Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Ageing Res Rev ; 65: 101200, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33130247

RESUMO

Sarcopenia represents a muscle-wasting syndrome characterized by progressive and generalized degenerative loss of skeletal muscle mass, quality, and strength occurring during normal aging. Sarcopenia patients are mainly suffering from the loss in muscle strength and are faced with mobility disorders reducing their quality of life and are, therefore, at higher risk for morbidity (falls, bone fracture, metabolic diseases) and mortality. Several molecular mechanisms have been described as causes for sarcopenia that refer to very different levels of muscle physiology. These mechanisms cover e. g. function of hormones (e. g. IGF-1 and Insulin), muscle fiber composition and neuromuscular drive, myo-satellite cell potential to differentiate and proliferate, inflammatory pathways as well as intracellular mechanisms in the processes of proteostasis and mitochondrial function. In this review, we describe sarcopenia as a muscle-wasting syndrome distinct from other atrophic diseases and summarize the current view on molecular causes of sarcopenia development as well as open questions provoking further research efforts for establishing efficient lifestyle and therapeutic interventions.


Assuntos
Sarcopenia , Envelhecimento , Humanos , Músculo Esquelético/patologia , Atrofia Muscular/patologia , Qualidade de Vida , Sarcopenia/patologia
2.
Artigo em Inglês | MEDLINE | ID: mdl-27551276

RESUMO

Intact melanocortin signaling via the G protein-coupled receptors (GPCRs), melanocortin receptor 4 (MC4R), and melanocortin receptor 3 (MC3R) is crucial for body weight maintenance. So far, no connection between melanocortin signaling and hypothalamic inflammation has been reported. Using a bimolecular fluorescence complementation library screen, we identified a new interaction partner for these receptors, ring finger protein 11 (RNF11). RNF11 participates in the constitution of the A20 complex that is involved in reduction of tumor necrosis factor α (TNFα)-induced NFκB signaling, an important pathway in hypothalamic inflammation. Mice treated with high-fat diet (HFD) for 3 days demonstrated a trend toward an increase in hypothalamic Rnf11 expression, as shown for other inflammatory markers under HFD. Furthermore, Gs-mediated signaling of MC3/4R was demonstrated to be strongly reduced to 20-40% by co-expression of RNF11 despite unchanged total receptor expression. Cell surface expression was not affected for MC3R but resulted in a significant reduction of MC4R to 61% by co-expression with RNF11. Mechanisms linking HFD, inflammation, and metabolism remain partially understood. In this study, a new axis between signaling of specific body weight regulating GPCRs and factors involved in hypothalamic inflammation is suggested.

3.
J Cell Commun Signal ; 9(2): 143-50, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25663268

RESUMO

IGFBP-2 affects growth and metabolism and is thought to impact on energy homeostasis and the accretion of body fat via its heparin binding domains (HBD). In order to assess the function of the HBD present in the linker domain (HBD1) we have generated transgenic mice overexpressing mutant human IGFBP-2 lacking the PKKLRP sequence and carrying a PNNLAP sequence instead. Transgenic mice expressed high amounts of human IGFBP-2, while endogenous IGFBP-2 or IGF-I serum concentrations were not affected. In both genders we performed a longitudinal analysis of growth and metabolism including at least 4 separate time points between the age of 10 and 52 weeks. Body composition was assessed by nuclear magnetic resonance (NMR) analysis. Food intake was recorded by an automated online-monitoring. We describe negative effects of mutant human IGFBP-2 on body weight, longitudinal growth and lean body mass (p < 0.05). Very clearly, negative effects of mutant IGFBP-2 were not observed for fat mass accretion throughout life. Instead, relative fat mass was increased in transgenic mice of both genders (p < 0.05). In male mice transgene expression significantly increased absolute mass of total body fat over all age groups (p < 0.05). Food intake was increased in female but decreased in male transgenic mice at an age of 11 weeks. Thus our study clearly provides gender- and time-specific effects of HBD1-deficient hIGFBP-2 (H1d-BP-2) on fat mass accretion and food intake. While our data are in principal agreement with current knowledge on the role of HB-domains for fat accretion we now may also speculate on a role of HBD1 for the control of eating behavior.

4.
Endocrinology ; 156(1): 389-99, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25322465

RESUMO

Effective and safe antiobesity drugs are still needed in face of the obesity pandemic worldwide. Recent interventions in rodents revealed 3,5-diiodo-L-thyronine (3,5-T2) as a metabolically active iodothyronine affecting energy and lipid metabolism without thyromimetic side effects typically associated with T3 administration. Accordingly, 3,5-T2 has been proposed as a potential hypolipidemic agent for treatment of obesity and hepatic steatosis. In contrast to other observations, our experiments revealed dose-dependent thyromimetic effects of 3,5-T2 akin to those of T3 in diet-induced obese male C57BL/6J mice. 3,5-T2 treatment exerted a negative feedback regulation on the hypothalamus-pituitary-thyroid axis, similar to T3. This is demonstrated by decreased expression of genes responsive to thyroid hormones (TH) in pituitary resulting in a suppressed thyroid function with lower T4 and T3 concentrations in serum and liver of 3,5-T2-treated mice. Analyses of hepatic TH target genes involved in lipid metabolism revealed T3-like changes in gene expression and increased type I-deiodinase activity after application of 3,5-T2 (2.5 µg/g body weight). Reduced hepatic triglyceride and serum cholesterol concentrations reflected enhanced lipid metabolism. Desired increased metabolic rate and reduction of different fat depots were, however, compromised by increased food intake preventing significant body weight loss. Moreover, enlarged heart weights indicate potential cardiac side effects of 3,5-T2 beyond hepatic thyromimetic actions. Altogether, the observed thyromimetic effects of 3,5-T2 in several mouse TH target tissues raise concern about indiscriminate administration of 3,5-T2 as powerful natural hormone for the treatment of hyperlipidemia and pandemic obesity.


Assuntos
Composição Corporal/efeitos dos fármacos , Di-Iodotironinas/farmacologia , Metabolismo Energético/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Obesidade/induzido quimicamente , Glândula Tireoide/efeitos dos fármacos , Animais , Gorduras na Dieta/toxicidade , Ingestão de Alimentos/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/fisiologia , Masculino , Camundongos , Atividade Motora , Obesidade/metabolismo , Glândula Tireoide/fisiologia , Fatores de Tempo , Transcriptoma , Redução de Peso
5.
Mol Cell Endocrinol ; 381(1-2): 280-90, 2013 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-23994018

RESUMO

Signaling through the mammalian target of rapamycin complex 1 (mTORC1) and its effectors the S6-kinases (S6K) in the hypothalamus is thought to be involved in nutrient sensing and control of food intake. Given the anatomical proximity of this pathway to circuits for the hormone ghrelin, we investigated the potential role of the mTORC1/S6K pathway in mediating the metabolic effects of ghrelin. We found that ghrelin promoted phosphorylation of S6K1 in the mouse hypothalamic cell line N-41 and in the rat hypothalamus after intracerebroventricular administration. Rapamycin, an inhibitor of mTORC1, suppressed ghrelin-induced phosphorylation of hypothalamic S6K1 and increased food intake and insulin in rats. Chronic peripheral administration of ghrelin induced a significant increase in body weight, fat mass and food efficiency in wild-type and S6K2-knockout but not in S6K1-knockout mice. We therefore propose that ghrelin-induced hyperphagia, adiposity and insulin secretion are controlled by a central nervous system involving the mTORC1/S6K1 pathway.


Assuntos
Adiposidade/fisiologia , Ingestão de Energia , Grelina/fisiologia , Proteínas Quinases S6 Ribossômicas/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Tecido Adiposo Marrom/fisiologia , Animais , Linhagem Celular , Insulina/sangue , Canais Iônicos/metabolismo , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Knockout , Proteínas Mitocondriais/metabolismo , Complexos Multiproteicos/antagonistas & inibidores , Complexos Multiproteicos/metabolismo , Ratos , Ratos Wistar , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Proteína Desacopladora 1
6.
PLoS One ; 7(5): e37794, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22662224

RESUMO

BACKGROUND: Successful reduction of body weight (BW) is often followed by recidivism to obesity. BW-changes including BW-loss and -regain is associated with marked alterations in energy expenditure (EE) and adipose tissue (AT) metabolism. Since these processes are sex-specifically controlled, we investigated sexual dimorphisms in metabolic processes during BW-dynamics (gain-loss-regain). RESEARCH DESIGN: Obesity was induced in C57BL/6J male (m) and female (f) mice by 15 weeks high-fat diet (HFD) feeding. Subsequently BW was reduced (-20%) by caloric restriction (CR) followed by adaptive feeding, and a regain-phase. Measurement of EE, body composition, blood/organ sampling were performed after each feeding period. Lipolysis was analyzed ex-vivo in gonadal AT. RESULTS: Male mice exhibited accelerated BW-gain compared to females (relative BW-gain m:140.5±3.2%; f:103.7±6.5%; p<0.001). In consonance, lean mass-specific EE was significantly higher in females compared to males during BW-gain. Under CR female mice reached their target-BW significantly faster than male mice (m:12.2 days; f:7.6 days; p<0.001) accompanied by a sustained sex-difference in EE. In addition, female mice predominantly downsized gonadal AT whereas the relation between gonadal and total body fat was not altered in males. Accordingly, only females exhibited an increased rate of forskolin-stimulated lipolysis in AT associated with significantly higher glycerol concentrations, lower RER-values, and increased AT expression of adipose triglyceride lipase (ATGL) and hormone sensitive lipase (HSL). Analysis of AT lipolysis in estrogen receptor alpha (ERα)-deficient mice revealed a reduced lipolytic rate in the absence of ERα exclusively in females. Finally, re-feeding caused BW-regain faster in males than in females. CONCLUSION: The present study shows sex-specific dynamics during BW-gain-loss-regain. Female mice responded to CR with an increase in lipolytic activity, and augmented lipid-oxidation leading to more efficient weight loss. These processes likely involve ERα-dependent signaling in AT and sexual dimorphic regulation of genes involved in lipid metabolism.


Assuntos
Tecido Adiposo/metabolismo , Peso Corporal , Lipólise , Animais , Dieta Hiperlipídica , Receptor alfa de Estrogênio/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/metabolismo , Fatores Sexuais , Aumento de Peso , Redução de Peso
7.
PLoS One ; 7(5): e37026, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22615880

RESUMO

OBJECTIVES: Obesity and its distribution pattern are important factors for the prediction of the onset of diabetes in humans. Since several mouse models are suitable to study the pathophysiology of type 2 diabetes the aim was to validate a novel computed tomograph model (Aloka-Hitachi LCT-200) for the quantification of visceral, subcutaneous, brown and intrahepatic fat depots in mice. METHODS: Different lean and obese mouse models (C57BL/6, B6.V-Lep(ob), NZO) were used to determine the most adequate scanning parameters for the detection of the different fat depots. The data were compared with those obtained after preparation and weighing the fat depots. Liver fat content was determined by biochemical analysis. RESULTS: The correlations between weights of fat tissues on scale and weights determined by CT were significant for subcutaneous (r(2) = 0.995), visceral (r(2) = 0.990) and total white adipose tissue (r(2) = 0.992). Moreover, scans in the abdominal region, between lumbar vertebrae L4 to L5 correlated with whole-body fat distribution allowing experimenters to reduce scanning time and animal exposure to radiation and anesthesia. Test-retest reliability and measurements conducted by different experimenters showed a high reproducibility in the obtained results. Intrahepatic fat content estimated by CT was linearly related to biochemical analysis (r(2) = 0.915). Furthermore, brown fat mass correlated well with weighted brown fat depots (r(2) = 0.952). In addition, short-term cold-expose (4 °C, 4 hours) led to alterations in brown adipose tissue attributed to a reduction in triglyceride content that can be visualized as an increase in Hounsfield units by CT imaging. CONCLUSION: The 3D imaging of fat by CT provides reliable results in the quantification of total, visceral, subcutaneous, brown and intrahepatic fat in mice. This non-invasive method allows the conduction of longitudinal studies of obesity in mice and therefore enables experimenters to investigate the onset of complex diseases such as diabetes and obesity.


Assuntos
Tecido Adiposo Marrom/diagnóstico por imagem , Tecido Adiposo Branco/diagnóstico por imagem , Fígado Gorduroso/diagnóstico por imagem , Animais , Distribuição da Gordura Corporal , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/diagnóstico por imagem , Diabetes Mellitus Tipo 2/metabolismo , Fígado Gorduroso/diagnóstico , Feminino , Imageamento Tridimensional/métodos , Estudos Longitudinais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos/metabolismo , Radiografia Abdominal/métodos , Reprodutibilidade dos Testes , Tomografia Computadorizada por Raios X/métodos , Triglicerídeos/metabolismo
8.
Physiol Behav ; 105(3): 791-9, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22061427

RESUMO

The central melanocortin system regulates lipid metabolism in peripheral tissues such as white adipose tissue. Alterations in the activity of sympathetic nerves connecting hypothalamic cells expressing melanocortin 3/4 receptors (MC3/4R) with white adipocytes have been shown to partly mediate these effects. Interestingly, hypothalamic neurons producing corticotropin-releasing hormone and thyrotropin-releasing hormone co-express MC4R. Therefore we hypothesized that regulation of hypothalamo-pituitary adrenal (HPA) and hypothalamo-pituitary thyroid (HPT) axes activity by the central melanocortin system could contribute to its control of peripheral lipid metabolism. To test this hypothesis, we chronically infused rats intracerebroventricularly (i.c.v.) either with an MC3/4R antagonist (SHU9119), an MC3/4R agonist (MTII) or saline. Rats had been previously adrenalectomized (ADX) and supplemented daily with 1mg/kg corticosterone (s.c.), thyroidectomized (TDX) and supplemented daily with 10 µg/kgL-thyroxin (s.c.), or sham operated (SO). Blockade of MC3/4R signaling with SHU9119 increased food intake and body mass, irrespective of gland surgery. The increase in body mass was accompanied by higher epididymal white adipose tissue (eWAT) weight and higher mRNA content of lipogenic enzymes in eWAT. SHU9119 infusion increased triglyceride content in the liver of SO and TDX rats, but not in those of ADX rats. Concomitantly, mRNA expression of lipogenic enzymes in liver was increased in SO and TDX, but not in ADX rats. We conclude that the HPA and HPT axes do not play an essential role in mediating central melanocortinergic effects on white adipose tissue and liver lipid metabolism. However, while basal hepatic lipid metabolism does not depend on a functional HPA axis, the induction of hepatic lipogenesis due to central melanocortin system blockade does require a functional HPA axis.


Assuntos
Sistema Hipotálamo-Hipofisário/fisiologia , Fígado/metabolismo , Melanocortinas/metabolismo , Sistema Hipófise-Suprarrenal/fisiologia , Triglicerídeos/metabolismo , Adipócitos Brancos/efeitos dos fármacos , Adrenalectomia , Hormônio Adrenocorticotrópico/genética , Hormônio Adrenocorticotrópico/metabolismo , Animais , Peso Corporal/efeitos dos fármacos , Corticosterona/administração & dosagem , Corticosterona/metabolismo , Sistemas de Liberação de Medicamentos , Ingestão de Alimentos/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Injeções Intraventriculares , Masculino , Hormônios Estimuladores de Melanócitos/farmacologia , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Ratos , Ratos Wistar , Receptores da Corticotropina/agonistas , Receptores da Corticotropina/antagonistas & inibidores , Tireoidectomia , Tiroxina/farmacologia , alfa-MSH/análogos & derivados , alfa-MSH/farmacologia
9.
Endocrinology ; 152(12): 4641-51, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21990309

RESUMO

Dysregulation of fatty acid oxidation plays a pivotal role in the pathophysiology of obesity and insulin resistance. Medium- and short-chain-3-hydroxyacyl-coenzyme A (CoA) dehydrogenase (SCHAD) (gene name, hadh) catalyze the third reaction of the mitochondrial ß-oxidation cascade, the oxidation of 3-hydroxyacyl-CoA to 3-ketoacyl-CoA, for medium- and short-chain fatty acids. We identified hadh as a putative obesity gene by comparison of two genome-wide scans, a quantitative trait locus analysis previously performed in the polygenic obese New Zealand obese mouse and an earlier described small interfering RNA-mediated mutagenesis in Caenorhabditis elegans. In the present study, we show that mice lacking SCHAD (hadh(-/-)) displayed a lower body weight and a reduced fat mass in comparison with hadh(+/+) mice under high-fat diet conditions, presumably due to an impaired fuel efficiency, the loss of acylcarnitines via the urine, and increased body temperature. Food intake, total energy expenditure, and locomotor activity were not altered in knockout mice. Hadh(-/-) mice exhibited normal fat tolerance at 20 C. However, during cold exposure, knockout mice were unable to clear triglycerides from the plasma and to maintain their normal body temperature, indicating that SCHAD plays an important role in adaptive thermogenesis. Blood glucose concentrations in the fasted and postprandial state were significantly lower in hadh(-/-) mice, whereas insulin levels were elevated. Accordingly, insulin secretion in response to glucose and glucose plus palmitate was elevated in isolated islets of knockout mice. Therefore, our data indicate that SCHAD is involved in thermogenesis, in the maintenance of body weight, and in the regulation of nutrient-stimulated insulin secretion.


Assuntos
Acil-CoA Desidrogenase/fisiologia , Peso Corporal , Butiril-CoA Desidrogenase/fisiologia , Insulina/metabolismo , Termogênese , Animais , Glicemia , Temperatura Baixa , Metabolismo Energético , Secreção de Insulina , Camundongos , Camundongos Knockout , Triglicerídeos/sangue
10.
Physiol Behav ; 105(1): 43-51, 2011 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-21513721

RESUMO

Ghrelin increases food intake and decreases energy expenditure, promoting a positive energy balance. We observed a single case of serious hypothermia during sustained ghrelin treatment in a male subject, suggesting that ghrelin may play a role in the regulation of body temperature. We therefore investigated the effect of ghrelin treatment on body temperature in rodents and humans under controlled conditions. Intriguingly, we could demonstrate ghrelin binding in axon terminals of the medial preoptic area of the hypothalamus located in the vicinity of cold-sensitive neurons. This localization of ghrelin receptors provides a potential anatomical basis for the regulation of body temperature by ghrelin. However, our follow-up studies also indicated that neither a chronic i.c.v. application of ghrelin in rats, nor a single s.c. injection under cold exposure in mice resulted in a relevant decrease in body core temperature. In addition, a four-hour intravenous ghrelin infusion did not decrease body surface temperature in healthy humans. We concluded that while there is a theoretical molecular basis for ghrelin to modify body temperature in mammals, its magnitude is irrelevant under physiologic circumstances. Hypothermia is not likely to represent a serious risk associated with this agent and pathway.


Assuntos
Temperatura Corporal/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Grelina/farmacologia , Hipotermia/induzido quimicamente , Adolescente , Adulto , Idoso , Animais , Ingestão de Alimentos/fisiologia , Metabolismo Energético/fisiologia , Humanos , Hipotermia/fisiopatologia , Masculino , Camundongos , Pessoa de Meia-Idade , Ratos , Ratos Wistar , Telemetria
11.
Endocrinology ; 149(6): 3009-15, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18308842

RESUMO

We recently reported that the hypothalamic homeobox domain transcription factor Bsx plays an essential role in the central nervous system control of spontaneous physical activity and the generation of hyperphagic responses. Moreover, we found Bsx to be a master regulator for the hypothalamic expression of key orexigenic neuropeptide Y and agouti gene-related protein. We now hypothesized that Bsx, which is expressed in the dorsomedial and arcuate nucleus (ARC) of the hypothalamus, is regulated by afferent signals in response to peripheral energy balance. Bsx expression was analyzed using in situ hybridization in fed vs. fasted (24 h) and ghrelin vs. leptin-treated rats, as well as in mice deficient for leptin or the ghrelin signaling. Ghrelin administration increased, whereas ghrelin receptor antagonist decreased ARC Bsx expression. Leptin injection attenuated the fasting-induced increase in ARC Bsx levels but had no effect in fed rats. Dorsomedial hypothalamic nucleus Bsx expression was unaffected by pharmacological modifications of leptin or ghrelin signaling. Obese leptin-deficient (ob/ob) mice, but not obese melanocortin 4 receptor-knockout mice, showed higher expression of Bsx, consistent with dependency from afferent leptin rather than increased adiposity per se. Interestingly, exposure to a high-fat diet triggered Bsx expression, consistent with the concept that decreased leptin signaling due to a high-fat diet induced leptin resistance. Our data indicate that ARC Bsx expression is specifically regulated by afferent energy balance signals, including input from leptin and ghrelin. Future studies will be necessary to test if Bsx may be involved in the pathogenesis of leptin resistance.


Assuntos
Ingestão de Alimentos/fisiologia , Atividade Motora/fisiologia , Proteína X Associada a bcl-2/fisiologia , Animais , Metabolismo Energético , Jejum , Grelina/sangue , Insulina/sangue , Leptina/sangue , Masculino , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Proteína X Associada a bcl-2/genética
12.
J Clin Invest ; 117(11): 3475-88, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17885689

RESUMO

Disruptions of the melanocortin signaling system have been linked to obesity. We investigated a possible role of the central nervous melanocortin system (CNS-Mcr) in the control of adiposity through effects on nutrient partitioning and cellular lipid metabolism independent of nutrient intake. We report that pharmacological inhibition of melanocortin receptors (Mcr) in rats and genetic disruption of Mc4r in mice directly and potently promoted lipid uptake, triglyceride synthesis, and fat accumulation in white adipose tissue (WAT), while increased CNS-Mcr signaling triggered lipid mobilization. These effects were independent of food intake and preceded changes in adiposity. In addition, decreased CNS-Mcr signaling promoted increased insulin sensitivity and glucose uptake in WAT while decreasing glucose utilization in muscle and brown adipose tissue. Such CNS control of peripheral nutrient partitioning depended on sympathetic nervous system function and was enhanced by synergistic effects on liver triglyceride synthesis. Our findings offer an explanation for enhanced adiposity resulting from decreased melanocortin signaling, even in the absence of hyperphagia, and are consistent with feeding-independent changes in substrate utilization as reflected by respiratory quotient, which is increased with chronic Mcr blockade in rodents and in humans with loss-of-function mutations in MC4R. We also reveal molecular underpinnings for direct control of the CNS-Mcr over lipid metabolism. These results suggest ways to design more efficient pharmacological methods for controlling adiposity.


Assuntos
Sistema Nervoso Central/metabolismo , Metabolismo dos Lipídeos , Melanocortinas/metabolismo , Transdução de Sinais/fisiologia , Adipócitos/citologia , Adipócitos/metabolismo , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Animais , Comportamento Animal/fisiologia , Ingestão de Alimentos , Glucose/metabolismo , Humanos , Insulina/metabolismo , Hormônios Estimuladores de Melanócitos/administração & dosagem , Hormônios Estimuladores de Melanócitos/metabolismo , Camundongos , Camundongos Knockout , Ratos , Ratos Sprague-Dawley , Receptor Tipo 4 de Melanocortina/genética , Receptor Tipo 4 de Melanocortina/metabolismo , Receptores de Melanocortina , alfa-MSH/administração & dosagem , alfa-MSH/análogos & derivados , alfa-MSH/metabolismo
13.
Nat Clin Pract Endocrinol Metab ; 3(10): 705-12, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17893689

RESUMO

The high prevalence of obesity and diabetes will lead to higher rates of morbidity and mortality. The search for drugs to treat these metabolic disorders has, therefore, intensified. The stomach-derived peptide ghrelin regulates food intake and body weight. Recent work suggests that ghrelin also controls glucose metabolism. In addition, current evidence suggests that most of the actions of ghrelin could contribute to the metabolic syndrome. The ghrelin signaling system is, therefore, a promising target for the development of new drugs for the treatment of obesity and diabetes. Agents that block the ghrelin signaling system might be especially useful targets. This Review summarizes the potential and the limitations of ghrelin as a tool to better understand, prevent and treat obesity and diabetes.


Assuntos
Diabetes Mellitus/metabolismo , Obesidade/metabolismo , Hormônios Peptídicos/metabolismo , Animais , Glicemia/metabolismo , Diabetes Mellitus/tratamento farmacológico , Metabolismo Energético/fisiologia , Grelina , Humanos , Obesidade/complicações , Obesidade/tratamento farmacológico , Hormônios Peptídicos/uso terapêutico
14.
Cell Metab ; 5(6): 450-63, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17550780

RESUMO

Food intake and activity-induced thermogenesis are important components of energy balance regulation. The molecular mechanism underlying the coordination of food intake with locomotory behavior to maintain energy homeostasis is unclear. We report that the brain-specific homeobox transcription factor Bsx is required for locomotory behavior, hyperphagia, and expression of the hypothalamic neuropeptides Npy and Agrp, which regulate feeding behavior and body weight. Mice lacking Bsx exhibit reduced locomotor activity and lower expression of Npy and Agrp. They also exhibit attenuated physiological responses to fasting, including reduced increase of Npy/Agrp expression, lack of food-seeking behavior, and reduced rebound hyperphagia. Furthermore, Bsx gene disruption rescues the obese phenotype of leptin-deficient ob/ob mice by reducing their hyperphagia without increasing their locomotor activity. Thus, Bsx represents an essential factor for NPY/AgRP neuronal function and locomotory behavior in the control of energy balance.


Assuntos
Proteína Relacionada com Agouti/metabolismo , Encéfalo/metabolismo , Proteínas de Homeodomínio/fisiologia , Hiperfagia/metabolismo , Locomoção/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Neuropeptídeo Y/metabolismo , Animais , Comportamento Animal , Peso Corporal , Encéfalo/patologia , Células Cultivadas , Imunoprecipitação da Cromatina , Ingestão de Energia , Metabolismo Energético , Jejum/fisiologia , Técnicas Imunoenzimáticas , Hibridização In Situ , Luciferases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Endocrinology ; 148(1): 21-6, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17008393

RESUMO

Ghrelin stimulates food intake and adiposity and thereby increases body weight (BW) in rodents after central as well as peripheral administration. Recently, it was discovered that the gene precursor of ghrelin encoded another secreted and bioactive peptide named obestatin. First reports appeared to demonstrate that this peptide requires an amidation for its biological activity and acts through the orphan receptor, GPR-39. Obestatin was shown to have actions opposite to ghrelin on food intake, BW, and gastric emptying. In the present study, we failed to observe any effect of obestatin on food intake, BW, body composition, energy expenditure, locomotor activity, respiratory quotient, or hypothalamic neuropeptides involved in energy balance regulation. In agreement with the first report, we were unable to find any effect of obestatin on GH secretion in vivo. Moreover, we were unable to find mRNA expression of GPR-39, the putative obestatin receptor, in the hypothalamus of rats. Therefore, the results presented here do not support a role of the obestatin/GPR-39 system in the regulation of energy balance.


Assuntos
Metabolismo Energético/efeitos dos fármacos , Hormônio do Crescimento/metabolismo , Hipotálamo/efeitos dos fármacos , Hormônios Peptídicos/farmacologia , Animais , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Alimentos/fisiologia , Metabolismo Energético/fisiologia , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/fisiologia , Grelina , Hipotálamo/metabolismo , Hipotálamo/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Hormônios Peptídicos/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Vagotomia , Aumento de Peso/efeitos dos fármacos , Aumento de Peso/fisiologia
16.
J Clin Invest ; 116(7): 1983-93, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16767221

RESUMO

Many homeostatic processes, including appetite and food intake, are controlled by neuroendocrine circuits involving the CNS. The CNS also directly regulates adipocyte metabolism, as we have shown here by examining central action of the orexigenic hormone ghrelin. Chronic central ghrelin infusion resulted in increases in the glucose utilization rate of white and brown adipose tissue without affecting skeletal muscle. In white adipocytes, mRNA expression of various fat storage-promoting enzymes such as lipoprotein lipase, acetyl-CoA carboxylase alpha, fatty acid synthase, and stearoyl-CoA desaturase-1 was markedly increased, while that of the rate-limiting step in fat oxidation, carnitine palmitoyl transferase-1alpha, was decreased. In brown adipocytes, central ghrelin infusion resulted in lowered expression of the thermogenesis-related mitochondrial uncoupling proteins 1 and 3. These ghrelin effects were dose dependent, occurred independently from ghrelin-induced hyperphagia, and seemed to be mediated by the sympathetic nervous system. Additionally, the expression of some fat storage enzymes was decreased in ghrelin-deficient mice, which led us to conclude that central ghrelin is of physiological relevance in the control of cell metabolism in adipose tissue. These results unravel the existence of what we believe to be a new CNS-based neuroendocrine circuit regulating metabolic homeostasis of adipose tissue.


Assuntos
Adipócitos/metabolismo , Encéfalo/metabolismo , Hormônios Peptídicos/metabolismo , Acetil-CoA Carboxilase/genética , Acetil-CoA Carboxilase/metabolismo , Adipócitos/citologia , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Animais , Encéfalo/anatomia & histologia , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Ingestão de Alimentos/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Ácido Graxo Sintases/genética , Ácido Graxo Sintases/metabolismo , Grelina , Glucose/metabolismo , Homeostase , Canais Iônicos , Lipase Lipoproteica/genética , Lipase Lipoproteica/metabolismo , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Proteínas Mitocondriais , Neuropeptídeo Y/genética , Neuropeptídeo Y/metabolismo , Hormônios Peptídicos/administração & dosagem , Hormônios Peptídicos/genética , Ratos , Ratos Wistar , Estearoil-CoA Dessaturase/genética , Estearoil-CoA Dessaturase/metabolismo , Proteína Desacopladora 1 , Proteína Desacopladora 3
17.
J Nutr ; 135(5): 1314-9, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15867332

RESUMO

Obesity represents one of the most urgent global health threats as well as one of the leading causes of death throughout industrialized nations. Efficacious and safe therapies remain at large. Attempts to decrease fat mass via pharmacological reduction of energy intake have had limited potency or intolerable side effects. Increasingly widespread sedentary lifestyle is often cited as a major contributor to the increasing prevalence of obesity. Moreover, low levels of spontaneous physical activity (SPA) are a major predictor of fat mass accumulation during overfeeding in humans, pointing to a substantial role for SPA in the control of energy balance. Despite this, very little is known about the molecular mechanisms by which SPA is regulated. The overview will attempt to summarize available information on neuroendocrine factors regulating SPA.


Assuntos
Metabolismo Energético , Atividade Motora/fisiologia , Sistemas Neurossecretores/fisiologia , Obesidade/fisiopatologia , Hormônios Peptídicos/fisiologia , Animais , Grelina , Homeostase , Humanos , Obesidade/prevenção & controle
18.
J Exp Biol ; 207(Pt 16): 2859-66, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15235014

RESUMO

According to the set-point theory of body mass, changes in body mass are perceived by the body, leading to activation of compensatory feedback mechanisms, which in turn restores the set-point body mass. However, this theory is still under debate. To test if mass per se might be sensed and regulated, we implanted loads corresponding to 10% (HI) or 2% (LO, control) of body mass into mice in addition to sham-operated mice (SO). We recorded body mass, food intake, energy expenditure and body composition over 14 weeks. Both male and female mice showed an initial stress-induced loss of body mass, which was more pronounced in males. Subsequently, male HI mice displayed a permanently decreased biological body mass (MBB, body mass exclusive of the implant mass), equivalent to approximately half of the mass of the implant, and obtained by a decrease in fat mass compared to SO males. In contrast, female HI mice rapidly recovered and maintained their initial MBB and body composition following a mass load. Initial lean body mass was maintained in all male and female groups, and energy intake was similar in all male and female groups. Body mass changes could not be explained by measurable changes in energy intake or expenditure. We conclude that changes in body mass are perceived and partially compensated in male but not in female mice, suggesting that mass-specific regulation of body mass might not play a major role in overall body mass regulation. Different compartments of the body are possibly regulated by different signals and stimuli. Our results suggest that lean body mass rather than body mass per se seems to be tightly regulated. Higher efficiency of energy utilization in females compared to males could explain the gender-specific changes in energy balance.


Assuntos
Peso Corporal/fisiologia , Metabolismo Energético/fisiologia , Homeostase , Camundongos/fisiologia , Análise de Variância , Animais , Metabolismo Basal , Composição Corporal , Índice de Massa Corporal , Ingestão de Alimentos/fisiologia , Feminino , Masculino , Atividade Motora/fisiologia , Fatores Sexuais , Fatores de Tempo
19.
Br J Nutr ; 88(3): 301-6, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12207840

RESUMO

Early postnatal overnutrition (PNO) induced by restricting litter size in rats leads to increased body-weight (BW) and body-fat gain in later life. PNO rats are used as an animal model of moderate obesity and early hyperinsulinism. We investigated whether the increased adiposity could be due to a decreased energy expenditure. Male newborn Wistar rats were raised in litters of either two (SL) or twelve pups (NL), weaned at 4 weeks of age and subsequently fed ad libitum. BW was recorded continuously until 12 weeks of age. Daily energy intake, total daily energy expenditure (EE, measured by indirect calorimetry) and body composition were measured in weaned pups at 5, 8 and 12 weeks of age. SL rats displayed increased BW compared with NL rats from week 2 to 5 and again from week 10 to 12. Lean body mass, body fat and protein content and total EE were increased in SL rats at week 5. The same linear correlation described the relationship between BW and total EE in NL and SL rats. At week 8 to 12 no differences in energy metabolism could be found, but the total fat content was increased in SL rats at week 12. Energy balance, i.e. assimilated energy minus EE, was no different between SL and NL at any time that it was measured. We conclude that although PNO rats display increased adiposity in early life, there seem to be no long-lasting effects on energy metabolism in later life, even if a tendency to increased adiposity can still be detected.


Assuntos
Metabolismo Energético , Obesidade/fisiopatologia , Tecido Adiposo/patologia , Envelhecimento/fisiologia , Animais , Animais Recém-Nascidos , Composição Corporal , Ingestão de Energia , Tamanho da Ninhada de Vivíparos , Masculino , Proteínas/análise , Ratos , Ratos Wistar , Aumento de Peso
20.
J Gravit Physiol ; 9(1): P189-90, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15002542

RESUMO

Numerous factors are involved to maintain a stable body mass (BM). We hypothesize that also weight per se can influence BM regulation. Male and female mice (FVB) were sham operated or implanted with a metabolic inert load weighing 2% (LO) or 10% (HI) of initial BM. BM, energy intake (El) and energy expenditure (EE) were measured. The artificial weight load influenced BM regulation gender specifically. In males, BM was reduced by 3-5% in HI when compared to SO and LO; EI was unchanged. In females, BM development was similar in all groups; EI was increased by 7 and 12% in LO and HI, respectively, when compared to SO. We conclude that the coupling of EI and EE seems to be tighter in females than in males in order to keep a stable BM.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA