Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
EMBO Rep ; 21(11): e50085, 2020 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-33043581

RESUMEN

The cultured brown adipocytes can oxidize glucose in vitro, but it is still not fully clear whether brown adipose tissue (BAT) could completely oxidize glucose in vivo. Although positron emission tomography (PET) with 18 F-fluorodeoxyglucose (18 F-FDG) showed a high level of glucose uptake in the activated BAT, the non-metabolizable 18 F-FDG cannot fully demonstrate intracellular glucose metabolism. Through in vivo [U-13 C]glucose tracing, here we show that chronic cold exposure dramatically activates glucose oxidation in BAT and the browning/beiging subcutaneous white adipose tissue (sWAT). Specifically, chronic cold exposure enhances glucose flux into the mitochondrial TCA cycle. Metabolic flux analysis models that ß3-adrenergic receptor (ß3-AR) agonist significantly enhances the flux of mitochondrial pyruvate uptake through mitochondrial pyruvate carrier (MPC) in the differentiated primary brown adipocytes. Furthermore, in vivo MPC inhibition blocks cold-induced glucose oxidation and impairs body temperature maintenance in mice. Together, mitochondrial pyruvate uptake and oxidation serve an important energy source in the chronic cold exposure activated BAT and beige adipose tissue, which supports a role for glucose oxidation in brown fat thermogenesis.


Asunto(s)
Tejido Adiposo Pardo , Glucosa , Tejido Adiposo Blanco , Animales , Frío , Fluorodesoxiglucosa F18 , Ratones , Termogénesis
2.
J Clin Invest ; 130(1): 247-257, 2020 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-31573981

RESUMEN

Brown adipose tissue (BAT), as the main site of adaptive thermogenesis, exerts beneficial metabolic effects on obesity and insulin resistance. BAT has been previously assumed to contain a homogeneous population of brown adipocytes. Utilizing multiple mouse models capable of genetically labeling different cellular populations, as well as single-cell RNA sequencing and 3D tissue profiling, we discovered a brown adipocyte subpopulation with low thermogenic activity coexisting with the classical high-thermogenic brown adipocytes within the BAT. Compared with the high-thermogenic brown adipocytes, these low-thermogenic brown adipocytes had substantially lower Ucp1 and Adipoq expression, larger lipid droplets, and lower mitochondrial content. Functional analyses showed that, unlike the high-thermogenic brown adipocytes, the low-thermogenic brown adipocytes have markedly lower basal mitochondrial respiration, and they are specialized in fatty acid uptake. Upon changes in environmental temperature, the 2 brown adipocyte subpopulations underwent dynamic interconversions. Cold exposure converted low-thermogenic brown adipocytes into high-thermogenic cells. A thermoneutral environment had the opposite effect. The recruitment of high-thermogenic brown adipocytes by cold stimulation is not affected by high-fat diet feeding, but it does substantially decline with age. Our results revealed a high degree of functional heterogeneity of brown adipocytes.


Asunto(s)
Adipocitos Marrones/metabolismo , Adiponectina/biosíntesis , Tejido Adiposo Pardo/metabolismo , Regulación de la Expresión Génica/fisiología , Termogénesis/fisiología , Proteína Desacopladora 1/biosíntesis , Adipocitos Marrones/citología , Tejido Adiposo Pardo/citología , Animales , Ratones
3.
EBioMedicine ; 24: 64-75, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28988979

RESUMEN

BACKGROUND: IRX3 was recently reported as the effector of the FTO variants. We aimed to test IRX3's roles in the browning program and to evaluate the association between the genetic variants in IRX3 and human obesity. METHODS: IRX3 expression was examined in beige adipocytes in human and mouse models, and further validated in induced beige adipocytes. The browning capacity of primary preadipocytes was assessed with IRX3 knockdown. Luciferase reporter analysis and ChIP assay were applied to investigate IRX3's effects on UCP1 transcriptional activity. Moreover, genetic analysis of IRX3 was performed in 861 young obese subjects and 916 controls. RESULTS: IRX3 expression was induced in the browning process and was positively correlated with the browning markers. IRX3 knockdown remarkably inhibited UCP1 expression in induced mouse and human beige adipocytes, and also repressed the uncoupled oxygen consumption rate. Further, IRX3 directly bound to UCP1 promoter and increased its transcriptional activity. Moreover, 17 rare heterozygous missense/frameshift IRX3 variants were identified, with a significant enrichment in obese subjects (P=0.038, OR=2.27; 95% CI, 1.02-5.05). CONCLUSIONS: IRX3 deficiency repressed the browning program of white adipocytes partially by regulating UCP1 transcriptional activity. Rare variants of IRX3 were associated with human obesity.


Asunto(s)
Adipocitos Marrones/metabolismo , Adipocitos Blancos/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Obesidad/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína Desacopladora 1/genética , Adulto , Animales , Estudios de Casos y Controles , Línea Celular , Modelos Animales de Enfermedad , Femenino , Mutación del Sistema de Lectura , Técnicas de Silenciamiento del Gen , Predisposición Genética a la Enfermedad , Humanos , Masculino , Ratones , Mutación , Mutación Missense , Obesidad/genética , Regiones Promotoras Genéticas , Termogénesis , Proteína Desacopladora 1/metabolismo , Adulto Joven
4.
Endocrine ; 56(2): 366-378, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28243972

RESUMEN

PURPOSE: DGAT2 is the critical catalyzing enzyme for triglyceride biosynthesis, and excess triglyceride accumulation in fat tissues is a fundamental process for obesity. Mutations in DGAT2 or other genes interacting with DGAT2 associated with adiposity have not been reported in human to date. METHODS: DGAT2 mutation was identified based on our in-home database-exome sequencing 227 young obese subjects (body-mass index (BMI), 35.1-61.7 kg/m2) and 219 lean controls (BMI, 17.5-23.0 kg/m2), further validated in 1190 lean subjects and the pedigree of the proband. The trios of the proband were further subjected to whole-exome sequencing to explore the candidate genes for obesity. The mutations in DGAT2 and FAAH were functionally evaluated in vitro. RESULTS: We detected two rare variants in DGAT2 with no significant difference between obese and lean individuals. One novel heterozygous nonsense variant c.382C > T (p.R128*) was identified in one obese subject but not in 219 lean subjects and another 1190 lean subjects. Notably, in vitro study showed that R128* mutation severely damaged the TG-biosynthesis ability of DGAT2, and all other R128* carriers in the pedigree were lean. Thus, we further identified a loss-of-function variant c. 944G > T (p.R315I) in FAAH in the proband inheriting from his obese father. Importantly, FAAH overexpression inhibited DGAT2 expression and TG synthesis, while R315I mutant largely eliminated this inhibitory effect. We first report loss-of-function mutations in DGAT2 and FAAH in one obese subject, which may interact with each other to affect the adiposity penetrance, providing a model of genetic interaction associated with human obesity.


Asunto(s)
Amidohidrolasas/genética , Diacilglicerol O-Acetiltransferasa/genética , Epistasis Genética , Mutación , Obesidad/genética , Adiposidad , Alelos , Animales , Línea Celular , Femenino , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Masculino , Ratones , Linaje
5.
Diabetes ; 66(4): 935-947, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28130309

RESUMEN

Some Shanghai Clinical Center f a role of Niemann-Pick type C1 (NPC1) for obesity traits. However, whether the loss-of-function mutations in NPC1 cause adiposity in humans remains unknown. We recruited 25 probands with rare autosomal-recessive Niemann-Pick type C (NP-C) disease and their parents in assessment of the effect of heterozygous NPC1 mutations on adiposity. We found that male NPC1+/- carriers had a significantly higher BMI than matched control subjects or the whole population-based control subjects. Consistently, male NPC1+/- mice had increased fat storage while eating a high-fat diet. We further conducted an in-depth assessment of rare variants in the NPC1 gene in young, severely obese subjects and lean control subjects and identified 17 rare nonsynonymous/frameshift variants in NPC1 (minor allele frequency <1%) that were significantly associated with an increased risk of obesity (3.40% vs. 0.73%, respectively, in obese patients and control subjects, P = 0.0008, odds ratio = 4.8, 95% CI 1.7-13.2), indicating that rare NPC1 variants were enriched in young, morbidly obese Chinese subjects. Importantly, participants carrying rare variants with severely damaged cholesterol-transporting ability had more fat accumulation than those with mild/no damage rare variants. In summary, rare loss-of-function NPC1 mutations were identified as being associated with human adiposity with a high penetrance, providing potential therapeutic interventions for obesity in addition to the role of NPC1 in the familial NP-C disease.


Asunto(s)
Pueblo Asiatico/genética , Proteínas Portadoras/genética , Dieta Alta en Grasa , Glicoproteínas de Membrana/genética , Obesidad Mórbida/genética , Proteínas/genética , Adulto , Animales , Estudios de Casos y Controles , China , Femenino , Mutación del Sistema de Lectura , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Variación Genética , Heterocigoto , Humanos , Péptidos y Proteínas de Señalización Intracelular , Masculino , Ratones , Persona de Mediana Edad , Proteína Niemann-Pick C1 , Obesidad/genética , Fenotipo , Adulto Joven
6.
Obesity (Silver Spring) ; 25(1): 252-260, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27925416

RESUMEN

OBJECTIVE: To determine the relationship of the gain-of-function variant A750T in leucine-rich repeat containing G protein-coupled receptor 4 (LGR4) with central obesity and related metabolic phenotypes. METHODS: The LGR4 A750T (c.2248 G > A) variant was detected by Sanger sequencing in a discovery young population and a validation community-based population with obesity from eastern China. Fat indices determined by anthropometry and computed tomography scans and clinical biochemical measurements were collected for association analysis. RESULTS: LGR4 A750T was significantly correlated with waist circumference (P = 0.030) and waist-to-height ratio (P < 0.001) in the young cohort (N = 594) and with waist-to-hip ratio (P = 0.013) in the community population (N = 1067). Combined analysis showed a significant correlation of the variant with waist circumference (P < 0.001) and waist-to-hip ratio (P = 0.021). Moreover, the variant had a remarkable correlation with abdominal visceral fat area (P = 0.004) and was associated with 2-h plasma insulin (P = 0.009) and the Matsuda index (P = 0.027) after an oral glucose tolerance test in young subjects with obesity. CONCLUSIONS: The LGR4 A750T variant may contribute to central obesity characterized by abdominal visceral fat accumulation.


Asunto(s)
Variación Genética , Obesidad Abdominal/epidemiología , Obesidad Abdominal/genética , Receptores Acoplados a Proteínas G/genética , Adolescente , Adulto , Glucemia/metabolismo , Presión Sanguínea , Índice de Masa Corporal , China , Colesterol/sangre , Técnicas de Genotipaje , Humanos , Insulina/sangre , Resistencia a la Insulina/genética , Grasa Intraabdominal , Modelos Lineales , Reproducibilidad de los Resultados , Tomografía Computarizada por Rayos X , Circunferencia de la Cintura , Relación Cintura-Cadera , Adulto Joven
7.
Biochem Biophys Res Commun ; 464(2): 396-400, 2015 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-26102032

RESUMEN

Leucine-rich repeat-containing G protein-coupled receptor 4 (Lgr4) is a newfound obese-associated gene. Previous study reveals that heterozygous mutation of Lgr4 correlates with decreased body weight in human. In our recent study, we demonstrate that Lgr4 ablation promotes browning of white adipose tissue and improves whole-body metabolic status. However little is known about its role in other metabolic tissues. Herein, we show that Lgr4 homozygous mutant (Lgr4(m/m)) mice show increased respiratory exchange ratio (RER, closer to 1.0) than wild-type mice at 12:00 AM (food-intake time for mice) while decreased RER (closer to 0.75) at 12:00 PM (fasting for mice), indicating a glucose-prone versus fatty acid-prone metabolic pattern, respectively. Furthermore, Lgr4 ablation increases lipid oxidation-related gene expression while suppresses glucose transporter type 4 (Glut4) levels in skeletal muscle under fasting condition. These data suggest that Lgr4 ablation enhances the flexibility of skeletal muscle to switch energy provider from glucose to fatty acid in response to glucose depletion. We further reveal the activation of Ampk/Sirt1/Pgc1α pathway during this adaptive fuel shift due to Lgr4 ablation. This study suggests that Lgr4 might serve as an adaptive regulator between glucose and lipid metabolism in skeletal muscle and reveals a potentially new regulator for a well-established adaptive network.


Asunto(s)
Adenilato Quinasa/metabolismo , Metabolismo Energético , Músculo Esquelético/metabolismo , Receptores Acoplados a Proteínas G/fisiología , Sirtuina 1/metabolismo , Factores de Transcripción/metabolismo , Animales , Ayuno , Insulina/metabolismo , Ratones , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Receptores Acoplados a Proteínas G/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...