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











Base de dados
Intervalo de ano de publicação
1.
Sci Transl Med ; 15(723): eadd4897, 2023 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-37992152

RESUMO

Deficiency in the adipose-derived hormone leptin or leptin receptor signaling causes class 3 obesity in individuals with genetic loss-of-function mutations in leptin or its receptor LEPR and metabolic and liver disease in individuals with hypoleptinemia secondary to lipoatrophy such as in individuals with generalized lipodystrophy. Therapies that restore leptin-LEPR signaling may resolve these metabolic sequelae. We developed a fully human monoclonal antibody (mAb), REGN4461 (mibavademab), that activates the human LEPR in the absence or presence of leptin. In obese leptin knockout mice, REGN4461 normalized body weight, food intake, blood glucose, and insulin sensitivity. In a mouse model of generalized lipodystrophy, REGN4461 alleviated hyperphagia, hyperglycemia, insulin resistance, dyslipidemia, and hepatic steatosis. In a phase 1, randomized, double-blind, placebo-controlled two-part study, REGN4461 was well tolerated with an acceptable safety profile. Treatment of individuals with overweight or obesity with REGN4461 decreased body weight over 12 weeks in those with low circulating leptin concentrations (<8 ng/ml) but had no effect on body weight in individuals with higher baseline leptin. Furthermore, compassionate-use treatment of a single patient with atypical partial lipodystrophy and a history of undetectable leptin concentrations associated with neutralizing antibodies to metreleptin was associated with noteable improvements in circulating triglycerides and hepatic steatosis. Collectively, these translational data unveil an agonist LEPR mAb that may provide clinical benefit in disorders associated with relatively low leptin concentrations.


Assuntos
Resistência à Insulina , Lipodistrofia Generalizada Congênita , Animais , Camundongos , Humanos , Leptina/uso terapêutico , Ensaios de Uso Compassivo , Receptores para Leptina/metabolismo , Lipodistrofia Generalizada Congênita/tratamento farmacológico , Obesidade/tratamento farmacológico , Anticorpos/uso terapêutico , Peso Corporal
2.
Sci Adv ; 9(11): eade4395, 2023 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-36930708

RESUMO

The interleukin-6 (IL-6) family cytokines signal through gp130 receptor homodimerization or heterodimerization with a second signaling receptor and play crucial roles in various cellular processes. We determined cryo-electron microscopy structures of five signaling complexes of this family, containing full receptor ectodomains bound to their respective ligands ciliary neurotrophic factor, cardiotrophin-like cytokine factor 1 (CLCF1), leukemia inhibitory factor, IL-27, and IL-6. Our structures collectively reveal similarities and differences in the assembly of these complexes. The acute bends at both signaling receptors in all complexes bring the membrane-proximal domains to a ~30 angstrom range but with distinct distances and orientations. We also reveal how CLCF1 engages its secretion chaperone cytokine receptor-like factor 1. Our data provide valuable insights for therapeutically targeting gp130-mediated signaling.


Assuntos
Antígenos CD , Interleucina-6 , Receptor gp130 de Citocina/metabolismo , Interleucina-6/metabolismo , Subunidade alfa de Receptor de Fator Inibidor de Leucemia/metabolismo , Microscopia Crioeletrônica , Antígenos CD/metabolismo , Glicoproteínas de Membrana/metabolismo , Citocinas/metabolismo
3.
Skelet Muscle ; 12(1): 26, 2022 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-36539856

RESUMO

BACKGROUND: Sarcopenia is defined as age-related low muscle mass and function, and can also describe the loss of muscle mass in certain medical conditions, such as sarcopenic obesity. Sarcopenic obesity describes loss of muscle and function in obese individuals; however, as sarcopenia is an age-related condition and obesity can occur in any age group, a more accurate term is obesity with low lean muscle mass (OLLMM). Given limited data on OLLMM (particularly in those aged < 65 years), the purpose of this study was to estimate the prevalence of OLLMM in adults aged ≥ 20 years in the USA. METHODS: Data from the National Health and Nutrition Examination Survey (NHANES) 2017-2018 and 1999-2006 were used. OLLMM was defined as an appendicular lean mass, adjusted for body mass index (BMI), cut-off point < 0.789 for males and < 0.512 for females, measured by dual-energy X-ray absorptiometry (DXA). DXA was only measured in individuals 20-59 years old in NHANES 2017-2018; we therefore utilized logistic regression models to predict OLLMM from NHANES 1999-2006 for those aged ≥ 60 years. The prevalence of OLLMM was estimated overall, and by sex, age, race/ethnicity, and clinical subgroup (high BMI, prediabetes, type 2 diabetes mellitus [T2DM], non-alcoholic fatty liver disease [NAFLD] with fibrosis, or post-bariatric surgery). Prevalence estimates were extrapolated to the USA population using NHANES sampling weights. RESULTS: We estimated that, during 2017-2018, 28.7 million or 15.9% of the USA population had OLLMM. The prevalence of OLLMM was greater in older individuals (8.1%, aged 20-59 years vs 28.3%, aged ≥ 60 years), highest (66.6%) in Mexican-American females aged ≥ 60 years, and lowest (2.6%) in non-Hispanic Black males aged 20-59 years. There was a higher prevalence of OLLMM in adults with prediabetes (19.7%), T2DM (34.5%), NAFLD with fibrosis (25.4%), or post-bariatric surgery (21.8%), compared with those without each condition. CONCLUSIONS: Overall, the burden of OLLMM in the USA is substantial, affecting almost 30 million adults. The prevalence of OLLMM increased with age, and among those with prediabetes, T2DM, NAFLD with fibrosis, or post-bariatric surgery. A unified definition of OLLMM will aid diagnosis and treatment strategies.


Assuntos
Diabetes Mellitus Tipo 2 , Hepatopatia Gordurosa não Alcoólica , Estado Pré-Diabético , Sarcopenia , Masculino , Adulto , Feminino , Humanos , Idoso , Adulto Jovem , Pessoa de Meia-Idade , Sarcopenia/epidemiologia , Inquéritos Nutricionais , Hepatopatia Gordurosa não Alcoólica/complicações , Diabetes Mellitus Tipo 2/complicações , Prevalência , Estado Pré-Diabético/complicações , Obesidade/complicações , Obesidade/epidemiologia , Fibrose , Músculos , Composição Corporal
4.
Nat Commun ; 13(1): 4844, 2022 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-35999217

RESUMO

Body fat distribution is a major, heritable risk factor for cardiometabolic disease, independent of overall adiposity. Using exome-sequencing in 618,375 individuals (including 160,058 non-Europeans) from the UK, Sweden and Mexico, we identify 16 genes associated with fat distribution at exome-wide significance. We show 6-fold larger effect for fat-distribution associated rare coding variants compared with fine-mapped common alleles, enrichment for genes expressed in adipose tissue and causal genes for partial lipodystrophies, and evidence of sex-dimorphism. We describe an association with favorable fat distribution (p = 1.8 × 10-09), favorable metabolic profile and protection from type 2 diabetes (~28% lower odds; p = 0.004) for heterozygous protein-truncating mutations in INHBE, which encodes a circulating growth factor of the activin family, highly and specifically expressed in hepatocytes. Our results suggest that inhibin ßE is a liver-expressed negative regulator of adipose storage whose blockade may be beneficial in fat distribution-associated metabolic disease.


Assuntos
Diabetes Mellitus Tipo 2 , Subunidades beta de Inibinas/genética , Tecido Adiposo , Adiposidade/genética , Diabetes Mellitus Tipo 2/genética , Exoma/genética , Humanos , Mutação
5.
J Cell Mol Med ; 25(20): 9878-9883, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34558178

RESUMO

NASH is a chronic liver disease that affects 3%-6% of individuals and requires urgent therapeutic developments. Isolating the key cell types in the liver is a necessary step towards understanding their function and roles in disease pathogenesis. However, traditional isolation methods through gradient centrifugation can only collect one or a few cell types simultaneously and pose technical difficulties when applied to NASH livers. Taking advantage of identified cell surface markers from liver single-cell RNAseq, here we established the combination of gradient centrifugation and antibody-based cell sorting techniques to isolate five key liver cell types (hepatocytes, endothelial cells, stellate cells, macrophages and other immune cells) from a single mouse liver. This method yielded high purity of each cell type from healthy and NASH livers. Our five-in-one protocol simultaneously isolates key liver cell types with high purity under normal and NASH conditions, enabling for systematic and accurate exploratory experiments such as RNA sequencing.


Assuntos
Separação Celular , Fígado/citologia , Hepatopatia Gordurosa não Alcoólica/patologia , Animais , Biomarcadores , Separação Celular/métodos , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Imunofluorescência , Hepatócitos/metabolismo , Imunofenotipagem , Macrófagos/metabolismo , Masculino , Camundongos , Hepatopatia Gordurosa não Alcoólica/etiologia
6.
JCI Insight ; 6(16)2021 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-34283813

RESUMO

Energy balance is controlled by interconnected brain regions in the hypothalamus, brainstem, cortex, and limbic system. Gene expression signatures of these regions can help elucidate the pathophysiology underlying obesity. RNA sequencing was conducted on P56 C57BL/6NTac male mice and E14.5 C57BL/6NTac embryo punch biopsies in 16 obesity-relevant brain regions. The expression of 190 known obesity-associated genes (monogenic, rare, and low-frequency coding variants; GWAS; syndromic) was analyzed in each anatomical region. Genes associated with these genetic categories of obesity had localized expression patterns across brain regions. Known monogenic obesity causal genes were highly enriched in the arcuate nucleus of the hypothalamus and developing hypothalamus. The obesity-associated genes clustered into distinct "modules" of similar expression profile, and these were distinct from expression modules formed by similar analysis with genes known to be associated with other disease phenotypes (type 1 and type 2 diabetes, autism, breast cancer) in the same energy balance-relevant brain regions.


Assuntos
Encéfalo/metabolismo , Metabolismo Energético/genética , Obesidade/metabolismo , Animais , Embrião de Mamíferos , Regulação da Expressão Gênica , Predisposição Genética para Doença , Masculino , Camundongos , Obesidade/genética , RNA-Seq
7.
Endocrinology ; 153(11): 5452-66, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22962259

RESUMO

Ghrelin is the endogenous ligand for the GH secretagogue receptor (GHSR) and robustly stimulates GH release from the anterior pituitary gland. Ghrelin also regulates the secretion of anterior pituitary hormones including TSH, LH, prolactin (PRL), and ACTH. However, the relative contribution of a direct action at the GHSR in the anterior pituitary gland vs. an indirect action at the GHSR in the hypothalamus remains undefined. We used a novel GHSR-enhanced green fluorescent protein (eGFP) reporter mouse to quantify GHSR coexpression with GH, TSH, LH, PRL, and ACTH anterior pituitary cells in males vs. females and in chow-fed or calorie-restricted (CR) mice. GHSR-eGFP-expressing cells were only observed in anterior pituitary. The number of GHSR-eGFP-expressing cells was higher in male compared with females, and CR did not affect the GHSR-eGFP cell number. Double staining revealed 77% of somatotrophs expressed GHSR-eGFP in both males and females. Nineteen percent and 12.6% of corticotrophs, 21% and 9% of lactotrophs, 18% and 19% of gonadotrophs, and 3% and 9% of males and females, respectively, expressed GHSR-eGFP. CR increased the number of TSH cells, but suppressed the number of lactotrophs and gonadotrophs, expressing GHSR-eGFP compared with controls. These studies support a robust stimulatory action of ghrelin via the GHSR on GH secretion and identify a previously unknown sexual dimorphism in the GHSR expression in the anterior pituitary. CR affects GHSR-eGFP expression on lactotrophs, gonadotrophs, and thyrotrophs, which may mediate reproductive function and energy metabolism during periods of negative energy balance. The low to moderate expression of GHSR-eGFP suggests that ghrelin plays a minor direct role on remaining anterior pituitary cells.


Assuntos
Adeno-Hipófise/metabolismo , Hormônios Adeno-Hipofisários/metabolismo , Receptores de Grelina/metabolismo , Hormônio Adrenocorticotrópico/metabolismo , Animais , Linhagem Celular , Feminino , Privação de Alimentos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hormônio do Crescimento/metabolismo , Hormônio Luteinizante/metabolismo , Masculino , Camundongos , Adeno-Hipófise/citologia , Prolactina/metabolismo , Tireotropina/metabolismo
8.
FASEB J ; 26(1): 259-71, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21965605

RESUMO

Ghrelin is a unique peptide gut hormone that requires post-translational modification to stimulate both feeding and growth hormone release. Ghrelin O-acyltransferase (GOAT) was identified as a specific acyl-transferase for ghrelin, and recent genetic deletion studies of the Goat gene (Goat(-/-)) uncovered the role of ghrelin in the regulation of glucose homeostasis. To further understand the physiological functions of the GOAT/ghrelin system, we have conducted a metabolomic and microarray profile of Goat-null mice, as well as determined Goat expression in different tissues using the lacZ reporter gene. Serum metabolite profile analysis revealed that Goat(-/-) mice exhibited increased secondary bile acids >2.5-fold. This was attributed to increased mRNA and protein expression of the ileal sodium-dependent bile acid transporter (ISBT) in the intestinal and biliary tract. Increased expression of additional solute carrier proteins, including Slc5a12 (>10-fold) were also detected in the small intestine and bile duct. Goat staining was consistently observed in the pituitary glands, stomach, and intestines, and to a lesser extent in the gallbladder and pancreatic duct. This is the first report that the GOAT/ghrelin system regulates bile acid metabolism, and these findings suggest a novel function of GOAT in the regulation of intestinal bile acid reabsorption..


Assuntos
Aciltransferases/genética , Aciltransferases/metabolismo , Ácidos e Sais Biliares/metabolismo , Absorção Intestinal/fisiologia , Metaboloma/genética , Acilação , Tecido Adiposo/anatomia & histologia , Tecido Adiposo/metabolismo , Animais , Sistema Biliar/metabolismo , Linhagem Celular , Cromossomos Artificiais Bacterianos/genética , Ingestão de Alimentos/fisiologia , Vesícula Biliar/metabolismo , Perfilação da Expressão Gênica , Grelina/metabolismo , Íleo/metabolismo , Óperon Lac/genética , Masculino , Proteínas de Membrana , Camundongos , Camundongos Mutantes , Adeno-Hipófise/metabolismo
9.
Cell Metab ; 7(5): 389-99, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18460330

RESUMO

Current evidence suggests that hypothalamic fatty acid metabolism may play a role in regulating food intake; however, confirmation that it is a physiologically relevant regulatory system of feeding is still incomplete. Here, we use pharmacological and genetic approaches to demonstrate that the physiological orexigenic response to ghrelin involves specific inhibition of fatty acid biosynthesis induced by AMP-activated protein kinase (AMPK) resulting in decreased hypothalamic levels of malonyl-CoA and increased carnitine palmitoyltransferase 1 (CPT1) activity. In addition, we also demonstrate that fasting downregulates fatty acid synthase (FAS) in a region-specific manner and that this effect is mediated by an AMPK and ghrelin-dependent mechanisms. Thus, decreasing AMPK activity in the ventromedial nucleus of the hypothalamus (VMH) is sufficient to inhibit ghrelin's effects on FAS expression and feeding. Overall, our results indicate that modulation of hypothalamic fatty acid metabolism specifically in the VMH in response to ghrelin is a physiological mechanism that controls feeding.


Assuntos
Ácidos Graxos/metabolismo , Grelina/fisiologia , Hipotálamo/metabolismo , Quinases Proteína-Quinases Ativadas por AMP , Animais , Western Blotting , Carnitina O-Palmitoiltransferase/metabolismo , Jejum/fisiologia , Ácido Graxo Sintases/antagonistas & inibidores , Ácido Graxo Sintases/metabolismo , Comportamento Alimentar , Hipotálamo/patologia , Hibridização In Situ , Leptina/metabolismo , Masculino , Malonil Coenzima A/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , Fosforilação , Proteínas Quinases/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor fas
10.
Am J Physiol Gastrointest Liver Physiol ; 294(3): G610-8, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18048479

RESUMO

Administration of chemically synthesized ghrelin (Ghr) peptide has been shown to increase food intake and body adiposity in most species. However, the biological role of endogenous Ghr in the molecular control of energy metabolism is far less understood. Mice deficient for either Ghr or its receptor (the growth hormone secretagogue receptor, GHS-R1a) seem to exhibit enhanced protection against high-fat diet-induced obesity but do not show a substantial metabolic phenotype on a standard diet. Here we present the first mouse mutant lacking both Ghr and the Ghr receptor. We demonstrate that simultaneous genetic disruption of both genes of the Ghr system leads to an enhanced energy metabolism phenotype. Ghr/Ghr receptor double knockout (dKO) mice exhibit decreased body weight, increased energy expenditure, and increased motor activity on a standard diet without exposure to a high caloric environment. Mice on the same genetic background lacking either the Ghr or the Ghr receptor gene did not exhibit such a phenotype on standard chow, thereby confirming earlier reports. No differences in food intake, meal pattern, or lean mass were observed between dKO, Ghr-deficient, Ghr receptor-deficient, and wild-type (WT) control mice. Only dKO showed a slight decrease in body length. In summary, simultaneous deletion of Ghr and its receptor enhances the metabolic phenotype of single gene-deficient mice compared with WT mice, possibly suggesting the existence of additional, as of yet unknown, molecular components of the endogenous Ghr system.


Assuntos
Metabolismo Energético/fisiologia , Grelina/deficiência , Grelina/genética , Atividade Motora/fisiologia , Receptores de Grelina/deficiência , Receptores de Grelina/genética , Alelos , Animais , Antropometria , Glicemia/metabolismo , Composição Corporal/genética , Composição Corporal/fisiologia , Temperatura Corporal/fisiologia , Peso Corporal/genética , Peso Corporal/fisiologia , Ingestão de Alimentos/genética , Ingestão de Alimentos/fisiologia , Deleção de Genes , Genótipo , Teste de Tolerância a Glucose , Resistência à Insulina/genética , Ligantes , Lipídeos/sangue , Camundongos , Camundongos Knockout , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
Gastroenterology ; 133(5): 1534-43, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17920065

RESUMO

BACKGROUND & AIMS: Peptide YY (PYY) and pancreatic polypeptide (PPY) are members of the neuropeptide Y peptide family. The neuropeptide Y receptor signaling pathway has been implicated in a number of physiologic processes, including the regulation of energy balance and bone mass. To investigate the contribution of endogenous PYY and PPY to these processes, we generated both Pyy- and Ppy-deficient mice. METHODS: Pyy(-/-) and Ppy(-/-) mice and their respective wild-type littermates were studied from 8 weeks to 9 months of age. Food intake, metabolic parameters, and locomotor activity were monitored using indirect calorimetry. Body composition and bone parameters were analyzed using dual energy x-ray absorptiometry, histomorphometry, and vertebral compression testing. RESULTS: Studies in these mice showed an osteopenic phenotype specific to the Pyy-deficient line, which included a reduction in trabecular bone mass and a functional deficit in bone strength. Furthermore, female Pyy(-/-) mice showed a greater sensitivity to ovariectomy-induced bone loss compared with wild-type littermates. No food intake or metabolic phenotype was apparent in male or female Pyy(-/-) mice on standard chow. However, female Pyy(-/-) mice on a high-fat diet showed a greater propensity to gain body weight and adiposity. No metabolic or osteopenic phenotype was observed in Ppy-deficient mice. CONCLUSIONS: These results indicate that endogenous PYY plays a critical role in regulating bone mass. In comparison, its role in regulating body weight is minor and is confined to situations of high-fat feeding.


Assuntos
Densidade Óssea/fisiologia , Osso e Ossos/metabolismo , Metabolismo Energético/fisiologia , Peptídeo YY/fisiologia , Absorciometria de Fóton , Adiposidade/efeitos dos fármacos , Animais , Composição Corporal/genética , Composição Corporal/fisiologia , Peso Corporal/efeitos dos fármacos , Densidade Óssea/genética , Doenças Ósseas Metabólicas/genética , Doenças Ósseas Metabólicas/metabolismo , Doenças Ósseas Metabólicas/fisiopatologia , Gorduras na Dieta/farmacologia , Modelos Animais de Doenças , Metabolismo Energético/genética , Feminino , Deleção de Genes , Masculino , Camundongos , Camundongos Transgênicos , Ovariectomia , Peptídeo YY/genética , Fenótipo , Coluna Vertebral/patologia , Coluna Vertebral/fisiopatologia
12.
Diabetes ; 56(5): 1350-6, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17303803

RESUMO

Trb3, a mammalian homolog of Drosophila tribbles, was proposed as a suppressor of Akt activity, predominantly in conditions of fasting and diabetes. Given these prior studies, we sought to determine whether Trb3 plays a major role in modulating hepatic insulin sensitivity. To answer this question, we produced mice in which a lacZ reporter was knocked into the locus containing the gene Trib3, resulting in a Trib3 null animal. Trib3 expression analyses demonstrated that the Trib3 is expressed in liver, adipose tissues, heart, kidney, lung, skin, small intestine, stomach, and denervated, but not normal, skeletal muscle. Trib3(-/-) mice are essentially identical to their wild-type littermates in overall appearance and body composition. Phenotypic analysis of Trib3(-/-) mice did not detect any alteration in serum glucose, insulin, or lipid levels; glucose or insulin tolerance; or energy metabolism. Studies in Trib3(-/-) hepatocytes revealed normal Akt and glycogen synthase kinase- 3beta phosphorylation patterns, glycogen levels, and expressions of key regulatory gluconeogenic and glycolytic genes. These data demonstrate that deletion of Trib3 has minimal effect on insulin-induced Akt activation in hepatic tissue, and, as such, they question any nonredundant role for Trb3 in the maintenance of glucose and energy homeostasis in mice.


Assuntos
Glucose/metabolismo , Insulina/fisiologia , Fígado/fisiologia , Acetil-CoA Carboxilase/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Cromossomos Artificiais Bacterianos , Genes Reporter , Insulina/farmacologia , Fígado/efeitos dos fármacos , Glicogênio Hepático/metabolismo , Camundongos , Camundongos Knockout , Proteína Oncogênica v-akt/efeitos dos fármacos , Proteína Oncogênica v-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
13.
Diabetes ; 55(12): 3372-80, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17130482

RESUMO

Despite altered regulation of insulin signaling, Pten(+/-) heterodeficient standard diet-fed mice, approximately 4 months old, exhibit normal fasting glucose and insulin levels. We report here a stable isotope flux phenotyping study of this "silent" phenotype, in which tissue-specific insulin effects in whole-body Pten(+/-)-deficient mice were dissected in vivo. Flux phenotyping showed gain of function in Pten(+/-) mice, seen as increased peripheral glucose disposal, and compensation by a metabolic feedback mechanism that 1) decreases hepatic glucose recycling via suppression of glucokinase expression in the basal state to preserve hepatic glucose production and 2) increases hepatic responsiveness in the fasted-to-fed transition. In Pten(+/-) mice, hepatic gene expression of glucokinase was 10-fold less than wild-type (Pten(+/+)) mice in the fasted state and reached Pten(+/+) values in the fed state. Glucose-6-phosphatase expression was the same for Pten(+/-) and Pten(+/+) mice in the fasted state, and its expression for Pten(+/-) was 25% of Pten(+/+) in the fed state. This study demonstrates how intra- and interorgan flux compensations can preserve glucose homeostasis (despite a specific gene defect that accelerates glucose disposal) and how flux phenotyping can dissect these tissue-specific flux compensations in mice presenting with a "silent" phenotype.


Assuntos
Fígado/fisiologia , Camundongos Knockout , PTEN Fosfo-Hidrolase/deficiência , PTEN Fosfo-Hidrolase/genética , Animais , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Ingestão de Alimentos , Jejum , Regulação Enzimológica da Expressão Gênica , Glucoquinase/genética , Teste de Tolerância a Glucose , Glucose-6-Fosfatase/genética , Insulina/farmacologia , Lipólise , Camundongos
14.
J Clin Invest ; 116(12): 3229-39, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17060947

RESUMO

The gut hormone ghrelin targets the brain to promote food intake and adiposity. The ghrelin receptor growth hormone secretagogue 1 receptor (GHSR) is present in hypothalamic centers controlling energy metabolism as well as in the ventral tegmental area (VTA), a region important for motivational aspects of multiple behaviors, including feeding. Here we show that in mice and rats, ghrelin bound to neurons of the VTA, where it triggered increased dopamine neuronal activity, synapse formation, and dopamine turnover in the nucleus accumbens in a GHSR-dependent manner. Direct VTA administration of ghrelin also triggered feeding, while intra-VTA delivery of a selective GHSR antagonist blocked the orexigenic effect of circulating ghrelin and blunted rebound feeding following fasting. In addition, ghrelin- and GHSR-deficient mice showed attenuated feeding responses to restricted feeding schedules. Taken together, these data suggest that the mesolimbic reward circuitry is targeted by peripheral ghrelin to influence physiological mechanisms related to feeding.


Assuntos
Apetite/efeitos dos fármacos , Dopamina/metabolismo , Neurônios/metabolismo , Hormônios Peptídicos/farmacologia , Potenciais de Ação/efeitos dos fármacos , Animais , Imunofluorescência/métodos , Grelina , Masculino , Mesencéfalo/citologia , Mesencéfalo/efeitos dos fármacos , Mesencéfalo/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Núcleo Accumbens/citologia , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Técnicas de Patch-Clamp , Hormônios Peptídicos/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Grelina , Fatores de Tempo , Área Tegmentar Ventral/citologia , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/metabolismo
15.
Nat Neurosci ; 9(3): 381-8, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16491079

RESUMO

The gut hormone and neuropeptide ghrelin affects energy balance and growth hormone release through hypothalamic action that involves synaptic plasticity in the melanocortin system. Ghrelin binding is also present in other brain areas, including the telencephalon, where its function remains elusive. Here we report that circulating ghrelin enters the hippocampus and binds to neurons of the hippocampal formation, where it promotes dendritic spine synapse formation and generation of long-term potentiation. These ghrelin-induced synaptic changes are paralleled by enhanced spatial learning and memory. Targeted disruption of the gene that encodes ghrelin resulted in decreased numbers of spine synapses in the CA1 region and impaired performance of mice in behavioral memory testing, both of which were rapidly reversed by ghrelin administration. Our observations reveal an endogenous function of ghrelin that links metabolic control with higher brain functions and suggest novel therapeutic strategies to enhance learning and memory processes.


Assuntos
Espinhas Dendríticas/metabolismo , Hipocampo/metabolismo , Memória/fisiologia , Hormônios Peptídicos/genética , Sinapses/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Espinhas Dendríticas/efeitos dos fármacos , Espinhas Dendríticas/ultraestrutura , Grelina , Hipocampo/efeitos dos fármacos , Hipocampo/ultraestrutura , Aprendizagem/efeitos dos fármacos , Aprendizagem/fisiologia , Potenciação de Longa Duração/efeitos dos fármacos , Potenciação de Longa Duração/fisiologia , Masculino , Memória/efeitos dos fármacos , Transtornos da Memória/tratamento farmacológico , Transtornos da Memória/genética , Transtornos da Memória/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nootrópicos/metabolismo , Nootrópicos/farmacologia , Hormônios Peptídicos/farmacologia , Ratos , Ratos Sprague-Dawley , Percepção Espacial/efeitos dos fármacos , Percepção Espacial/fisiologia , Sinapses/efeitos dos fármacos , Sinapses/ultraestrutura , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/genética
16.
J Clin Invest ; 115(12): 3573-8, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16322795

RESUMO

The gut peptide ghrelin, the endogenous ligand for the growth hormone secretagogue receptor, has been implicated not only in the regulation of pituitary growth hormone (GH) secretion but in a number of endocrine and nonendocrine functions, including appetitive behavior and carbohydrate substrate utilization. Nevertheless, recent genetic studies have failed to show any significant defects in GH levels, food intake, or body weight in adult ghrelin-deficient (Ghrl-/-) mice. Here we demonstrate that male Ghrl-/- mice are protected from the rapid weight gain induced by early exposure to a high-fat diet 3 weeks after weaning (6 weeks of age). This reduced weight gain was associated with decreased adiposity and increased energy expenditure and locomotor activity as the animals aged. Despite the absence of ghrelin, these Ghrl-/- mice showed a paradoxical preservation of the GH/IGF-1 axis, similar to that reported in lean compared with obese humans. These findings suggest an important role for endogenous ghrelin in the metabolic adaptation to nutrient availability.


Assuntos
Obesidade/genética , Hormônios Peptídicos/fisiologia , Ração Animal , Animais , Composição Corporal , Peso Corporal , Calorimetria , Carboidratos/química , Dieta , Genótipo , Grelina , Hormônio do Crescimento/metabolismo , Heterozigoto , Fator de Crescimento Insulin-Like I/metabolismo , Ligantes , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Biológicos , Neuropeptídeos/química , Oxigênio/metabolismo , Hormônios Peptídicos/metabolismo , Fenótipo , Hipófise/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo
17.
Nat Genet ; 37(10): 1072-81, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16170315

RESUMO

Multiple organs cooperate to regulate appetite, metabolism, and glucose and fatty acid homeostasis. Here, we identified and characterized lymphatic vasculature dysfunction as a cause of adult-onset obesity. We found that functional inactivation of a single allele of the homeobox gene Prox1 led to adult-onset obesity due to abnormal lymph leakage from mispatterned and ruptured lymphatic vessels. Prox1 heterozygous mice are a new model for adult-onset obesity and lymphatic vascular disease.


Assuntos
Proteínas de Homeodomínio/genética , Metabolismo dos Lipídeos/genética , Anormalidades Linfáticas/genética , Obesidade/genética , Alelos , Animais , Modelos Animais de Doenças , Deleção de Genes , Proteínas de Homeodomínio/metabolismo , Insulina/sangue , Leptina/sangue , Lipídeos/análise , Fígado/metabolismo , Linfa/metabolismo , Anormalidades Linfáticas/complicações , Vasos Linfáticos/anormalidades , Vasos Linfáticos/fisiopatologia , Camundongos , Camundongos Knockout , Obesidade/complicações , Proteínas Supressoras de Tumor
18.
Proc Natl Acad Sci U S A ; 102(7): 2496-501, 2005 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-15699348

RESUMO

We identified a glycoprotein hormone beta-subunit (OGH, also called GPB5) that, as a heterodimer with the alpha-subunit GPA2, serves as a second ligand for the thyroid-stimulating hormone receptor. Mice in which the OGH gene is deleted (OGH-/-) are indistinguishable from WT littermates in body weight, response to high-fat diet, metabolic parameters, body composition, and insulin tolerance. Mice engineered to transgenically globally overexpress OGH (OGH-TG) develop approximately 2-fold elevations in their basal thyroid levels and weigh slightly less than WT littermates despite increased food intake because of an increase in their metabolic rates. Moreover, when OGH-TG mice are challenged with a high-fat diet, they gain significantly less weight and body fat than their WT littermates. The OGH-TG mice also have reduced blood glucose, insulin, cholesterol, and triglycerides. In contrast to other approaches in which the thyroid axis is activated, OGH-TG mice exhibit only minor changes in heart rate and blood pressure. Our findings suggest that constitutive low-level activation of the thyroid axis (via OGH or other means) may provide a beneficial therapeutic approach for combating diet-induced obesity.


Assuntos
Glicoproteínas/genética , Obesidade/genética , Hormônios Peptídicos/genética , Animais , Peso Corporal , Gorduras na Dieta/administração & dosagem , Expressão Gênica , Genes Reporter , Óperon Lac , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Modelos Biológicos , Dados de Sequência Molecular , Obesidade/metabolismo , Obesidade/patologia , Fenótipo
19.
Nat Med ; 11(2): 199-205, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15654325

RESUMO

Genetic ablation of Inppl1, which encodes SHIP2 (SH2-domain containing inositol 5-phosphatase 2), was previously reported to induce severe insulin sensitivity, leading to early postnatal death. In the previous study, the targeting construct left the first eighteen exons encoding Inppl1 intact, generating a Inppl1(EX19-28-/-) mouse, and apparently also deleted a second gene, Phox2a. We report a new SHIP2 knockout (Inppl1(-/-)) targeted to the translation-initiating ATG, which is null for Inppl1 mRNA and protein. Inppl1(-/-) mice are viable, have normal glucose and insulin levels, and normal insulin and glucose tolerances. The Inppl1(-/-) mice are, however, highly resistant to weight gain when placed on a high-fat diet. These results suggest that inhibition of SHIP2 would be useful in the effort to ameliorate diet-induced obesity, but call into question a dominant role of SHIP2 in modulating glucose homeostasis.


Assuntos
Gorduras na Dieta/metabolismo , Obesidade/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Animais , Análise Química do Sangue , Peso Corporal , Éxons , Feminino , Deleção de Genes , Genes Reporter , Glucose/metabolismo , Homeostase , Inositol Polifosfato 5-Fosfatases , Insulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases , Monoéster Fosfórico Hidrolases/genética , Transdução de Sinais , Distribuição Tecidual
20.
Proc Natl Acad Sci U S A ; 101(21): 8227-32, 2004 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-15148384

RESUMO

Ghrelin is a recently identified growth hormone (GH) secretogogue whose administration not only induces GH release but also stimulates food intake, increases adiposity, and reduces fat utilization in mice. The effect on food intake appears to be independent of GH release and instead due to direct activation of orexigenic neurons in the arcuate nucleus of the hypothalamus. The effects of ghrelin administration on food intake have led to the suggestion that inhibitors of endogenous ghrelin could be useful in curbing appetite and combating obesity. To further study the role of endogenous ghrelin in appetite and body weight regulation, we generated ghrelin-deficient (ghrl(-/-)) mice, in which the ghrelin gene was precisely replaced with a lacZ reporter gene. ghrl(-/-) mice were viable and exhibited normal growth rates as well as normal spontaneous food intake patterns, normal basal levels of hypothalamic orexigenic and anorexigenic neuropeptides, and no impairment of reflexive hyperphagia after fasting. These results indicate that endogenous ghrelin is not an essential regulator of food intake and has, at most, a redundant role in the regulation of appetite. However, analyses of ghrl(-/-) mice demonstrate that endogenous ghrelin plays a prominent role in determining the type of metabolic substrate (i.e., fat vs. carbohydrate) that is used for maintenance of energy balance, particularly under conditions of high fat intake.


Assuntos
Metabolismo Energético , Comportamento Alimentar/fisiologia , Deleção de Genes , Hormônios Peptídicos/deficiência , Animais , Apetite/fisiologia , Peso Corporal , Metabolismo dos Carboidratos , Ingestão de Alimentos , Jejum , Gorduras/metabolismo , Grelina , Camundongos , Hormônios Peptídicos/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA