Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Cureus ; 14(6): e25989, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35859973

RESUMEN

Background Patients require vitamin and mineral supplementation after bariatric surgery to prevent the development of micronutrient deficiencies. Consuming oral supplements is challenging due to gastric volume restriction. A transdermal patch dosage form may provide adequate micronutrient supplementation without pill burden. The study aims to determine the percentage of patients who have two or more nutritional deficiencies one year after surgery and to determine serum nutrient concentrations and gastrointestinal symptoms over time. Methods Patients who planned to undergo bariatric surgery and preferred transdermal patches versus oral supplements were recruited during preoperative office visits. Enrolled patients were instructed to use a transdermal multivitamin patch as per the manufacturer's instructions. Serum nutrient concentrations and Gastrointestinal Symptom Response Scale scores were determined at baseline and three months, six months, and one year after surgery. Results Ninety-two participants completed the study protocol. Twenty-five participants had a full panel of study labs one year after surgery. Among these patients, 19% had two or more micronutrient deficiencies. Vitamin D was the most common deficiency followed by vitamin B6; however, median serum concentrations of both nutrients increased over time. Vitamin B1, folate, and zinc deficiencies were also observed. There were no changes in gastrointestinal symptoms. Conclusions Additional studies, including randomized controlled trials, are required to determine if the PatchMD Multivitamin Plus patch (Pilot Rd. STE. B, Las Vegas) can provide adequate supplementation of vitamins and minerals. The patch was not associated with changes in gastrointestinal symptoms.

2.
Biol Psychiatry ; 91(10): 869-878, 2022 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-34593204

RESUMEN

The understanding of the neural control of appetite sheds light on the pathogenesis of eating disorders such as anorexia nervosa and obesity. Both diseases are a result of maladaptive eating behaviors (overeating or undereating) and are associated with life-threatening health problems. The fine regulation of appetite involves genetic, physiological, and environmental factors, which are detected and integrated in the brain by specific neuronal populations. For centuries, the hypothalamus has been the center of attention in the scientific community as a key regulator of appetite. The hypothalamus receives and sends axonal projections to several other brain regions that are important for the integration of sensory and emotional information. These connections ensure that appropriate behavioral decisions are made depending on the individual's emotional state and environment. Thus, the mechanisms by which higher-order brain regions integrate exteroceptive information to coordinate feeding is of great importance. In this review, we will focus on the functional and anatomical projections connecting the hypothalamus to the limbic system and higher-order brain centers in the cortex. We will also address the mechanisms by which specific neuronal populations located in higher-order centers regulate appetite and how maladaptive eating behaviors might arise from altered connections among cortical and subcortical areas with the hypothalamus.


Asunto(s)
Apetito , Trastornos de Alimentación y de la Ingestión de Alimentos , Encéfalo , Humanos , Hipotálamo , Obesidad
4.
Cell Rep ; 19(3): 655-667, 2017 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-28423326

RESUMEN

Translational profiling methodologies enable the systematic characterization of cell types in complex tissues, such as the mammalian brain, where neuronal isolation is exceptionally difficult. Here, we report a versatile strategy for profiling CNS cell types in a spatiotemporally restricted fashion by engineering a Cre-dependent adeno-associated virus expressing an EGFP-tagged ribosomal protein (AAV-FLEX-EGFPL10a) to access translating mRNAs by translating ribosome affinity purification (TRAP). We demonstrate the utility of this AAV to target a variety of genetically and anatomically defined neural populations expressing Cre recombinase and illustrate the ability of this viral TRAP (vTRAP) approach to recapitulate the molecular profiles obtained by bacTRAP in corticothalamic neurons across multiple serotypes. Furthermore, spatially restricting adeno-associated virus (AAV) injections enabled the elucidation of regional differences in gene expression within this cell type. Altogether, these results establish the broad applicability of the vTRAP strategy for the molecular dissection of any CNS or peripheral cell type that can be engineered to express Cre.


Asunto(s)
Cromatografía de Afinidad/métodos , Biosíntesis de Proteínas , Ribosomas/metabolismo , Virus/metabolismo , Animales , Biomarcadores/metabolismo , Dependovirus/metabolismo , Femenino , Regulación de la Expresión Génica , Proteínas Fluorescentes Verdes/metabolismo , Hormonas Hipotalámicas/metabolismo , Hipotálamo/metabolismo , Masculino , Melaninas/metabolismo , Ratones , Neuronas/metabolismo , Hormonas Hipofisarias/metabolismo , Reproducibilidad de los Resultados , Serotipificación
5.
J Clin Invest ; 126(12): 4727-4734, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27906690

RESUMEN

Leptin is an adipose tissue hormone that functions as an afferent signal in a negative feedback loop that maintains homeostatic control of adipose tissue mass. This endocrine system thus serves a critical evolutionary function by protecting individuals from the risks associated with being too thin (starvation) or too obese (predation and temperature dysregulation). Mutations in leptin or its receptor cause massive obesity in mice and humans, and leptin can effectively treat obesity in leptin-deficient patients. Leptin acts on neurons in the hypothalamus and elsewhere to elicit its effects, and mutations that affect the function of this neural circuit cause Mendelian forms of obesity. Leptin levels fall during starvation and elicit adaptive responses in many other physiologic systems, the net effect of which is to reduce energy expenditure. These effects include cessation of menstruation, insulin resistance, alterations of immune function, and neuroendocrine dysfunction, among others. Some or all of these effects are also seen in patients with constitutively low leptin levels, such as occur in lipodystrophy. Leptin is an approved treatment for generalized lipodystrophy, a condition associated with severe metabolic disease, and has also shown potential for the treatment of other types of diabetes. In addition, leptin restores reproductive capacity and increases bone mineral density in patients with hypothalamic amenorrhea, an infertility syndrome in females. Most obese patients have high endogenous levels of leptin, in some instances as a result of mutations in the neural circuit on which leptin acts, though in most cases, the pathogenesis of leptin resistance is not known. Obese patients with leptin resistance show a variable response to exogenous leptin but may respond to a combination of leptin plus amylin. Overall, the identification of leptin has provided a framework for studying the pathogenesis of obesity in the general population, clarified the nature of the biologic response to starvation, and helped to advance our understanding of the neural mechanisms that control feeding.


Asunto(s)
Tejido Adiposo , Diabetes Mellitus , Leptina , Lipodistrofia , Obesidad , Inanición , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patología , Humanos , Hipotálamo/metabolismo , Hipotálamo/patología , Leptina/genética , Leptina/metabolismo , Leptina/uso terapéutico , Lipodistrofia/tratamiento farmacológico , Lipodistrofia/genética , Lipodistrofia/metabolismo , Lipodistrofia/patología , Ratones , Neuronas/metabolismo , Neuronas/patología , Obesidad/tratamiento farmacológico , Obesidad/genética , Obesidad/metabolismo , Obesidad/patología , Receptores de Leptina/genética , Receptores de Leptina/metabolismo , Inanición/tratamiento farmacológico , Inanición/genética , Inanición/metabolismo , Inanición/patología
6.
Cell Metab ; 22(6): 1059-67, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26655697

RESUMEN

In this report we evaluated the functions of hypothalamic amylin in vivo and in vitro. Profiling of hypothalamic neurons revealed that islet amyloid polypeptide (Iapp, precursor to amylin) is expressed in neurons in the lateral hypothalamus, arcuate nucleus, medial preoptic area, and elsewhere. Hypothalamic expression of lapp is markedly decreased in ob/ob mice and normalized by exogenous leptin. In slices, amylin and leptin had similar electrophysiologic effects on lateral hypothalamic leptin receptor ObRb-expressing neurons, while the amylin antagonist AC187 inhibited their activity and blunted the effect of leptin. Finally, i.c.v. infusion of AC187 acutely reduced the anorectic effects of leptin. These data show that hypothalamic amylin is transcriptionally regulated by leptin, that it can act directly on ObRb neurons in concert with leptin, and that it regulates feeding. These findings provide a potential mechanism for the increased efficacy of a metreleptin/pramlintide combination therapy for obesity.


Asunto(s)
Hipotálamo/metabolismo , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Leptina/metabolismo , Animales , Ingestión de Alimentos/efectos de los fármacos , Encefalinas/genética , Encefalinas/metabolismo , Femenino , Hipotálamo/efectos de los fármacos , Polipéptido Amiloide de los Islotes Pancreáticos/antagonistas & inhibidores , Polipéptido Amiloide de los Islotes Pancreáticos/farmacología , Leptina/sangre , Leptina/farmacología , Masculino , Potenciales de la Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Microscopía Confocal , Neuronas/metabolismo , Técnicas de Placa-Clamp , Fragmentos de Péptidos/farmacología , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , ARN Mensajero/metabolismo , Receptores de Leptina/genética , Receptores de Leptina/metabolismo
7.
Cell Metab ; 18(4): 596-607, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24093682

RESUMEN

Comprehensive transcriptional profiling of glucose-sensing neurons is challenging because of low expression levels of glucokinase (Gck) and other key proteins that transduce a glucose signal. To overcome this, we generated and validated transgenic mice with a neuronal/endocrine-specific Gck promoter driving cre expression and mated them to mice with cre-dependent expression of an EGFP-tagged ribosomal protein construct (EEF1A1-LSL.EGFPL10) that can be used to map and profile cells. We found significant Gck expression in hypothalamic and limbic regions in cells that are activated following administration of glucose or 2-deoxyglucose. Transcriptional profiling from Gck-cre/EEF1A1-LSL.EGFPL10 mice enriched known and previously unknown glucose-sensing populations including neurons expressing growth hormone releasing hormone (GHRH). Electrophysiological recordings show that hypoglycemia activates GHRH neurons, suggesting a mechanistic link between hypoglycemia and growth hormone release. These studies provide a means for mapping glucose-sensitive neurons and for generating transcriptional profiles from other cell types expressing cre in a cell-specific manner.


Asunto(s)
Perfilación de la Expresión Génica , Hormona Liberadora de Hormona del Crecimiento/metabolismo , Neuronas/metabolismo , Animales , Desoxiglucosa/farmacología , Glucoquinasa/genética , Glucoquinasa/metabolismo , Glucosa/farmacología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hipoglucemia/metabolismo , Hipoglucemia/patología , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/efectos de los fármacos , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/metabolismo
8.
Nat Neurosci ; 16(11): 1637-43, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24056699

RESUMEN

Rapid-eye movement (REM) sleep correlates with neuronal activity in the brainstem, basal forebrain and lateral hypothalamus. Lateral hypothalamus melanin-concentrating hormone (MCH)-expressing neurons are active during sleep, but their effects on REM sleep remain unclear. Using optogenetic tools in newly generated Tg(Pmch-cre) mice, we found that acute activation of MCH neurons (ChETA, SSFO) at the onset of REM sleep extended the duration of REM, but not non-REM, sleep episodes. In contrast, their acute silencing (eNpHR3.0, archaerhodopsin) reduced the frequency and amplitude of hippocampal theta rhythm without affecting REM sleep duration. In vitro activation of MCH neuron terminals induced GABAA-mediated inhibitory postsynaptic currents in wake-promoting histaminergic neurons of the tuberomammillary nucleus (TMN), and in vivo activation of MCH neuron terminals in TMN or medial septum also prolonged REM sleep episodes. Collectively, these results suggest that activation of MCH neurons maintains REM sleep, possibly through inhibition of arousal circuits in the mammalian brain.


Asunto(s)
Hipotálamo/fisiología , Red Nerviosa/fisiología , Vías Nerviosas/fisiología , Optogenética , Sueño REM/fisiología , 6-Ciano 7-nitroquinoxalina 2,3-diona/farmacología , Animales , Animales Recién Nacidos , Bicuculina/farmacología , Channelrhodopsins , Antagonistas de Aminoácidos Excitadores/farmacología , Antagonistas de Receptores de GABA-A/farmacología , Regulación de la Expresión Génica , Hormonas Hipotalámicas/genética , Hipotálamo/citología , Hipotálamo/efectos de los fármacos , Melaninas/genética , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Red Nerviosa/efectos de los fármacos , Vías Nerviosas/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/fisiología , Hormonas Hipofisarias/genética , Ritmo Teta/efectos de los fármacos , Ritmo Teta/genética , Transducción Genética , Valina/análogos & derivados , Valina/farmacología
9.
Elife ; 2: e01462, 2013 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-24381247

RESUMEN

Sugars that contain glucose, such as sucrose, are generally preferred to artificial sweeteners owing to their post-ingestive rewarding effect, which elevates striatal dopamine (DA) release. While the post-ingestive rewarding effect, which artificial sweeteners do not have, signals the nutrient value of sugar and influences food preference, the neural circuitry that mediates the rewarding effect of glucose is unknown. In this study, we show that optogenetic activation of melanin-concentrating hormone (MCH) neurons during intake of the artificial sweetener sucralose increases striatal dopamine levels and inverts the normal preference for sucrose vs sucralose. Conversely, animals with ablation of MCH neurons no longer prefer sucrose to sucralose and show reduced striatal DA release upon sucrose ingestion. We further show that MCH neurons project to reward areas and are required for the post-ingestive rewarding effect of sucrose in sweet-blind Trpm5(-/-) mice. These studies identify an essential component of the neural pathways linking nutrient sensing and food reward. DOI: http://dx.doi.org/10.7554/eLife.01462.001.


Asunto(s)
Glucosa/metabolismo , Hipotálamo/metabolismo , Melaninas/metabolismo , Neuronas/metabolismo , Valor Nutritivo , Animales , Hipotálamo/química , Ratones , Recompensa
10.
Cell ; 151(5): 1126-37, 2012 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-23178128

RESUMEN

The mammalian brain is composed of thousands of interacting neural cell types. Systematic approaches to establish the molecular identity of functional populations of neurons would advance our understanding of neural mechanisms controlling behavior. Here, we show that ribosomal protein S6, a structural component of the ribosome, becomes phosphorylated in neurons activated by a wide range of stimuli. We show that these phosphorylated ribosomes can be captured from mouse brain homogenates, thereby enriching directly for the mRNAs expressed in discrete subpopulations of activated cells. We use this approach to identify neurons in the hypothalamus regulated by changes in salt balance or food availability. We show that galanin neurons are activated by fasting and that prodynorphin neurons restrain food intake during scheduled feeding. These studies identify elements of the neural circuit that controls food intake and illustrate how the activity-dependent capture of cell-type-specific transcripts can elucidate the functional organization of a complex tissue.


Asunto(s)
Encéfalo/metabolismo , Neuronas/metabolismo , Ribosomas/metabolismo , Transcriptoma , Animales , Encéfalo/citología , Ayuno , Conducta Alimentaria , Hipotálamo/citología , Hipotálamo/metabolismo , Ratones , Fosforilación , Proteína S6 Ribosómica/metabolismo
11.
Curr Med Res Opin ; 28(2): 195-201, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22208675

RESUMEN

BACKGROUND AND OBJECTIVE: Dabigatran is a new oral anticoagulant recently approved for the prevention of stroke or systemic embolism in patients with atrial fibrillation (AF). Based on its pharmacokinetic profile, dabigatran is dosed twice daily. This analysis provides a quantitative rationale for the selection of the dose regimen in this population. METHODS: The pharmacokinetic profile of dabigatran was simulated for AF patients given a total daily dose of 300 mg, either once or twice daily. Simulations were based on a population pharmacokinetic model supplemented with data collected from 9522 patients enrolled in a pivotal phase III study (RE-LY). RESULTS: The typical RE-LY patient (male, 72 years old, Caucasian, weight 80 kg, creatinine clearance 68.64 mL/min) treated with dabigatran 150 mg twice daily showed less than two-fold difference between peak-trough plasma levels compared with a five-fold difference when the same total dose (300 mg) was administered once daily. For patients who miss or delay taking one scheduled dabigatran dose, twice daily dosing maintained adequate minimum trough concentrations better than once daily dosing. Pharmacokinetic data collected from a phase II study and RE-LY were consistent with the simulation results. The study did not access comparative efficacy and bleeding data for once versus twice daily dosing. CONCLUSION: Pharmacokinetic simulations show that a twice daily regimen in patients with AF minimizes daily fluctuations in plasma concentrations of dabigatran and can maintain trough concentrations sufficient to prevent the development of thrombi while at the same time minimizing the risk of bleeding due to supratherapeutic peak plasma concentrations. The efficacy and safety of this dosing regimen is supported by clinical data from the RE-LY trial.


Asunto(s)
Fibrilación Atrial/patología , Bencimidazoles/administración & dosificación , Bencimidazoles/farmacocinética , Accidente Cerebrovascular/prevención & control , beta-Alanina/análogos & derivados , Anciano , Anticoagulantes/administración & dosificación , Anticoagulantes/uso terapéutico , Fibrilación Atrial/tratamiento farmacológico , Bencimidazoles/uso terapéutico , Ensayos Clínicos Fase II como Asunto , Ensayos Clínicos Fase III como Asunto , Simulación por Computador , Dabigatrán , Esquema de Medicación , Humanos , Masculino , Ensayos Clínicos Controlados Aleatorios como Asunto , Accidente Cerebrovascular/tratamiento farmacológico , Trombina/antagonistas & inhibidores , beta-Alanina/administración & dosificación , beta-Alanina/farmacocinética , beta-Alanina/uso terapéutico
12.
J Clin Invest ; 121(1): 355-68, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21183787

RESUMEN

Studies in humans and rodents indicate that a minimum amount of stored energy is required for normal pubertal development. The adipocyte-derived hormone leptin is a key metabolic signal to the neuroendocrine reproductive axis. Humans and mice lacking leptin or the leptin receptor (LepR) (ob/ob and db/db mice, respectively) are infertile and fail to enter puberty. Leptin administration to leptin-deficient subjects and ob/ob mice induces puberty and restores fertility, but the exact site or sites of leptin action are unclear. Here, we found that genetic deletion of LepR selectively from hypothalamic Kiss1 neurons in mice had no effect on puberty or fertility, indicating that direct leptin signaling in Kiss1 neurons is not required for these processes. However, bilateral lesions of the ventral premammillary nucleus (PMV) of ob/ob mice blunted the ability of exogenous leptin to induce sexual maturation. Moreover, unilateral reexpression of endogenous LepR in PMV neurons was sufficient to induce puberty and improve fertility in female LepR-null mice. This LepR reexpression also normalized the increased hypothalamic GnRH content characteristic of leptin-signaling deficiency. These data suggest that the PMV is a key site for leptin's permissive action at the onset of puberty and support the hypothesis that the multiple actions of leptin to control metabolism and reproduction are anatomically dissociated.


Asunto(s)
Hipotálamo/metabolismo , Leptina/metabolismo , Proteínas/metabolismo , Maduración Sexual/fisiología , Animales , Secuencia de Bases , Femenino , Fertilidad/genética , Fertilidad/fisiología , Expresión Génica , Humanos , Kisspeptinas , Leptina/deficiencia , Leptina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Ratones Transgénicos , Neuronas/metabolismo , Embarazo , Proteínas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Leptina/deficiencia , Receptores de Leptina/genética , Receptores de Leptina/metabolismo , Maduración Sexual/genética , Transducción de Señal
13.
Aesthet Surg J ; 30(6): 783-8, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21131449

RESUMEN

BACKGROUND: Various techniques have been described for periorbital rejuvenation and correction of the ptotic brow, including the coronal brow lift, the endoscopic brow lift, anterior hairline foreheadplasty in the subgaleal, subperiosteal, or subcutaneous planes, and the subcutaneous temporal brow lift. OBJECTIVES: The authors present results from a series of 28 patients treated with subcutaneous temporal brow lift over nearly four years. METHODS: A retrospective chart review was conducted of 28 patients who were treated with subcutaneous temporal brow lift by the senior author (JDF) between July 2003 and January 2007. All patients underwent the same subcutaneous temporal brow lift procedure under local anesthesia in an office-based setting. No combined procedures were performed on any of the patients in this series. RESULTS: Of the 28 patients, 27 were female and one was male; mean age was 54 years. Five patients underwent a unilateral brow lift for asymmetry and 23 patients underwent a bilateral procedure. The mean length of follow-up was 10.8 months. Scarring was minimal and rated as "good" or "excellent" by both patients and surgeon. The effectiveness of the browlift was also rated as "good" or "excellent" by all but one patient. Two patients underwent revision-one for scar revision and the other for a greater degree of lift. There were no incidences of hematoma, infection, numbness, or excessive scarring. CONCLUSIONS: The subcutaneous temporal brow lift is an effective, reproducible, and inexpensive technique that can be performed safely under local anesthesia.


Asunto(s)
Anestesia Local , Cejas , Rejuvenecimiento , Ritidoplastia/métodos , Adulto , Anciano , Femenino , Frente , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Tejido Subcutáneo
14.
Proc Natl Acad Sci U S A ; 107(15): 7024-9, 2010 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-20351287

RESUMEN

The autonomic nervous system regulates fuel availability and energy storage in the liver, adipose tissue, and other organs; however, the molecular components of this neural circuit are poorly understood. We sought to identify neural populations that project from the CNS indirectly through multisynaptic pathways to liver and epididymal white fat in mice using pseudorabies virus strains expressing different reporters together with BAC transgenesis and immunohistochemistry. Neurons common to both circuits were identified in subpopulations of the paraventricular nucleus of the hypothalamus (PVH) by double labeling with markers expressed in viruses injected in both sites. The lateral hypothalamus and arcuate nucleus of the hypothalamus and brainstem regions (nucleus of the solitary tract and A5 region) also project to both tissues but are labeled at later times. Connections from these same sites to the PVH were evident after direct injection of virus into the PVH, suggesting that these regions lie upstream of the PVH in a common pathway to liver and adipose tissue (two metabolically active organs). These common populations of brainstem and hypothalamic neurons express neuropeptide Y and proopiomelanocortin in the arcuate nucleus, melanin-concentrating hormone, and orexin in the lateral hypothalamus and in the corticotrophin-releasing hormone and oxytocin in the PVH. The delineation of this circuitry will facilitate a functional analysis of the possible role of these potential command-like neurons to modulate autonomic outflow and coordinate metabolic responses in liver and adipose tissue.


Asunto(s)
Tejido Adiposo/patología , Hipotálamo/patología , Hígado/patología , Neuronas/metabolismo , Animales , Tronco Encefálico/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Hormonas Hipotalámicas/metabolismo , Hipotálamo/metabolismo , Inmunohistoquímica/métodos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Hígado/metabolismo , Masculino , Melaninas/metabolismo , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Neuropéptido Y/metabolismo , Neuropéptidos/metabolismo , Orexinas , Hormonas Hipofisarias/metabolismo , Proopiomelanocortina/metabolismo
15.
Cell Metab ; 5(1): 21-33, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17189204

RESUMEN

The active thyroid hormone, triiodothyronine (T3), regulates mitochondrial uncoupling protein activity and related thermogenesis in peripheral tissues. Type 2 deiodinase (DII), an enzyme that catalyzes active thyroid hormone production, and mitochondrial uncoupling protein 2 (UCP2) are also present in the hypothalamic arcuate nucleus, where their interaction and physiological significance have not been explored. Here, we report that DII-producing glial cells are in direct apposition to neurons coexpressing neuropeptide Y (NPY), agouti-related protein (AgRP), and UCP2. Fasting increased DII activity and local thyroid hormone production in the arcuate nucleus in parallel with increased GDP-regulated UCP2-dependent mitochondrial uncoupling. Fasting-induced T3-mediated UCP2 activation resulted in mitochondrial proliferation in NPY/AgRP neurons, an event that was critical for increased excitability of these orexigenic neurons and consequent rebound feeding following food deprivation. These results reveal a physiological role for a thyroid-hormone-regulated mitochondrial uncoupling in hypothalamic neuronal networks.


Asunto(s)
Núcleo Arqueado del Hipotálamo/metabolismo , Ayuno , Conducta Alimentaria , Canales Iónicos/metabolismo , Proteínas Mitocondriales/metabolismo , Neuronas/metabolismo , Termogénesis , Triyodotironina/metabolismo , Proteína Relacionada con Agouti , Animales , Núcleo Arqueado del Hipotálamo/citología , Ingestión de Alimentos , Proteínas Fluorescentes Verdes , Guanosina Difosfato/metabolismo , Hipotálamo/citología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Yoduro Peroxidasa/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Neuroglía/metabolismo , Neuropéptido Y/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteína Desacopladora 2 , Yodotironina Deyodinasa Tipo II
16.
J Neurosci ; 26(2): 389-97, 2006 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-16407534

RESUMEN

The observation that loss of orexin (hypocretin) neurons causes human narcolepsy raises the possibility that other acquired disorders might also result from loss of hypothalamic neurons. To test this possibility for body weight, mice with selective loss of melanin concentrating hormone (MCH) neurons were generated. MCH was chosen to test because induced mutations of the MCH gene in mice cause hypophagia and leanness. Mice with ablation of MCH neurons were generated using toxin (ataxin-3)-mediated ablation strategy. The mice appeared normal but, after 7 weeks, developed reduced body weight, body length, fat mass, lean mass, and leptin levels. Leanness was characterized by hypophagia and increased energy expenditure. To study the role of MCH neurons on obesity secondary to leptin deficiency, we generated mice deficient in both ob gene product (leptin) and MCH neurons. Absence of MCH neurons in ob/ob mice improved obesity, diabetes, and hepatic steatosis, suggesting that MCH neurons are important mediators of the response to leptin deficiency. These data show that loss of MCH neurons can lead to an acquired leanness. This has implications for the pathogenesis of acquired changes of body weight and might be considered in clinical settings characterized by substantial weight changes later in life.


Asunto(s)
Hormonas Hipotalámicas/fisiología , Hipotálamo/fisiopatología , Leptina/deficiencia , Melaninas/fisiología , Proteínas del Tejido Nervioso/fisiología , Neuronas/fisiología , Hormonas Hipofisarias/fisiología , Delgadez/etiología , Tejido Adiposo/patología , Animales , Ataxina-3 , Glucemia/análisis , Muerte Celular , Cruzamientos Genéticos , Diabetes Mellitus Experimental/fisiopatología , Ingestión de Energía , Metabolismo Energético , Ayuno/sangre , Hígado Graso/fisiopatología , Femenino , Expresión Génica , Hormonas Hipotalámicas/análisis , Hipotálamo/patología , Insulina/sangre , Leptina/genética , Leptina/uso terapéutico , Enfermedad de Machado-Joseph/genética , Masculino , Melaninas/análisis , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Neuropéptidos/metabolismo , Proteínas Nucleares , Obesidad/genética , Obesidad/fisiopatología , Hormonas Hipofisarias/análisis , Proteínas Recombinantes de Fusión/fisiología , Proteínas Represoras , Factores de Transcripción , Pérdida de Peso
17.
J Comp Neurol ; 482(3): 217-43, 2005 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-15690487

RESUMEN

The central neuropeptide Y (NPY) Y1 receptor (Y1-R) system has been implicated in feeding, endocrine, and autonomic regulation. In the present study, we systematically examined the brain distribution of Y1-R mRNA in rodents by using radioisotopic in situ hybridization histochemistry (ISHH) with a novel sensitive cRNA probe. Within the rat hypothalamus, Y1-R-specific hybridization was observed in the anteroventral periventricular, ventromedial preoptic, suprachiasmatic, paraventricular (PVH), dorsomedial, ventromedial, arcuate, and mamillary nuclei. In the rat, Y1-R mRNA expression was also seen in the subfornical organ, anterior hypothalamic area, dorsal hypothalamic area, and in the lateral hypothalamic area. In addition, Y1-R hybridization was evident in several extrahypothalamic forebrain and hindbrain sites involved in feeding and/or autonomic regulation in the rat. A similar distribution pattern of Y1-R mRNA was observed in the mouse brain. Moreover, by using a transgenic mouse line expressing green fluorescent protein under the control of the melanocortin-4 receptor (MC4-R) promoter, we observed Y1-R mRNA expression in MC4-R-positive cells in several brain sites such as the PVH and central nucleus of the amygdala. Additionally, dual-label ISHH demonstrated that hypophysiotropic PVH cells coexpress Y1-R and pro-thyrotropin-releasing hormone mRNAs in the rat. These observations are consistent with the proposed roles of the central NPY/Y1-R system in energy homeostasis.


Asunto(s)
Mapeo Encefálico , Hipotálamo/metabolismo , ARN Mensajero/metabolismo , Receptor de Melanocortina Tipo 4/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropéptido/metabolismo , Animales , Regulación del Apetito/fisiología , Conducta Alimentaria/fisiología , Hibridación Fluorescente in Situ , Masculino , Ratones , Ratones Transgénicos , Prosencéfalo/metabolismo , ARN Complementario/análisis , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/genética , Receptores de Neuropéptido/genética , Rombencéfalo , Distribución Tisular
18.
Proc Natl Acad Sci U S A ; 102(7): 2537-42, 2005 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-15699332

RESUMEN

Food restriction and weight loss result in reduced plasma leptin, which is associated with a pleiotropic biologic response. However, because weight loss itself is also associated with changes in numerous other humoral and metabolic signals, it can be difficult to determine the precise features of the biologic response to acute leptin deficiency. To study this response in the absence of changes in nutritional state, we have developed a protocol that allows such analysis in normal, non-food-restricted animals. Wild-type mice are treated with high-dose leptin until fat mass is depleted and, as a consequence, endogenous leptin production is reduced. At this point, exogenous leptin is abruptly withdrawn, thus inducing a state of leptin deficiency in otherwise normal mice. Leptin deficiency is sustained by feeding the animals only as much as they consumed voluntarily before leptin withdrawal. The biologic response to leptin deficiency induced in this manner includes altered neuropeptide levels, decreased energy expenditure, and impaired reproductive and immune function. Replacement of leptin at physiological concentrations after withdrawal of high-dosage leptin blunts, but does not completely block, the hyperphagia and weight regain caused by acute leptin deficiency, nor does it correct the resulting reproductive and immune dysfunction. This suggests that high-dosage leptin treatment induces a state of partial leptin resistance. In aggregate, these studies establish the role of acute hypoleptinemia in regulating energy balance, the immune system, and reproductive function, and further suggest that high-dosage leptin treatment can induce a state of acquired leptin resistance.


Asunto(s)
Leptina/deficiencia , Animales , Resistencia a Medicamentos , Ingestión de Alimentos/fisiología , Metabolismo Energético , Femenino , Inmunidad , Leptina/administración & dosificación , Leptina/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Sistemas Neurosecretores/fisiología
19.
Proc Natl Acad Sci U S A ; 100(18): 10540-5, 2003 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-12925742

RESUMEN

Gold-thioglucose (GTG) induces lesions in the ventromedial nucleus of the hypothalamus, resulting in hyperphagia and obesity. To identify genes involved in the hypothalamic regulation of energy homeostasis, we used a screen for genes that are dysregulated in GTG-induced obese mice. We found that GPR7, the endogenous G protein-coupled receptor for the recently identified ligands neuropeptide B and neuropeptide W, was down-regulated in hypothalamus after GTG treatment. Here we show that male GPR7-/- mice develop an adult-onset obese phenotype that progressively worsens with age and was greatly exacerbated when animals are fed a high-fat diet. GPR7-/- male mice were hyperphagic and had decreased energy expenditure and locomotor activity. Plasma levels of glucose, leptin, and insulin were also elevated in these mice. GPR7-/- male mice had decreased hypothalamic neuropeptide Y RNA levels and increased proopiomelanocortin RNA levels, a set of effects opposite to those evident in ob/ob mice. Furthermore, ob/ob GPR7-/- and Ay/a GPR7-/- double mutant male mice had an increased body weight compared with normal ob/ob or Ay/a male mice, suggesting that the obesity of GPR7-/- mice is independent of leptin and melanocortin signaling. Female mice did not show any significant weight increase or associated metabolic defects. These data suggest a potential role for GPR7 and its endogenous ligands, neuropeptide B and neuropeptide W, in regulating energy homeostasis independent of leptin and melanocortin signaling in a sexually dimorphic manner.


Asunto(s)
Metabolismo Energético , Obesidad/etiología , Receptores de Neuropéptido/fisiología , Animales , Composición Corporal , Peso Corporal , Ingestión de Alimentos , Femenino , Hipotálamo/química , Masculino , Ratones , Ratones Endogámicos CBA , Actividad Motora , Receptores Acoplados a Proteínas G , Receptores de Neuropéptido/análisis
20.
J Neurosci ; 23(18): 7143-54, 2003 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-12904474

RESUMEN

The melanocortin-4 receptor (MC4-R) is an important regulator of energy homeostasis, and evidence suggests that MC4-R-expressing neurons are downstream targets of leptin action. MC4-Rs are broadly expressed in the CNS, and the distribution of MC4-R mRNA has been analyzed most extensively in the rat. However, relatively little is known concerning chemical profiles of MC4-R-expressing neurons. The extent to which central melanocortins act presynaptically or postsynaptically on MC4-Rs is also unknown. To address these issues, we have generated a transgenic mouse line expressing green fluorescent protein (GFP) under the control of the MC4-R promoter, using a modified bacterial artificial chromosome. We have confirmed that the CNS distribution of GFP-producing cells is identical to that of MC4-R mRNA in wild-type mice and that nearly all GFP-producing cells coexpress MC4-R mRNA. For example, cells coexpressing GFP and MC4-R mRNA were distributed in the paraventricular hypothalamic nucleus (PVH) and the dorsal motor nucleus of the vagus (DMV). MC4-R promotor-driven GFP expression was found in PVH cells producing thyrotropin-releasing hormone and in cholinergic DMV cells. Finally, we have observed that a synthetic MC3/4-R agonist, MT-II, depolarizes some GFP-expressing cells, suggesting that MC4-Rs function postsynaptically in some instances and may function presynaptically in others. These studies extend our knowledge of the distribution and function of the MC4-R. The transgenic mouse line should be useful for future studies on the role of melanocortin signaling in regulating feeding behavior and autonomic homeostasis.


Asunto(s)
Expresión Génica/fisiología , Proteínas Luminiscentes/biosíntesis , Regiones Promotoras Genéticas/fisiología , Receptores de Corticotropina/genética , Animales , Encéfalo/anatomía & histología , Encéfalo/metabolismo , Encéfalo/fisiología , Cromosomas Artificiales Bacterianos , Proteínas Fluorescentes Verdes , Hipotálamo/citología , Hipotálamo/fisiología , Técnicas In Vitro , Ligandos , Proteínas Luminiscentes/genética , Bulbo Raquídeo/metabolismo , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Neuronas/fisiología , Núcleo Hipotalámico Paraventricular/metabolismo , Técnicas de Placa-Clamp , ARN Mensajero/biosíntesis , Receptor de Melanocortina Tipo 4 , Receptores de Corticotropina/biosíntesis , Nervio Vago/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA