to perform a systems and comparative analysis in gene expression in skeletal muscle under conditions of negative energy balance. Gastrocnemius skeletal muscle RNA pools were generated from the following groups of rats: cycling diestrous females, cycling females with 48 h of fasting, lactation, and lactation + leptin. Of those known genes studied, 35 genes were up-regulated and 49 were down-regulated during lactation. Leptin treatment during lactation reversed the differential regulation of about 80% of these genes, demonstrating the importance of the leptin suppression to the changes in skeletal muscle metabolism. GenMAPP analysis revealed a coordinated regulation at key steps in glycolysis/gluconeogenesis, the tricarboxylic acid cycle, and lipid metabolism, indicating an increased rate of lactate production through glycolysis and reduced fatty acid degradation in skeletal muscle during lactation. Particular interest was paid to those genes that changed in a similar manner to UCP3 mRNA. Many of these genes that were decreased during lactation and increased during fasting are involved in fatty acid degradation and transport, including acyl-coenzyme A dehydrogenase for medium chain fatty acid, carnitine palmitoyltransferase 1, and fatty acid translocase. The current studies provide a basis for investigating the mechanisms underlying metabolic adaptations during lactation and fasting and highlight the importance of UCP3 in lipid metabolism.


Asunto(s)
Adaptación Fisiológica/fisiología , Perfilación de la Expresión Génica , Lactancia/fisiología , Músculo Esquelético/metabolismo , Animales , Proteínas Portadoras/genética , Ciclo del Ácido Cítrico/fisiología , Metabolismo Energético/fisiología , Ayuno/fisiología , Ácidos Grasos/metabolismo , Femenino , Gluconeogénesis/fisiología , Glucólisis/fisiología , Canales Iónicos , Leptina/farmacología , Proteínas Mitocondriales , Análisis de Secuencia por Matrices de Oligonucleótidos , Embarazo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética/fisiología , Proteína Desacopladora 3
16.
J Neurosci ; 24(22): 5091-100, 2004 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-15175378

RESUMEN

In several hyperphagic models, including lactation, in which hypothalamic melanocortin signaling is reduced, a novel expression of NPY mRNA in the dorsomedial hypothalamus (DMH) has been observed, suggesting that melanocortin signaling and the induced NPY in the DMH may constitute unique neurocircuitry in mediating energy balance. Using lactating rats as a model, the present study first showed that in the DMH abundant alpha-MSH and agouti-related protein fibers are in close apposition to NPY-positive cells. However, no NPY and MC4R (a melanocortin receptor) double-labeled neurons were observed. These data suggested that melanocortin input may synapse on presynaptic terminals that then synapse on DMH NPY cells. To study the function of DMH MC4Rs in energy balance, an MC3/4R-selective agonist, melanotan II (MTII), was injected bilaterally into the DMH. MTII injection significantly suppressed feeding induced by 24 hr fasting or suckling-induced hyperphagia. Furthermore, MTII treatment greatly attenuated suckling-induced NPY expression in the DMH. MTII treatment also stimulated uncoupling protein 1 activity in the brown adipose tissue of suckling female rats, indicative of increased sympathetic outflow. In summary, the present study demonstrated that the melanocortin system in the DMH not only plays an important role in inducing NPY expression in the DMH of lactating rats but also in regulating energy homeostasis, at least in part, by modulating appetite and energy expenditure.


Asunto(s)
Hiperfagia/fisiopatología , Hipotálamo/metabolismo , Lactancia/fisiología , Neuropéptido Y/metabolismo , Receptor de Melanocortina Tipo 4/metabolismo , alfa-MSH/análogos & derivados , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/metabolismo , Proteína Relacionada con Agouti , Animales , Regulación del Apetito/fisiología , Proteínas Portadoras/genética , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Metabolismo Energético/fisiología , Femenino , Expresión Génica/efectos de los fármacos , Glutamato Descarboxilasa/genética , Homeostasis/fisiología , Hipotálamo/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular , Canales Iónicos , Isoenzimas/genética , Leptina/sangre , Proteínas de la Membrana/genética , Proteínas Mitocondriales , Modelos Animales , Fibras Nerviosas/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuropéptido Y/genética , Péptidos Cíclicos/farmacología , Proteínas/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor de Melanocortina Tipo 4/agonistas , Receptor de Melanocortina Tipo 4/genética , Proteína Desacopladora 1 , alfa-MSH/metabolismo , alfa-MSH/farmacología
17.
Physiol Behav ; 79(1): 47-63, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12818709

RESUMEN

Early onset obesity and type II diabetes is rapidly becoming an epidemic, especially within the United States. This dramatic increase is likely due to many factors including both prenatal and postnatal environmental cues. The purpose of this review is to highlight some of the recent advances in our knowledge of the development of the hypothalamic circuits involved in the regulation of energy balance, with a focus on the neuropeptide Y (NPY) system. Unlike the adult rat, during the postnatal period NPY is transiently expressed in several hypothalamic regions, along with the expected expression within the arcuate nucleus (ARH). These transient populations of NPY neurons during the postnatal period may provide local NPY production to sustain the necessary energy intake during this critical growth phase. This may be physiologically important since ARH-NPY projections do not fully develop until the 3rd postnatal week. The significance of this ontogeny is that many peripheral metabolic signals have little effect of feeding prior to the development of the ARH projections. The essential questions now are whether prenatal and/or postnatal exposure to high levels of insulin or leptin during development can cause permanent changes in the function of hypothalamic circuits. It is vital to understand not only the natural development of the hypothalamic circuits that regulate energy homeostasis, but also their abnormal development caused by maternal and postnatal environmental cues. This will be pivotal for designing intervention and therapeutics to treat early onset obesity/type II diabetes, which may very well need to be different from those designed to prevent/treat adult onset obesity/type II diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/fisiopatología , Diabetes Mellitus/fisiopatología , Hipotálamo/fisiología , Neuropéptido Y/fisiología , Obesidad , Animales , Núcleo Arqueado del Hipotálamo/fisiopatología , Femenino , Humanos , Insulina/fisiología , Leptina/fisiología , Masculino , Red Nerviosa/fisiopatología , Embarazo , Efectos Tardíos de la Exposición Prenatal , Ratas , Ratas Endogámicas , alfa-MSH/fisiología
18.
Brain Res ; 973(2): 223-32, 2003 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-12738066

RESUMEN

An abundance of physiological data suggests an interaction between neuropeptide Y (NPY) and corticotropin-releasing hormone (CRH) in the regulation of endocrine and autonomic functions. Previously, studies in our laboratory have indicated that NPY neurons in the arcuate nucleus of the hypothalamus (ARH) project to and come in close contact with CRH neurons in the paraventricular nucleus of the hypothalamus (PVH). Conversely, it has been demonstrated that the ventromedial portion of the ARH, an area containing NPY neurons, displays CRH receptor binding and CRH receptor mRNA. These data suggest a possible reciprocal feedback regulation between NPY and CRH neurons. The ARH also contains several other populations of neurons that may be targets of the CRH system and express CRH receptors; most notable are tuberoinfundibular dopaminergic neurons (TIDA). The PVH is an important component in the regulation of prolactin secretion and may play a role in the suppression of TIDA activity, which is a critical step in the prolactin stress response. The purpose of the present study was to characterize the distribution and cellular localization of CRH R(1) receptor-like immunoreactivity (CRH R(1)-ir) in the rat hypothalamus and to determine the phenotype of neurons in the ARH that contain CRH R(1)-ir. CRH R(1)-ir was present throughout the rat brain. Hypothalamic regions with the highest levels of immunostaining were the supraoptic nucleus, magnocellular PVH, ARH, and suprachiasmatic nucleus. Double label immunofluorescence was used to demonstrate that CRH R(1)-ir in the ARH was localized to NPY cell bodies. Furthermore, TIDA neurons in the ARH also displayed CRH R(1)-ir. However, despite an abundance of CRH R(1)-ir cells in the ARH, CRH-ir fiber innervation to the ARH was extremely sparse. Therefore, although this study provides neuroanatomical evidence for direct CRH R(1) regulation of ARH NPY and TIDA neurons in the rat, it is not consistent with the idea of a reciprocal feedback loop and suggests the involvement of other CRH-like ligands, such as urocortin.


Asunto(s)
Núcleo Arqueado del Hipotálamo/metabolismo , Hormona Liberadora de Corticotropina/metabolismo , Dopamina/metabolismo , Hipotálamo/metabolismo , Neuronas/metabolismo , Neuropéptido Y/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/citología , Hormona Liberadora de Corticotropina/genética , Femenino , Hipotálamo/anatomía & histología , Inmunohistoquímica/métodos , Hibridación in Situ/métodos , Microscopía Confocal/instrumentación , Vías Nerviosas/citología , Vías Nerviosas/metabolismo , Prosencéfalo/anatomía & histología , Prosencéfalo/metabolismo , Ratas
19.
Neuron ; 37(4): 649-61, 2003 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-12597862

RESUMEN

The gastrointestinal peptide hormone ghrelin stimulates appetite in rodents and humans via hypothalamic actions. We discovered expression of ghrelin in a previously uncharacterized group of neurons adjacent to the third ventricle between the dorsal, ventral, paraventricular, and arcuate hypothalamic nuclei. These neurons send efferents onto key hypothalamic circuits, including those producing neuropeptide Y (NPY), Agouti-related protein (AGRP), proopiomelanocortin (POMC) products, and corticotropin-releasing hormone (CRH). Within the hypothalamus, ghrelin bound mostly on presynaptic terminals of NPY neurons. Using electrophysiological recordings, we found that ghrelin stimulated the activity of arcuate NPY neurons and mimicked the effect of NPY in the paraventricular nucleus of the hypothalamus (PVH). We propose that at these sites, release of ghrelin may stimulate the release of orexigenic peptides and neurotransmitters, thus representing a novel regulatory circuit controlling energy homeostasis.


Asunto(s)
Sistema Nervioso Central/metabolismo , Metabolismo Energético/fisiología , Homeostasis/fisiología , Hipotálamo/metabolismo , Red Nerviosa/metabolismo , Hormonas Peptídicas/metabolismo , Proteínas , Proteína Relacionada con Agouti , Animales , Sistema Nervioso Central/citología , Hormona Liberadora de Corticotropina/biosíntesis , Femenino , Ghrelina , Hipotálamo/citología , Hipotálamo/efectos de los fármacos , Técnicas In Vitro , Péptidos y Proteínas de Señalización Intercelular , Proteínas Luminiscentes/biosíntesis , Ratones , Ratones Noqueados , Ratones Transgénicos , Neuronas/citología , Neuronas/metabolismo , Neuropéptido Y/biosíntesis , Especificidad de Órganos , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/metabolismo , Técnicas de Placa-Clamp , Hormonas Peptídicas/farmacología , Terminales Presinápticos/metabolismo , Proopiomelanocortina/biosíntesis , Unión Proteica/fisiología , Biosíntesis de Proteínas , Ratas
20.
J Neurosci ; 23(4): 1487-97, 2003 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-12598637

RESUMEN

The receptor subtypes that mediate the effects of neuropeptide Y (NPY) on food intake have not been clearly defined. The NPY Y4 receptor has been identified recently as a potential mediator of the regulation of food intake. The purpose of the present study was to characterize the central site of action of the Y4 receptor using a combination of neuroanatomical and physiological approaches. Using immunocytochemistry, Y4-like immunoreactivity was found to be colocalized with orexin cell bodies in the lateral hypothalamic area (LHA) and orexin fibers throughout the brain. In situ hybridization confirmed the expression of Y4 mRNA in orexin neurons. To determine the functional interaction between Y4 receptors and orexin neurons, we examined the effects of rat pancreatic polypeptide (rPP), a Y4-selective ligand, or NPY, a nonselective ligand, administered directly into the LHA on the stimulation of food and water intake and c-Fos expression. Both rPP and NPY significantly increased food and water intake when they were administered into the LHA, although NPY was a more potent stimulator of food intake. Furthermore, both NPY and rPP significantly stimulated c-Fos expression in the LHA. However, whereas rPP stimulated c-Fos expression in orexin neurons, NPY did not. Neither rPP nor NPY stimulated c-Fos in melanin-concentrating hormone neurons, but both activated neurons of an unknown phenotype in the LHA. These results suggest that a functional Y4 receptor is expressed on orexin neurons and that these neurons are activated in response to a ligand with high affinity for the Y4 receptor (rPP). Although these data suggest a role for central Y4 receptors, the endogenous ligand for this receptor has yet to be clearly established.


Asunto(s)
Proteínas Portadoras/análisis , Ingestión de Alimentos , Hipotálamo/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Neuronas/metabolismo , Neuropéptidos/análisis , Receptores de Neuropéptido Y/fisiología , Animales , Ingestión de Líquidos/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Femenino , Hipotálamo/química , Inmunohistoquímica , Hibridación in Situ , Masculino , Neuronas/química , Neuropéptido Y/farmacología , Receptores de Orexina , Orexinas , Polipéptido Pancreático/farmacología , Proteínas Proto-Oncogénicas c-fos/metabolismo , ARN Mensajero/biosíntesis , Ratas , Receptores Acoplados a Proteínas G , Receptores de Neuropéptido , Receptores de Neuropéptido Y/genética , Receptores de Neuropéptido Y/metabolismo , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA