Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Physiol ; 594(4): 1069-85, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26613645

RESUMEN

KEY POINTS: The basal forebrain is an important component of the ascending arousal system and may be a key site through which the orexin neurons promote arousal. It has long been known that orexin-A and -B excite basal forebrain cholinergic neurons, but orexin-producing neurons also make the inhibitory peptide dynorphin. Using whole-cell recordings in brain slices, we found that dynorphin-A directly inhibits basal forebrain cholinergic neurons via κ-opioid receptors, and decreases afferent excitatory synaptic input to these neurons. While the effects of dynorphin-A and orexin-A desensitize over multiple applications, co-application of dynorphin-A and orexin-A produces a sustained response that reverses depending on the membrane potential of basal forebrain cholinergic neurons. At -40 mV the net effect of the co-application is inhibition by dynorphin-A, whereas at -70 mV the excitatory response to orexin-A prevails. ABSTRACT: The basal forebrain (BF) is an essential component of the ascending arousal systems and may be a key site through which the orexin (also known as hypocretin) neurons drive arousal and promote the maintenance of normal wakefulness. All orexin neurons also make dynorphin, and nearly all brain regions innervated by the orexin neurons express kappa opiate receptors, the main receptor for dynorphin. This is remarkable because orexin excites target neurons including BF neurons, but dynorphin has inhibitory effects. We identified the sources of dynorphin input to the magnocellular preoptic nucleus and substantia innominata (MCPO/SI) in mice and determined the effects of dynorphin-A on MCPO/SI cholinergic neurons using patch-clamp recordings in brain slices. We found that the orexin neurons are the main source of dynorphin input to the MCPO/SI region, and dynorphin-A inhibits MCPO/SI cholinergic neurons through κ-opioid receptors by (1) activation of a G protein-coupled inwardly rectifying potassium current, (2) inhibition of a voltage-gated Ca(2+) current and (3) presynaptic depression of the glutamatergic input to these neurons. The responses both to dynorphin-A and to orexin-A desensitize, but co-application of dynorphin-A and orexin-A produces a sustained response. In addition, the polarity of the response to the co-application depends on the membrane potential of BF neurons; at -40 mV the net effect of the co-application is inhibition by dynorphin-A, whereas at -70 mV the excitatory response to orexin-A prevails. This suggests that depending on their state of activation, BF cholinergic neurons can be excited or inhibited by signals from the orexin neurons.


Asunto(s)
Neuronas Colinérgicas/metabolismo , Dinorfinas/metabolismo , Área Preóptica/metabolismo , Sustancia Innominada/metabolismo , Sinapsis/metabolismo , Animales , Canales de Calcio/metabolismo , Neuronas Colinérgicas/fisiología , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Ratones , Ratones Endogámicos C57BL , Orexinas/metabolismo , Área Preóptica/citología , Área Preóptica/fisiología , Receptores Opioides/metabolismo , Sustancia Innominada/citología , Sustancia Innominada/fisiología , Sinapsis/fisiología , Potenciales Sinápticos
2.
Nat Med ; 1(12): 1311-4, 1995 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-7489415

RESUMEN

The regulation of body weight and composition involves input from genes and the environment, demonstrated, for example, by the variable susceptibility of inbred strains of mice to obesity when offered a high-fat diet. The identification of the gene responsible for obesity in the ob/ob mouse provides a new approach to defining links between diet and genetics in the regulation of body weight. The ob gene protein product, leptin, is an adipocyte-derived circulating protein. Administration of recombinant leptin reduces food intake and increases energy expenditure in ob/ob mice, suggesting that it signals to the brain the magnitude of fat stores. Information on the regulation of this protein is limited. In several rodent models of obesity including db/db, fa/fa, yellow (Ay/a) VMH-lesioned, and those induced by gold thioglucose, monosodium glutamate, and transgenic ablation of brown adipose tissue, leptin mRNA expression and the level of circulating leptin are increased, suggesting resistance to one or more of its actions. We have assessed the impact of increased dietary fat on circulating leptin levels in normal FVB mice and FVB mice with transgene-induced ablation of brown adipose tissue. We find that high-fat diet evokes a sustained increase in circulating leptin in both normal and transgenic mice, with leptin levels accurately reflecting the amount of body lipid across a broad range of body fat. However, despite increased leptin levels, animals fed a high-fat diet became obese without decreasing their caloric intake, suggesting that a high content of dietary fat changes the 'set point' for body weight, at least in part by limiting the action of leptin.


Asunto(s)
Grasas de la Dieta/metabolismo , Metabolismo de los Lípidos , Obesidad/metabolismo , Proteínas/metabolismo , Secuencia de Aminoácidos , Animales , Peso Corporal , Femenino , Leptina , Masculino , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular
3.
Nat Med ; 6(8): 924-8, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10932232

RESUMEN

The prevalence of type 2 diabetes mellitus is growing worldwide. By the year 2020, 250 million people will be afflicted. Most forms of type 2 diabetes are polygenic with complex inheritance patterns, and penetrance is strongly influenced by environmental factors. The specific genes involved are not yet known, but impaired glucose uptake in skeletal muscle is an early, genetically determined defect that is present in non-diabetic relatives of diabetic subjects. The rate-limiting step in muscle glucose use is the transmembrane transport of glucose mediated by glucose transporter (GLUT) 4 (ref. 4), which is expressed mainly in skeletal muscle, heart and adipose tissue. GLUT4 mediates glucose transport stimulated by insulin and contraction/exercise. The importance of GLUT4 and glucose uptake in muscle, however, was challenged by two recent observations. Whereas heterozygous GLUT4 knockout mice show moderate glucose intolerance, homozygous whole-body GLUT4 knockout (GLUT4-null) mice have only mild perturbations in glucose homeostasis and have growth retardation, depletion of fat stores, cardiac hypertrophy and failure, and a shortened life span. Moreover, muscle-specific inactivation of the insulin receptor results in minimal, if any, change in glucose tolerance. To determine the importance of glucose uptake into muscle for glucose homeostasis, we disrupted GLUT4 selectively in mouse muscles. A profound reduction in basal glucose transport and near-absence of stimulation by insulin or contraction resulted. These mice showed severe insulin resistance and glucose intolerance from an early age. Thus, GLUT4-mediated glucose transport in muscle is essential to the maintenance of normal glucose homeostasis.


Asunto(s)
Resistencia a la Insulina/fisiología , Proteínas de Transporte de Monosacáridos/deficiencia , Proteínas de Transporte de Monosacáridos/genética , Proteínas Musculares , Músculo Esquelético/metabolismo , Animales , Secuencia de Bases , Transporte Biológico Activo/efectos de los fármacos , Cartilla de ADN/genética , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Transportador de Glucosa de Tipo 4 , Humanos , Técnicas In Vitro , Insulina/farmacología , Resistencia a la Insulina/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Transporte de Monosacáridos/metabolismo , Contracción Muscular/fisiología , Músculo Esquelético/efectos de los fármacos
4.
J Exp Med ; 185(1): 171-5, 1997 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-8996253

RESUMEN

Several inflammatory cytokines, most notably tumor necrosis factor (TNF) and IL-1, induce anorexia and loss of lean body mass, common manifestations of acute and chronic inflammatory conditions. In C57BL/6 female mice, the administration of TNF, IL-1, and, to a lesser extent, leukemia inhibitory factor (LIF), produced a prompt and dose-dependent increase in serum leptin levels and leptin mRNA expression in fat. IL-10, IL-4, ciliary neurotrophic factor, and IL-2, cytokines not known to induce anorexia or decrease food intake, had no effect on leptin gene expression or serum leptin levels. After administration of Escherichia coli lipopolysaccharide (LPS), leptin gene expression and leptin levels were increased. These findings suggest that leptin levels may be one mechanism by which anorexia is induced during acute inflammatory conditions.


Asunto(s)
Tejido Adiposo/metabolismo , Anorexia , Citocinas/farmacología , Inflamación , Interleucina-6 , Biosíntesis de Proteínas , Transcripción Genética/efectos de los fármacos , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/inmunología , Animales , Factor Neurotrófico Ciliar , Escherichia coli , Femenino , Inhibidores de Crecimiento/farmacología , Humanos , Interleucina-10/farmacología , Interleucina-2/farmacología , Interleucina-4/farmacología , Interleucinas/farmacología , Cinética , Leptina , Factor Inhibidor de Leucemia , Lipopolisacáridos/farmacología , Linfocinas/farmacología , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/farmacología , Proteínas/análisis , ARN Mensajero/biosíntesis , Proteínas Recombinantes/farmacología , Factor de Necrosis Tumoral alfa/farmacología
6.
Curr Biol ; 8(15): R517-20, 1998 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-9705924

RESUMEN

Brown adipocytes play important roles in the regulation of fat storage and body temperature, by virtue of their ability to uncouple mitochondrial fuel oxidation and ATP synthesis. The discovery of a tissue-specific transcriptional coactivator provides new insights into the regulation of thermogenesis by brown adipocytes.


Asunto(s)
Adaptación Fisiológica/fisiología , Regulación de la Temperatura Corporal/fisiología , Adipocitos , Tejido Adiposo Pardo/citología , Tejido Adiposo Pardo/fisiología , Animales , Proteínas Portadoras/genética , AMP Cíclico/metabolismo , Canales Iónicos , Proteínas de la Membrana/genética , Proteínas Mitocondriales , Receptores Citoplasmáticos y Nucleares/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología , Desacopladores , Proteína Desacopladora 1
7.
J Clin Invest ; 96(3): 1658-63, 1995 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-7657836

RESUMEN

The mutant gene responsible for obesity in the ob/ob mouse was recently identified by positional cloning (Zhang Y., R. Proenca, M. Maffel, M. Barone, L. Leopold, and J.M. Friedman. 1994. Nature (Lond.) 372:425). The encoded protein and to represent and "adipostat" signal reflecting the state of energy stores. We confirm that the adipocyte is the source of ob mRNA and that the predicted 16-kD ob protein is present in rodent serum as detected by Western blot. To evaluate the hypothesis that it might represent an adipostat, we assessed serum levels of ob protein and expression of ob mRNA in adipose cells and tissue of rodents in response to a variety of perturbations which effect body fat mass. Both ob protein and ob mRNA expression are markedly increased in obesity. The levels of ob protein are approximately 5-10-fold elevated in serum of db/db mice, in mice with hypothalamic lesions caused by neonatal administration of monosodium glutamate (MSG), and in mice with toxigene induced brown fat ablation, (UCP-DTA). Very parallel changes are observed in adipocyte ob mRNA expression in these models and in ob/ob mice. As predicted however, no serum ob protein could be detected in the ob/ob mice. By contrast to obesity, starvation of normal rats and mice for 1-3 d markedly suppresses ob mRNA abundance, and this is reversed with refeeding. Similarly, ob protein concentration in normal mice falls to undetectable levels with starvation. In the ob/ob, UCP-DTA and MSG models, overexpression of ob mRNA is reversed by caloric restriction. These data support the hypothesis that expression of ob mRNA and protein are regulated as a function of energy stores, and that ob serves as a circulating feedback signal to sites involved in regulation of energy homeostasis.


Asunto(s)
Tejido Adiposo/metabolismo , Fenómenos Fisiológicos Nutricionales de los Animales , Expresión Génica , Obesidad/genética , Obesidad/fisiopatología , Biosíntesis de Proteínas , Adipocitos/metabolismo , Secuencia de Aminoácidos , Animales , Anticuerpos , Secuencia de Bases , Western Blotting , Cartilla de ADN , Leptina , Masculino , Ratones , Ratones Obesos , Datos de Secuencia Molecular , Péptidos/química , Péptidos/inmunología , Reacción en Cadena de la Polimerasa , ARN Mensajero/biosíntesis , Conejos/inmunología , Ratas , Ratas Sprague-Dawley
8.
J Clin Invest ; 97(11): 2553-61, 1996 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-8647948

RESUMEN

The orphan nuclear receptor, peroxisome proliferator-activated receptor (PPAR) gamma, is implicated in mediating expression of fat-specific genes and in activating the program of adipocyte differentiation. The potential for regulation of PPAR gamma gene expression in vivo is unknown. We cloned a partial mouse PPAR gamma cDNA and developed an RNase protection assay that permits simultaneous quantitation of mRNAs for both gamma l and gamma 2 isoforms encoded by the PPAR gamma gene. Probes for detection of adipocyte P2, the obese gene product, leptin, and 18S mRNAs were also employed. Both gamma l and gamma 2 mRNAs were abundantly expressed in adipose tissue. PPAR gamma 1 expression was also detected at lower levels in liver, spleen, and heart; whereas, gamma l and gamma 2 mRNA were expressed at low levels in skeletal muscle. Adipose tissue levels of gamma l and gamma 2 were not altered in two murine models of obesity (gold thioglucose and ob/ob), but were modestly increased in mice with toxigene-induced brown fat ablation uncoupling protein diphtheria toxin A mice. Fasting (12-48 h) was associated with an 80% fall in PPAR gamma 2 and a 50% fall in PPAR gamma mRNA levels in adipose tissue. Western blot analysis demonstrated a marked effect of fasting to reduce PPAR gamma protein levels in adipose tissue. Similar effects of fasting on PPAR gamma mRNAs were noted in all three models of obesity. Insulin-deficient (streptozotocin) diabetes suppressed adipose tissue gamma l and gamma 2 expression by 75% in normal mice with partial restoration during insulin treatment. Levels of adipose tissue PPAR gamma 2 mRNA were increased by 50% in normal mice exposed to a high fat diet. In obese uncoupling protein diphtheria toxin A mice, high fat feeding resulted in de novo induction of PPAR gamma 2 expression in liver. We conclude (a) PPAR gamma 2 mRNA expression is most abundant in adipocytes in normal mice, but lower level expression is seen in skeletal muscle; (b) expression of adipose tissue gamma1 or gamma2 mRNAs is increased in only one of the three models of obesity; (c) PPAR gamma 1 and gamma 2 expression is downregulated by fasting and insulin-deficient diabetes; and (d) exposure of mice to a high fat diet increases adipose tissue expression of PPAR gamma (in normal mice) and induces PPAR gamma 2 mRNA expression in liver (in obese mice). These findings demonstrate in vivo modulation of PPAR gamma mRNA levels over a fourfold range and provide an additional level of regulation for the control of adipocyte development and function.


Asunto(s)
Grasas de la Dieta , Regulación de la Expresión Génica , Obesidad/metabolismo , Receptores Citoplasmáticos y Nucleares/biosíntesis , Factores de Transcripción/biosíntesis , Transcripción Genética , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/fisiología , Animales , Aurotioglucosa/farmacología , Secuencia de Bases , Cartilla de ADN , Diabetes Mellitus Experimental/metabolismo , Toxina Diftérica/toxicidad , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Leptina , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Datos de Secuencia Molecular , Músculo Esquelético/metabolismo , Miocardio/metabolismo , Obesidad/genética , Especificidad de Órganos , Reacción en Cadena de la Polimerasa , Biosíntesis de Proteínas , Proteínas/genética , ARN Mensajero/biosíntesis , ARN Ribosómico 16S/biosíntesis , Valores de Referencia , Bazo/metabolismo , Transcripción Genética/efectos de los fármacos
9.
J Clin Invest ; 101(1): 1-9, 1998 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-9421459

RESUMEN

The ability to regulate specific genes of energy metabolism in response to fasting and feeding is an important adaptation allowing survival of intermittent food supplies. However, little is known about transcription factors involved in such responses in higher organisms. We show here that gene expression in adipose tissue for adipocyte determination differentiation dependent factor (ADD) 1/sterol regulatory element binding protein (SREBP) 1, a basic-helix-loop-helix protein that has a dual DNA-binding specificity, is reduced dramatically upon fasting and elevated upon refeeding; this parallels closely the regulation of two adipose cell genes that are crucial in energy homeostasis, fatty acid synthetase (FAS) and leptin. This elevation of ADD1/SREBP1, leptin, and FAS that is induced by feeding in vivo is mimicked by exposure of cultured adipocytes to insulin, the classic hormone of the fed state. We also show that the promoters for both leptin and FAS are transactivated by ADD1/SREBP1. A mutation in the basic domain of ADD1/SREBP1 that allows E-box binding but destroys sterol regulatory element-1 binding prevents leptin gene transactivation but has no effect on the increase in FAS promoter function. Molecular dissection of the FAS promoter shows that most if not all of this action of ADD1/SREBP1 is through an E-box motif at -64 to -59, contained with a sequence identified previously as the major insulin response element of this gene. These results indicate that ADD1/SREBP1 is a key transcription factor linking changes in nutritional status and insulin levels to the expression of certain genes that regulate systemic energy metabolism.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT , Proteínas de Unión al ADN/metabolismo , Ácido Graso Sintasas/genética , Secuencias Hélice-Asa-Hélice , Proteínas Nucleares/metabolismo , Proteínas/genética , Factores de Transcripción , Células 3T3 , Animales , Línea Celular , Proteínas de Unión al ADN/genética , Ingestión de Alimentos , Ácido Graso Sintasas/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Insulina/farmacología , Leptina , Ratones , Ratones Endogámicos C57BL , Proteínas Nucleares/genética , Proteínas/metabolismo , Ratas , Proteína 1 de Unión a los Elementos Reguladores de Esteroles , Activación Transcripcional
10.
J Clin Invest ; 104(12): 1703-14, 1999 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-10606624

RESUMEN

Glucose enters the heart via GLUT1 and GLUT4 glucose transporters. GLUT4-deficient mice develop striking cardiac hypertrophy and die prematurely. Whether their cardiac changes are caused primarily by GLUT4 deficiency in cardiomyocytes or by metabolic changes resulting from the absence of GLUT4 in skeletal muscle and adipose tissue is unclear. To determine the role of GLUT4 in the heart we used cre-loxP recombination to generate G4H(-/-) mice in which GLUT4 expression is abolished in the heart but is present in skeletal muscle and adipose tissue. Life span and serum concentrations of insulin, glucose, FFAs, lactate, and beta-hydroxybutyrate were normal. Basal cardiac glucose transport and GLUT1 expression were both increased approximately 3-fold in G4H(-/-) mice, but insulin-stimulated glucose uptake was abolished. G4H(-/-) mice develop modest cardiac hypertrophy associated with increased myocyte size and induction of atrial natriuretic and brain natriuretic peptide gene expression in the ventricles. Myocardial fibrosis did not occur. Basal and isoproterenol-stimulated isovolumic contractile performance was preserved. Thus, selective ablation of GLUT4 in the heart initiates a series of events that results in compensated cardiac hypertrophy.


Asunto(s)
Cardiomegalia/etiología , Proteínas de Transporte de Monosacáridos/fisiología , Proteínas Musculares , Contracción Miocárdica , Animales , Factor Natriurético Atrial/genética , Cardiomegalia/fisiopatología , Femenino , Glucosa/metabolismo , Transportador de Glucosa de Tipo 1 , Transportador de Glucosa de Tipo 4 , Masculino , Ratones , Ratones Transgénicos , Proteínas de Transporte de Monosacáridos/genética , Miocardio/metabolismo , Péptido Natriurético Encefálico/genética , Tamaño de los Órganos
11.
Diabetes ; 36(1): 14-9, 1987 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-3792662

RESUMEN

Previous studies indicated that protein sparing in skeletal muscle during prolonged starvation depends on the availability of lipid fuels. To test this relationship further, fasted rats conserving protein were treated in vivo for 6-8 h with the antilipolytic agent nicotinic acid (NA) or with tetradecylglycidate (TDGD), an inhibitor of long-chain fatty acid oxidation. After treatment, protein synthesis and degradation in skeletal muscle were evaluated with the perfused rat hindquarter. NA treatment decreased plasma 3-hydroxybutyrate and free fatty acids and increased plasma urea and urine urea excretion, indicating increased breakdown of body protein. TDGD produced similar metabolic effects, except that plasma free fatty acids were markedly increased as a result of inhibition of fatty acid oxidation. NA and TDGD also decreased plasma insulin and increased plasma corticosteroid. Inhibition of lipid metabolism in vivo resulted in accelerated loss of protein from skeletal muscle due to decreased protein synthesis and increased protein breakdown. NA increased both total (i.e., tyrosine release) and myofibrillar (i.e., 3-methylhistidine release) protein breakdown, whereas TDGD increased the breakdown of only nonmyofibrillar proteins (i.e., 3-methylhistidine release by perfused hindquarter was not altered). These data indicate that lipid fuels may directly modulate protein metabolism in muscle during prolonged starvation and may prevent a rise in catabolic hormones. They also indicate that free fatty acids may directly attenuate the breakdown of myofibrillar proteins in muscle during prolonged starvation and that this may be unrelated to their oxidation.


Asunto(s)
Metabolismo de los Lípidos , Proteínas Musculares/metabolismo , Músculos/metabolismo , Inanición/metabolismo , Ácido 3-Hidroxibutírico , Animales , Nitrógeno de la Urea Sanguínea , Compuestos Epoxi/farmacología , Ácidos Grasos/farmacología , Hidroxibutiratos/sangre , Masculino , Proteínas Musculares/biosíntesis , Niacina/farmacología , Ratas , Ratas Endogámicas , Tioglicolatos/farmacología
12.
Diabetes ; 49(2): 143-56, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10868929

RESUMEN

Mitochondria use energy derived from fuel combustion to create a proton electrochemical gradient across the mitochondrial inner membrane. This intermediate form of energy is then used by ATP synthase to synthesize ATP. Uncoupling protein-1 (UCP1) is a brown fat-specific mitochondrial inner membrane protein with proton transport activity. UCP1 catalyzes a highly regulated proton leak, converting energy stored within the mitochondrial proton electrochemical potential gradient to heat. This uncouples fuel oxidation from conversion of ADP to ATP. In rodents, UCP1 activity and brown fat contribute importantly to whole-body energy expenditure. Recently, two additional mitochondrial carriers with high similarity to UCP1 were molecularly cloned. In contrast to UCP1, UCP2 is expressed widely, and UCP3 is expressed preferentially in skeletal muscle. Biochemical studies indicate that UCP2 and UCP3, like UCP1, have uncoupling activity. While UCP1 is known to play an important role in regulating heat production during cold exposure, the biological functions of UCP2 and UCP3 are unknown. Possible functions include 1) control of adaptive thermogenesis in response to cold exposure and diet, 2) control of reactive oxygen species production by mitochondria, 3) regulation of ATP synthesis, and 4) regulation of fatty acid oxidation. This article will survey present knowledge regarding UCP1, UCP2, and UCP3, and review proposed functions for the two new uncoupling proteins.


Asunto(s)
Proteínas Portadoras/fisiología , Metabolismo Energético/fisiología , Proteínas de Transporte de Membrana , Mitocondrias/metabolismo , Proteínas Mitocondriales , Proteínas/fisiología , Animales , Proteínas Portadoras/genética , Expresión Génica , Ligamiento Genético , Variación Genética , Humanos , Canales Iónicos , Modelos Biológicos , Proteínas/genética , Proteína Desacopladora 2 , Proteína Desacopladora 3
13.
Diabetes ; 44(11): 1266-73, 1995 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-7589822

RESUMEN

We recently created a new model of murine obesity through transgenic ablation of brown adipose tissue (BAT) using a tissue-specific toxigene (6). The goal of the present study was to further define the altered glucose homeostasis and insulin resistance in these transgenic animals. Despite an approximately 30% increase in total body lipid, no abnormalities were observed in 6-week-old transgenic animals. At the age of 22-26 weeks, marked obesity in transgenic mice was associated with significant increases in blood glucose and plasma insulin levels and an abnormal response to both intraperitoneal glucose and insulin tolerance tests. Glucose transport in soleus muscle was reduced, with the response to insulin stimulation blunted by up to 85% in males and 55% in females. The total number of insulin receptors was decreased by 36% in muscle and 59% in adipose tissue of transgenic animals. Insulin receptor tyrosine kinase activity, which was assessed following maximal insulin stimulation in vivo, was reduced in transgenic animals by 59% in muscle and 56% in fat. GLUT4 mRNA and protein was unchanged in muscle of transgenic animals compared with in that of controls but was significantly reduced in adipose tissue. In conclusion, primary BAT deficiency results in the development of glucose intolerance or diabetes and severe insulin resistance with both receptor and postreceptor components. These animals should be a useful model for studies of obesity-linked diabetes and insulin resistance and related complications.


Asunto(s)
Tejido Adiposo Pardo/anatomía & histología , Diabetes Mellitus Tipo 2/fisiopatología , Diabetes Mellitus/fisiopatología , Resistencia a la Insulina , Proteínas Musculares , Obesidad , Tejido Adiposo/metabolismo , Envejecimiento , Animales , Transporte Biológico/efectos de los fármacos , Glucemia/metabolismo , Membrana Celular/metabolismo , Desoxiglucosa/metabolismo , Diabetes Mellitus/sangre , Diabetes Mellitus/genética , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/genética , Toxina Diftérica/biosíntesis , Femenino , Expresión Génica , Prueba de Tolerancia a la Glucosa , Transportador de Glucosa de Tipo 4 , Insulina/sangre , Insulina/farmacología , Resistencia a la Insulina/genética , Masculino , Ratones , Ratones Transgénicos , Proteínas de Transporte de Monosacáridos/biosíntesis , Músculo Esquelético/metabolismo , ARN Mensajero/análisis , ARN Mensajero/biosíntesis , Receptor de Insulina/metabolismo , Valores de Referencia , Caracteres Sexuales , Factor de Necrosis Tumoral alfa/biosíntesis
14.
Diabetes ; 47(2): 230-8, 1998 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9519718

RESUMEN

Brown adipose tissue (BAT) has the capacity for uncoupled mitochondrial respiration and is proposed to be a key site for regulating energy expenditure in rodents. To better define the role of BAT in energy homeostasis, we previously created a line of transgenic mice with deficiency of BAT (UCP promoter-driven diphtheria toxin A transgenic mice [UCP-DTA]) mice. These mice develop obesity that initially is due to decreased energy expenditure and later accompanied by hyperphagia despite increased levels of circulating leptin. In addition, the obesity of these mice is accompanied by severe insulin-resistant diabetes and hyperlipidemia. To better define the basis for leptin resistance in this model, we treated UCP-DTA mice with leptin (300 microg i.p., b.i.d.) and compared their response with that of leptin-treated ob/ob and FVB control mice (30 microg i.p., b.i.d.). Leptin treatment of FVB and ob/ob mice decreased their body weight and food intake and improved their glucose homeostasis. In contrast, tenfold higher dosages of leptin had no effect on body weight, food intake, or circulating insulin or glucose concentrations of UCP-DTA mice. Hypothalamic neuropeptide Y (NPY) mRNA expression was lower in UCP-DTA mice than in littermate control FVB mice in the fed state, and increased progressively in response to food restriction as leptin levels fell. In parallel to the levels of hypothalamic NPY, corticosterone levels were initially suppressed and rose with food restriction. Thus food intake, body weight, and insulin and glucose homeostasis of UCP-DTA mice are all extraordinarily resistant to leptin, whereas hypothalamic NPY and the hypothalamopituitary adrenal (HPA) axis may remain under leptin control. Further elucidation of the mechanisms underlying leptin resistance in UCP-DTA mice may provide valuable insights into the basis for leptin resistance in human obesity.


Asunto(s)
Tejido Adiposo Pardo/fisiología , Proteínas Portadoras/genética , Toxina Diftérica/genética , Hipotálamo/metabolismo , Proteínas de la Membrana/genética , Neuropéptido Y/metabolismo , Proteínas/farmacología , Animales , Glucemia/metabolismo , Corticosterona/sangre , Resistencia a Medicamentos , Metabolismo Energético , Femenino , Hipotálamo/efectos de los fármacos , Insulina/sangre , Canales Iónicos , Leptina , Masculino , Ratones , Ratones Transgénicos , Proteínas Mitocondriales , Neuropéptido Y/genética , Obesidad/genética , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Proteína Desacopladora 1
15.
Diabetes ; 47(9): 1464-71, 1998 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9726236

RESUMEN

Beta-adrenergic receptors (ARs) are expressed predominantly in adipose tissue, and beta3-selective agonists are effective anti-obesity drugs in rodents. Rodent and human beta3-ARs differ with respect to expression in white versus brown adipocytes as well as their ability to be stimulated by beta3-AR-selective agonists. Humans express beta3-AR mRNA abundantly in brown but not white adipocytes, while rodents express beta3-AR mRNA abundantly in both sites. To determine the basis for this difference, we have transgenically introduced 74 kilobases (kb) of human beta3-AR genomic sequence into gene knockout mice lacking beta3-ARs. Importantly, human beta3-AR mRNA was expressed only in brown adipose tissue (BAT) of transgenic mice, with little or no expression being detected in white adipose tissue (WAT), liver, stomach, small intestine, skeletal muscle, and heart. This pattern of expression differed from that observed in mice bearing a murine beta3-AR genomic transgene in which beta3-AR mRNA was expressed in both WAT and BAT, but not in other sites. Furthermore, we have transgenically introduced smaller human constructs containing -14.5 and -0.6 kb of upstream sequence into beta3-AR gene knockout mice. Both -14.5 and -0.6 kb constructs were expressed in BAT but not WAT. Thus, human but not murine cis-regulatory elements direct beta3-AR gene expression preferentially to brown adipocytes. Identification of responsible cis-regulatory element(s) and relevant trans-acting factor(s) should provide insight into mechanisms controlling human beta3-AR gene expression. In addition, the beta3-AR agonist, CGP-12177, stimulated oxygen consumption in mice expressing human but not murine beta3-ARs by 91% compared with only 49% in control beta3-AR gene knockout mice, demonstrating that the human beta3-AR can functionally couple with energy expenditure. These "humanized" mice should assist us in the development of drugs that may become effective anti-obesity agents in humans.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Tejido Adiposo/metabolismo , Receptores Adrenérgicos beta/genética , Secuencias Reguladoras de Ácidos Nucleicos , Antagonistas Adrenérgicos beta/farmacología , Animales , Células CHO , Línea Celular , Cricetinae , Regulación de la Expresión Génica , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Especificidad de Órganos , Consumo de Oxígeno/efectos de los fármacos , Propanolaminas/farmacología , ARN Mensajero/biosíntesis , Receptores Adrenérgicos beta/biosíntesis , Receptores Adrenérgicos beta/fisiología , Receptores Adrenérgicos beta 3 , Proteínas Recombinantes/biosíntesis , Transcripción Genética , Transfección
16.
Diabetes ; 45(7): 909-14, 1996 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-8666142

RESUMEN

To examine potential interactions between leptin and the beta3 adrenergic system in the regulation of food intake, we determined the effects of treatment with a selective beta3 adrenergic receptor (AR) agonist (CL 316,243 [1 mg/kg]) on body weight, food intake, and leptin expression. Studies were carried out in C57Bl/6J and FVB male control mice as well as in mice with targeted disruption of the beta3 AR gene. These findings were correlated with measurement of the expression in hypothalamus of neuropeptide Y (NPY) and melanin concentrating hormone (MCH), two neuropeptides that may be involved in the central regulation of food intake. Treatment with CL 316,243 (1 mg/kg) for 12 or 24 h decreased leptin mRNA abundance and circulating levels to 20% of baseline in normal animals. No effect of the CL 316,243 compound was seen in mice with targeted disruption of the beta3 AR gene. Despite the failing leptin levels, beta3 agonist administration acutely suppressed food intake. Finally, the induced suppression of food intake and leptin levels occurred despite unchanged or increased hypothalamic expression of the orexigenic neuropeptides NPY and MCH. Thus, beta3 AR agonists via beta3 ARs suppress leptin levels acutely and simultaneously suppress food intake via a mechanism that operates downstream of leptin and two of its putative central targets.


Asunto(s)
Antagonistas Adrenérgicos beta/farmacología , Dioxoles/farmacología , Conducta Alimentaria/fisiología , Expresión Génica/efectos de los fármacos , Biosíntesis de Proteínas , Receptores Adrenérgicos beta/fisiología , Tejido Adiposo/metabolismo , Secuencia de Aminoácidos , Animales , Anticuerpos , Ayuno , Conducta Alimentaria/efectos de los fármacos , Femenino , Hormonas Hipotalámicas/biosíntesis , Hipotálamo/metabolismo , Leptina , Masculino , Melaninas/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Datos de Secuencia Molecular , Neuropéptido Y/biosíntesis , Fragmentos de Péptidos/química , Fragmentos de Péptidos/inmunología , Hormonas Hipofisarias/biosíntesis , Proteínas/antagonistas & inhibidores , ARN Mensajero/biosíntesis , Receptores Adrenérgicos beta/efectos de los fármacos , Receptores Adrenérgicos beta 3 , Receptores de Leptina , Caracteres Sexuales
17.
Physiol Genomics ; 5(3): 137-45, 2001 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-11285367

RESUMEN

The cDNA of an uncoupling protein (UCP) homolog has been cloned from the swallow-tailed hummingbird, Eupetomena macroura. The hummingbird uncoupling protein (HmUCP) cDNA was amplified from pectoral muscle (flight muscle) using RT-PCR and primers for conserved domains of various known UCP homologs. The rapid amplification of cDNA ends (RACE) method was used to complete the cloning of the 5' and 3' ends of the open reading frame. The HmUCP coding region contains 915 nucleotides, and the deduced protein sequence consists of 304 amino acids, being approximately 72, 70, and 55% identical to human UCP3, UCP2, and UCP1, respectively. The uncoupling activity of this novel protein was characterized in yeast. In this expression system, the 12CA5-tagged HmUCP fusion protein was detected by Western blot in the enriched mitochondrial fraction. Similarly to rat UCP1, HmUCP decreased the mitochondrial membrane potential as measured in whole yeast by uptake of the fluorescent potential-sensitive dye 3',3-dihexyloxacarbocyanine iodide. The HmUCP mRNA is primarily expressed in skeletal muscle, but high levels can also be detected in heart and liver, as assessed by Northern blot analysis. Lowering the room's temperature to 12-14 degrees C triggered the cycle torpor/rewarming, typical of hummingbirds. Both in the pectoral muscle and heart, HmUCP mRNA levels were 1.5- to 3.4-fold higher during torpor. In conclusion, this is the first report of an UCP homolog in birds. The data indicate that HmUCP has the potential to function as an UCP and could play a thermogenic role during rewarming.


Asunto(s)
Aves/genética , Proteínas Portadoras/genética , Proteínas de la Membrana/genética , Proteínas de Transporte de Membrana , Proteínas Mitocondriales , Proteínas/genética , Secuencia de Aminoácidos , Animales , Aves/fisiología , Clonación Molecular , Canales Iónicos , Potenciales de la Membrana , Mitocondrias/fisiología , Datos de Secuencia Molecular , Filogenia , ARN Mensajero/biosíntesis , Saccharomyces cerevisiae/fisiología , Homología de Secuencia de Aminoácido , Termogénesis , Distribución Tisular , Proteína Desacopladora 1 , Proteína Desacopladora 2 , Proteína Desacopladora 3
18.
Endocrinology ; 127(6): 2898-906, 1990 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-2249632

RESUMEN

Adipsin is a serine protease with complement factor D activity that is synthesized by adipocytes and secreted into the blood stream. Expression of adipsin is deficient in models of genetic (ob/ob, db/db) and acquired (monosodium glutamate-lesioned) obesity, but the cellular mechanisms responsible for this deficiency are unknown. Because hyperinsulinemia is frequently associated with obesity, we evaluated the effects of this hormone and insulin-like growth factor 1 (IGF-1) on adipsin secretion and adipsin messenger RNA (mRNA) levels in 3T3-F442A adipocytes. In the present study, we report that in fully differentiated adipocytes (after 11 days post confluence), insulin exposure progressively decreases adipsin secretion by 40%, 67%, and 78% after 2, 4, and 6 days of treatment. The inhibition of adipsin secretion by insulin is the result of a corresponding decrease in adipsin mRNA and is specific since two other differentiation-dependent fat cell mRNAs encoding aP2 (a fatty acid binding protein) and glycerophosphate dehydrogenase (GPD), are unaffected. Insulin suppresses adipsin gene expression via high affinity insulin receptors, because physiological levels of insulin produce this effect, and dose-response curves for insulin stimulation of 2-deoxyglucose uptake and glucose utilization are similar to insulin's effect on adipsin. In contrast, insulin when present during days 1-8 post confluence (during differentiation) markedly increases adipsin secretion and adipsin mRNA levels. This stimulation is due to the ability of insulin to accelerate differentiation as evidenced by corresponding increases in aP2 and GPD mRNAs as well. Insulin and IGF-1 are equipotent in this effect, suggesting that both insulin and IGF-1 receptors can mediate this response. In summary, during the differentiation of 3T3-F442A adipocytes, insulin stimulates adipsin gene expression by accelerating differentiation. As the cells become mature adipocytes, they acquire some differentiation-dependent factor, which couples insulin receptor stimulation to inhibition of adipsin gene expression. This model should aid our search for the molecular links between insulin receptor stimulation and altered gene expression.


Asunto(s)
Tejido Adiposo/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/farmacología , Insulina/farmacología , Serina Endopeptidasas/genética , Tejido Adiposo/citología , Tejido Adiposo/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular , Factor D del Complemento , Relación Dosis-Respuesta a Droga , Cinética , Ratones , Radioinmunoensayo , Serina Endopeptidasas/análisis
19.
Endocrinology ; 137(1): 21-9, 1996 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-8536614

RESUMEN

Previous studies have indicated that rodents are relatively resistant to diet-induced obesity and that this resistance may be mediated in part by the capacity for diet-induced thermogenesis in brown adipose tissue (BAT). To test this hypothesis, we fed UCP-DTA transgenic with toxigene-mediated ablation of BAT and their control littermates a "Western diet" [21% (wt/wt) fat] or normal mouse chow [6.5% (wt/wt) fat]. The diets were begun at weaning (19 days old). At the age of 12 weeks, transgenic mice receiving the Western diet were markedly obese. The increased body weight and total body lipid content were significantly greater in transgenic mice receiving the Western diet than were the additive individual effects of Western diet (in control mice) and decreased BAT (in chow-fed mice), suggesting a synergistic interaction between diminished BAT and diet. A synergistic effect of Western diet and BAT ablation was also observed for morbid metabolic complications, such as insulin resistance, hyperglycemia, and hyperlipidemia. These metabolic changes were accompanied by increased expression of tumor necrosis factor-alpha and decreased expression of GLUT4 and beta 3-adrenergic receptor messenger RNA levels in white adipose tissue of UCP-DTA transgenic mice receiving the Western diet compared to those in the other experimental groups. As previously described, transgenic mice with diminished brown fat are hyperphagic. Of note, the degree of hyperphagia in transgenics compared to controls was similar whether the animals were fed chow or a Western diet. Thus, the synergistic effect of Western diet on obesity in transgenic mice was not mediated by a further stimulation of food intake. Overall, this study demonstrates the existence of a synergistic interaction between decreased BAT and Western diet to cause marked obesity and its accompanying disorders, such as insulin resistance and hyperlipidemia, and gives further support for the view that an important function of BAT is protection from diet-induced obesity, diabetes, and insulin resistance.


Asunto(s)
Tejido Adiposo Pardo/fisiología , Diabetes Mellitus/etiología , Grasas de la Dieta/efectos adversos , Hiperlipidemias/etiología , Obesidad/etiología , Animales , Composición Corporal , Peso Corporal , Proteínas Portadoras/metabolismo , Susceptibilidad a Enfermedades , Ingestión de Alimentos , Femenino , Canales Iónicos , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Proteínas Mitocondriales , ARN Mensajero/metabolismo , Receptores Adrenérgicos beta/genética , Desacopladores/metabolismo , Proteína Desacopladora 1
20.
Endocrinology ; 138(2): 797-804, 1997 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9003017

RESUMEN

Leptin is synthesized exclusively by adipocytes and acts on the hypothalamus to regulate energy balance. Previous messenger RNA expression studies demonstrated that leptin is expressed in white adipocytes and also in brown adipose tissue, however expression in brown fat is markedly lower than in white fat. This suggests the possibility that leptin expression in brown adipose tissue is due to the presence of white adipocytes that reside within brown adipose tissue, and that brown adipocytes actually do not express leptin. To address this point, we performed immunohistochemistry on paraffin sections and studied leptin protein expression in different depots of white and brown fat of lean and obese (db/db) mice. To establish the cell type expressing leptin, we also assessed the size and organization of lipid droplets, the ultrastructural features of mitochondria, and the presence or absence of uncoupling protein, a brown fat-specific marker. In white adipose tissue of lean and obese (db/db) mice, leptin protein was expressed in adipocytes of various sizes (range examined: 19.67-200 microns), including adipocytes at the multilocular stage of differentiation. Leptin staining was more intense in some depots (retroperitoneal), and appeared to decrease with fasting. In brown adipose tissue of lean animals, multilocular uncoupling protein (UCP)-positive brown adipocytes had typical brown mitochondria and were leptin-negative, both in fed and fasted conditions. At the periphery of the interscapular brown adipose tissue depot, unilocular, UCP-negative adipocytes (mean diameter: 41.55 microns) with white-type mitochondria were observed, and these cells were leptin-positive. In obese (db/db) animals, brown fat was composed mainly of small unilocular, UCP-positive. adipocytes (mean diameter: 40.08 microns), which were also leptin-positive. At the periphery of the organ, numerous large, unilocular, UCP-negative adipocytes (mean diameter: 73.65 microns) with white-like mitochondria were present. As expected, these cells were also leptin-positive. In summary, classical brown adipocytes differ from white adipocytes, not only by their morphology and UCP expression, but also by their apparent lack of detectable leptin expression. db/db brown adipocytes, however, were unilocular and leptin-positive. The molecular mechanisms mediating expression of leptin in white but not brown adipocytes of lean animals, and the significant expression of leptin in brown adipocytes of db/db mice will be the focus of future studies.


Asunto(s)
Tejido Adiposo Pardo/química , Tejido Adiposo/química , Proteínas Portadoras/análisis , Inmunohistoquímica , Proteínas de la Membrana/análisis , Proteínas/análisis , Adipocitos/química , Secuencia de Aminoácidos , Animales , Ayuno , Femenino , Canales Iónicos , Leptina , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Mitocondriales , Datos de Secuencia Molecular , Obesidad/metabolismo , Control de Calidad , Proteína Desacopladora 1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA