Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Nature ; 601(7893): 446-451, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34937935

RESUMEN

Exosomes and other small extracellular vesicles (sEVs) provide a unique mode of cell-to-cell communication in which microRNAs (miRNAs) produced and released from one cell are taken up by cells at a distance where they can enact changes in gene expression1-3. However, the mechanism by which miRNAs are sorted into exosomes/sEVs or retained in cells remains largely unknown. Here we demonstrate that miRNAs possess sorting sequences that determine their secretion in sEVs (EXOmotifs) or cellular retention (CELLmotifs) and that different cell types, including white and brown adipocytes, endothelium, liver and muscle, make preferential use of specific sorting sequences, thus defining the sEV miRNA profile of that cell type. Insertion or deletion of these CELLmotifs or EXOmotifs in a miRNA increases or decreases retention in the cell of production or secretion into exosomes/sEVs. Two RNA-binding proteins, Alyref and Fus, are involved in the export of miRNAs carrying one of the strongest EXOmotifs, CGGGAG. Increased miRNA delivery mediated by EXOmotifs leads to enhanced inhibition of target genes in distant cells. Thus, this miRNA code not only provides important insights that link circulating exosomal miRNAs to tissues of origin, but also provides an approach for improved targeting in RNA-mediated therapies.


Asunto(s)
Vesículas Extracelulares , MicroARNs , Adipocitos/citología , Comunicación Celular , Endotelio/citología , Exosomas/genética , Exosomas/metabolismo , Vesículas Extracelulares/genética , Vesículas Extracelulares/metabolismo , Hígado/citología , MicroARNs/genética , MicroARNs/metabolismo , Músculos/citología
2.
Mol Cell ; 74(4): 844-857.e7, 2019 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-31000437

RESUMEN

Brown adipose tissue (BAT) is rich in mitochondria and plays important roles in energy expenditure, thermogenesis, and glucose homeostasis. We find that levels of mitochondrial protein succinylation and malonylation are high in BAT and subject to physiological and genetic regulation. BAT-specific deletion of Sirt5, a mitochondrial desuccinylase and demalonylase, results in dramatic increases in global protein succinylation and malonylation. Mass spectrometry-based quantification of succinylation reveals that Sirt5 regulates the key thermogenic protein in BAT, UCP1. Mutation of the two succinylated lysines in UCP1 to acyl-mimetic glutamine and glutamic acid significantly decreases its stability and activity. The reduced function of UCP1 and other proteins in Sirt5KO BAT results in impaired mitochondria respiration, defective mitophagy, and metabolic inflexibility. Thus, succinylation of UCP1 and other mitochondrial proteins plays an important role in BAT and in regulation of energy homeostasis.


Asunto(s)
Metabolismo Energético/genética , Mitocondrias/metabolismo , Obesidad/genética , Sirtuinas/genética , Proteína Desacopladora 1/genética , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Pardo/patología , Animales , Regulación de la Expresión Génica , Glucosa/metabolismo , Ratones , Ratones Noqueados , Mitocondrias/genética , Proteínas Mitocondriales/genética , Obesidad/metabolismo , Obesidad/patología , Proteómica/métodos , Ácido Succínico/metabolismo , Termogénesis/genética , Proteína Desacopladora 1/metabolismo
3.
Proc Natl Acad Sci U S A ; 121(11): e2316544121, 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38442155

RESUMEN

Muscle regeneration is a complex process relying on precise teamwork between multiple cell types, including muscle stem cells (MuSCs) and fibroadipogenic progenitors (FAPs). FAPs are also the main source of intramuscular adipose tissue (IMAT). Muscles without FAPs exhibit decreased IMAT infiltration but also deficient muscle regeneration, indicating the importance of FAPs in the repair process. Here, we demonstrate the presence of bidirectional crosstalk between FAPs and MuSCs via their secretion of extracellular vesicles (EVs) containing distinct clusters of miRNAs that is crucial for normal muscle regeneration. Thus, after acute muscle injury, there is activation of FAPs leading to a transient rise in IMAT. These FAPs also release EVs enriched with a selected group of miRNAs, a number of which come from an imprinted region on chromosome 12. The most abundant of these is miR-127-3p, which targets the sphingosine-1-phosphate receptor S1pr3 and activates myogenesis. Indeed, intramuscular injection of EVs from immortalized FAPs speeds regeneration of injured muscle. In late stages of muscle repair, in a feedback loop, MuSCs and their derived myoblasts/myotubes secrete EVs enriched in miR-206-3p and miR-27a/b-3p. The miRNAs repress FAP adipogenesis, allowing full muscle regeneration. Together, the reciprocal communication between FAPs and muscle cells via miRNAs in their secreted EVs plays a critical role in limiting IMAT infiltration while stimulating muscle regeneration, hence providing an important mechanism for skeletal muscle repair and homeostasis.


Asunto(s)
Vesículas Extracelulares , MicroARNs , Células Satélite del Músculo Esquelético , Fibras Musculares Esqueléticas , Comunicación , MicroARNs/genética , Regeneración/genética
4.
Mol Cell ; 55(3): 372-82, 2014 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-25002143

RESUMEN

Brown and beige/brite fats generate heat via uncoupled respiration to defend against cold. The total mass and activity of thermogenic adipose tissues are also tightly linked to systemic energy and nutrient homeostasis. Despite originating from distinct progenitors, brown and beige adipocytes acquire remarkably similar molecular and metabolic characteristics during differentiation through the action of a network of transcription factors and cofactors. How this regulatory network interfaces with long noncoding RNAs (lncRNAs), an emerging class of developmental regulators, remains largely unexplored. Here, we globally profiled lncRNA gene expression during thermogenic adipocyte formation and identified Brown fat lncRNA 1 (Blnc1) as a nuclear lncRNA that promotes brown and beige adipocyte differentiation and function. Blnc1 forms a ribonucleoprotein complex with transcription factor EBF2 to stimulate the thermogenic gene program. Further, Blnc1 itself is a target of EBF2, thereby forming a feedforward regulatory loop to drive adipogenesis toward thermogenic phenotype.


Asunto(s)
Adipogénesis , Tejido Adiposo Pardo/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , ARN Largo no Codificante/metabolismo , Animales , Línea Celular , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Células HEK293 , Humanos , Masculino , Ratones Endogámicos C57BL , ARN Largo no Codificante/genética , Ribonucleoproteínas/metabolismo
5.
Proc Natl Acad Sci U S A ; 114(34): E7111-E7120, 2017 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-28784777

RESUMEN

Brown and beige adipocytes convert chemical energy into heat through uncoupled respiration to defend against cold stress. Beyond thermogenesis, brown and beige fats engage other metabolic tissues via secreted factors to influence systemic energy metabolism. How the protein and long noncoding RNA (lncRNA) regulatory networks act in concert to regulate key aspects of thermogenic adipocyte biology remains largely unknown. Here we developed a genome-wide functional screen to interrogate the transcription factors and cofactors in thermogenic gene activation and identified zinc finger and BTB domain-containing 7b (Zbtb7b) as a potent driver of brown fat development and thermogenesis and cold-induced beige fat formation. Zbtb7b is required for activation of the thermogenic gene program in brown and beige adipocytes. Genetic ablation of Zbtb7b impaired cold-induced transcriptional remodeling in brown fat, rendering mice sensitive to cold temperature, and diminished browning of inguinal white fat. Proteomic analysis revealed a mechanistic link between Zbtb7b and the lncRNA regulatory pathway through which Zbtb7b recruits the brown fat lncRNA 1 (Blnc1)/heterogeneous nuclear ribonucleoprotein U (hnRNPU) ribonucleoprotein complex to activate thermogenic gene expression in adipocytes. These findings illustrate the emerging concept of a protein-lncRNA regulatory network in the control of adipose tissue biology and energy metabolism.


Asunto(s)
Tejido Adiposo Beige/metabolismo , Tejido Adiposo Pardo/metabolismo , Proteínas de Unión al ADN/metabolismo , Termogénesis , Factores de Transcripción/metabolismo , Adipocitos/citología , Adipocitos/metabolismo , Tejido Adiposo Beige/crecimiento & desarrollo , Tejido Adiposo Pardo/crecimiento & desarrollo , Animales , Células Cultivadas , Proteínas de Unión al ADN/genética , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Largo no Codificante , Factores de Transcripción/genética
6.
Diabetes ; 70(8): 1857-1873, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34031123

RESUMEN

The brain is now recognized as an insulin-sensitive tissue; however, the role of changing insulin concentrations in the peripheral circulation in gene expression in the brain is largely unknown. Here, we performed a hyperinsulinemic-euglycemic clamp on 3-month-old male C57BL/6 mice for 3 h. We show that, in comparison with results in saline-infused controls, increases in peripheral insulin within the physiological range regulate expression of a broad network of genes in the brain. Insulin regulates distinct pathways in the hypothalamus (HTM), hippocampus, and nucleus accumbens. Insulin shows its most robust effect in the HTM and regulates multiple genes involved in neurotransmission, including upregulating expression of multiple subunits of GABA-A receptors, Na+ and K+ channels, and SNARE proteins; differentially modulating glutamate receptors; and suppressing multiple neuropeptides. Insulin also strongly modulates metabolic genes in the HTM, suppressing genes in the glycolysis and pentose phosphate pathways, while increasing expression of genes regulating pyruvate dehydrogenase and long-chain fatty acyl-CoA and cholesterol biosynthesis, thereby rerouting of carbon substrates from glucose metabolism to lipid metabolism required for the biogenesis of membranes for neuronal and glial function and synaptic remodeling. Furthermore, based on the transcriptional signatures, these changes in gene expression involve neurons, astrocytes, oligodendrocytes, microglia, and endothelial cells. Thus, peripheral insulin acutely and potently regulates expression of a broad network of genes involved in neurotransmission and brain metabolism. Dysregulation of these pathways could have dramatic effects in normal physiology and diabetes.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Hipocampo/metabolismo , Hipotálamo/metabolismo , Insulina/farmacología , Lipogénesis/fisiología , Núcleo Accumbens/metabolismo , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Técnica de Clampeo de la Glucosa , Hipocampo/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Lipogénesis/efectos de los fármacos , Masculino , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Núcleo Accumbens/efectos de los fármacos
7.
Diabetes ; 69(11): 2294-2309, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32868340

RESUMEN

Skeletal muscle insulin resistance is a prominent early feature in the pathogenesis of type 2 diabetes. In attempt to overcome this defect, we generated mice overexpressing insulin receptors (IR) specifically in skeletal muscle (IRMOE). On normal chow, IRMOE mice have body weight similar to that of controls but an increase in lean mass and glycolytic muscle fibers and reduced fat mass. IRMOE mice also show higher basal phosphorylation of IR, IRS-1, and Akt in muscle and improved glucose tolerance compared with controls. When challenged with high-fat diet (HFD), IRMOE mice are protected from diet-induced obesity. This is associated with reduced inflammation in fat and liver, improved glucose tolerance, and improved systemic insulin sensitivity. Surprisingly, however, in both chow and HFD-fed mice, insulin-stimulated Akt phosphorylation is significantly reduced in muscle of IRMOE mice, indicating postreceptor insulin resistance. RNA sequencing reveals downregulation of several postreceptor signaling proteins that contribute to this resistance. Thus, enhancing early insulin signaling in muscle by overexpression of the IR protects mice from diet-induced obesity and its effects on glucose metabolism. However, chronic overstimulation of this pathway leads to postreceptor desensitization, indicating the critical balance between normal signaling and hyperstimulation of the insulin signaling pathway.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Regulación de la Expresión Génica/efectos de los fármacos , Intolerancia a la Glucosa/inducido químicamente , Resistencia a la Insulina/fisiología , Receptor de Insulina/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Composición Corporal , Grasas de la Dieta/farmacología , Metabolismo Energético , Técnica de Clampeo de la Glucosa , Hígado/metabolismo , Ratones , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/fisiología , Obesidad/inducido químicamente , Receptor de Insulina/genética , Análisis de Secuencia de ARN
8.
J Clin Invest ; 129(10): 3990-4000, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31573548

RESUMEN

Over the past decade, great progress has been made in understanding the complexity of adipose tissue biology and its role in metabolism. This includes new insights into the multiple layers of adipose tissue heterogeneity, not only differences between white and brown adipocytes, but also differences in white adipose tissue at the depot level and even heterogeneity of white adipocytes within a single depot. These inter- and intra-depot differences in adipocytes are developmentally programmed and contribute to the wide range of effects observed in disorders with fat excess (overweight/obesity) or fat loss (lipodystrophy). Recent studies also highlight the underappreciated dynamic nature of adipose tissue, including potential to undergo rapid turnover and dedifferentiation and as a source of stem cells. Finally, we explore the rapidly expanding field of adipose tissue as an endocrine organ, and how adipose tissue communicates with other tissues to regulate systemic metabolism both centrally and peripherally through secretion of adipocyte-derived peptide hormones, inflammatory mediators, signaling lipids, and miRNAs packaged in exosomes. Together these attributes and complexities create a robust, multidimensional signaling network that is central to metabolic homeostasis.


Asunto(s)
Adipocitos/fisiología , Tejido Adiposo/fisiopatología , Síndrome Metabólico/etiología , Adipocitos/patología , Adipoquinas/fisiología , Tejido Adiposo/patología , Animales , Exosomas/metabolismo , Humanos , Resistencia a la Insulina/fisiología , Metabolismo de los Lípidos , Lipodistrofia/etiología , Lipodistrofia/patología , Lipodistrofia/fisiopatología , Síndrome Metabólico/patología , Síndrome Metabólico/fisiopatología , MicroARNs/metabolismo , Transducción de Señal
9.
Cell Metab ; 30(4): 735-753.e4, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31577934

RESUMEN

Dietary sugars, fructose and glucose, promote hepatic de novo lipogenesis and modify the effects of a high-fat diet (HFD) on the development of insulin resistance. Here, we show that fructose and glucose supplementation of an HFD exert divergent effects on hepatic mitochondrial function and fatty acid oxidation. This is mediated via three different nodes of regulation, including differential effects on malonyl-CoA levels, effects on mitochondrial size/protein abundance, and acetylation of mitochondrial proteins. HFD- and HFD plus fructose-fed mice have decreased CTP1a activity, the rate-limiting enzyme of fatty acid oxidation, whereas knockdown of fructose metabolism increases CPT1a and its acylcarnitine products. Furthermore, fructose-supplemented HFD leads to increased acetylation of ACADL and CPT1a, which is associated with decreased fat metabolism. In summary, dietary fructose, but not glucose, supplementation of HFD impairs mitochondrial size, function, and protein acetylation, resulting in decreased fatty acid oxidation and development of metabolic dysregulation.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Azúcares de la Dieta/efectos adversos , Ácidos Grasos/metabolismo , Fructosa/efectos adversos , Hígado/metabolismo , Proteínas Mitocondriales , Obesidad/metabolismo , Animales , Línea Celular , Glucosa/efectos adversos , Lipogénesis , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Procesamiento Proteico-Postraduccional , Transcripción Genética
11.
Mol Metab ; 6(8): 863-872, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28752050

RESUMEN

OBJECTIVE: Brown and white adipose tissue exerts pleiotropic effects on systemic energy metabolism in part by releasing endocrine factors. Neuregulin 4 (Nrg4) was recently identified as a brown fat-enriched secreted factor that ameliorates diet-induced metabolic disorders, including insulin resistance and hepatic steatosis. However, the physiological mechanisms through which Nrg4 regulates energy balance and glucose and lipid metabolism remain incompletely understood. The aims of the current study were: i) to investigate the regulation of adipose Nrg4 expression during obesity and the physiological signals involved, ii) to elucidate the mechanisms underlying Nrg4 regulation of energy balance and glucose and lipid metabolism, and iii) to explore whether Nrg4 regulates adipose tissue secretome gene expression and adipokine secretion. METHODS: We examined the correlation of adipose Nrg4 expression with obesity in a cohort of diet-induced obese mice and investigated the upstream signals that regulate Nrg4 expression. We performed metabolic cage and hyperinsulinemic-euglycemic clamp studies in Nrg4 transgenic mice to dissect the metabolic pathways regulated by Nrg4. We investigated how Nrg4 regulates hepatic lipid metabolism in the fasting state and explored the effects of Nrg4 on adipose tissue gene expression, particularly those encoding secreted factors. RESULTS: Adipose Nrg4 expression is inversely correlated with adiposity and regulated by pro-inflammatory and anti-inflammatory signaling. Transgenic expression of Nrg4 increases energy expenditure and augments whole body glucose metabolism. Nrg4 protects mice from diet-induced hepatic steatosis in part through activation of hepatic fatty acid oxidation and ketogenesis. Finally, Nrg4 promotes a healthy adipokine profile during obesity. CONCLUSIONS: Nrg4 exerts pleiotropic beneficial effects on energy balance and glucose and lipid metabolism to ameliorate obesity-associated metabolic disorders. Biologic therapeutics based on Nrg4 may improve both type 2 diabetes and non-alcoholic fatty liver disease (NAFLD) in patients.


Asunto(s)
Adipoquinas/sangre , Ácidos Grasos/metabolismo , Neurregulinas/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Obesidad/metabolismo , Adipocitos/metabolismo , Animales , Células Cultivadas , Dieta Alta en Grasa/efectos adversos , Metabolismo Energético , Glucosa/metabolismo , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Neurregulinas/genética , Enfermedad del Hígado Graso no Alcohólico/etiología , Obesidad/etiología
12.
J Clin Invest ; 127(11): 4059-4074, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-28972537

RESUMEN

Overconsumption of high-fat diet (HFD) and sugar-sweetened beverages are risk factors for developing obesity, insulin resistance, and fatty liver disease. Here we have dissected mechanisms underlying this association using mice fed either chow or HFD with or without fructose- or glucose-supplemented water. In chow-fed mice, there was no major physiological difference between fructose and glucose supplementation. On the other hand, mice on HFD supplemented with fructose developed more pronounced obesity, glucose intolerance, and hepatomegaly as compared to glucose-supplemented HFD mice, despite similar caloric intake. Fructose and glucose supplementation also had distinct effects on expression of the lipogenic transcription factors ChREBP and SREBP1c. While both sugars increased ChREBP-ß, fructose supplementation uniquely increased SREBP1c and downstream fatty acid synthesis genes, resulting in reduced liver insulin signaling. In contrast, glucose enhanced total ChREBP expression and triglyceride synthesis but was associated with improved hepatic insulin signaling. Metabolomic and RNA sequence analysis confirmed dichotomous effects of fructose and glucose supplementation on liver metabolism in spite of inducing similar hepatic lipid accumulation. Ketohexokinase, the first enzyme of fructose metabolism, was increased in fructose-fed mice and in obese humans with steatohepatitis. Knockdown of ketohexokinase in liver improved hepatic steatosis and glucose tolerance in fructose-supplemented mice. Thus, fructose is a component of dietary sugar that is distinctively associated with poor metabolic outcomes, whereas increased glucose intake may be protective.


Asunto(s)
Fructosa/farmacología , Glucosa/farmacología , Insulina/fisiología , Lipogénesis/efectos de los fármacos , Hígado/efectos de los fármacos , Adolescente , Animales , Dieta Alta en Grasa/efectos adversos , Inducción Enzimática , Ácidos Grasos/biosíntesis , Fructoquinasas/genética , Fructoquinasas/metabolismo , Intolerancia a la Glucosa , Humanos , Resistencia a la Insulina , Hígado/enzimología , Masculino , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/enzimología , Enfermedad del Hígado Graso no Alcohólico/etiología , Obesidad/enzimología , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Activación Transcripcional , Transcriptoma , Regulación hacia Arriba
13.
Trends Endocrinol Metab ; 26(5): 231-7, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25843910

RESUMEN

Brown fat is highly active in fuel oxidation and dissipates chemical energy through uncoupling protein (UCP)1-mediated heat production. Activation of brown fat leads to increased energy expenditure, reduced adiposity, and lower plasma glucose and lipid levels, thus contributing to better homeostasis. Uncoupled respiration and thermogenesis have been considered to be responsible for the metabolic benefits of brown adipose tissue. Recent studies have demonstrated that brown adipocytes also secrete factors that act locally and systemically to influence fuel and energy metabolism. This review discusses the evidence supporting a thermogenesis-independent role of brown fat, particularly through its release of secreted factors, and their implications in physiology and therapeutic development.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Termogénesis , Adipocitos Marrones/metabolismo , Adiponectina/fisiología , Tejido Adiposo Pardo/inervación , Animales , Proteínas Morfogenéticas Óseas/fisiología , Metabolismo Energético , Factores de Crecimiento de Fibroblastos/fisiología , Homeostasis , Humanos , Canales Iónicos/fisiología , Proteínas Mitocondriales/fisiología , Factor de Crecimiento Nervioso/fisiología , Neurregulinas/fisiología , Obesidad , Proteína Desacopladora 1 , Factor A de Crecimiento Endotelial Vascular/fisiología
14.
Nat Med ; 20(12): 1436-1443, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25401691

RESUMEN

Brown fat activates uncoupled respiration in response to cold temperature and contributes to systemic metabolic homeostasis. To date, the metabolic action of brown fat has been primarily attributed to its role in fuel oxidation and uncoupling protein 1 (UCP1)-mediated thermogenesis. Whether brown fat engages other tissues through secreted factors remains largely unexplored. Here we show that neuregulin 4 (Nrg4), a member of the epidermal growth factor (EGF) family of extracellular ligands, is highly expressed in adipose tissues, enriched in brown fat and markedly increased during brown adipocyte differentiation. Adipose tissue Nrg4 expression was reduced in rodent and human obesity. Gain- and loss-of-function studies in mice demonstrated that Nrg4 protects against diet-induced insulin resistance and hepatic steatosis through attenuating hepatic lipogenic signaling. Mechanistically, Nrg4 activates ErbB3 and ErbB4 signaling in hepatocytes and negatively regulates de novo lipogenesis mediated by LXR and SREBP1c in a cell-autonomous manner. These results establish Nrg4 as a brown fat-enriched endocrine factor with therapeutic potential for the treatment of obesity-associated disorders, including type 2 diabetes and nonalcoholic fatty liver disease (NAFLD).


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Lipogénesis , Hígado/metabolismo , Neurregulinas/genética , Obesidad/genética , ARN Mensajero/genética , Receptor ErbB-3/metabolismo , Receptor ErbB-4/metabolismo , Células 3T3-L1 , Adipocitos Marrones/metabolismo , Adipogénesis , Animales , Hígado Graso/genética , Hígado Graso/metabolismo , Células HEK293 , Humanos , Resistencia a la Insulina , Receptores X del Hígado , Ratones , Neurregulinas/metabolismo , Obesidad/metabolismo , Receptores Nucleares Huérfanos/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo
15.
Diabetes ; 63(4): 1340-52, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24379350

RESUMEN

Chronic low-grade inflammation is emerging as a pathogenic link between obesity and metabolic disease. Persistent immune activation in white adipose tissue (WAT) impairs insulin sensitivity and systemic metabolism, in part, through the actions of proinflammatory cytokines. Whether obesity engages an adaptive mechanism to counteract chronic inflammation in adipose tissues has not been elucidated. Here we identified otopetrin 1 (Otop1) as a component of a counterinflammatory pathway that is induced in WAT during obesity. Otop1 expression is markedly increased in obese mouse WAT and is stimulated by tumor necrosis factor-α in cultured adipocytes. Otop1 mutant mice respond to high-fat diet with pronounced insulin resistance and hepatic steatosis, accompanied by augmented adipose tissue inflammation. Otop1 attenuates interferon-γ (IFN-γ) signaling in adipocytes through selective downregulation of the transcription factor STAT1. Using a tagged vector, we found that Otop1 physically interacts with endogenous STAT1. Thus, Otop1 defines a unique target of cytokine signaling that attenuates obesity-induced adipose tissue inflammation and plays an adaptive role in maintaining metabolic homeostasis in obesity.


Asunto(s)
Tejido Adiposo/patología , Inflamación/prevención & control , Proteínas de la Membrana/farmacología , Obesidad/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Dieta Alta en Grasa , Homeostasis/efectos de los fármacos , Inflamación/inmunología , Resistencia a la Insulina/fisiología , Interferón gamma/efectos de los fármacos , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Factor de Transcripción STAT1/metabolismo
16.
PLoS One ; 8(8): e70109, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23990898

RESUMEN

The biological clock plays an important role in integrating nutrient and energy metabolism with other cellular processes. Previous studies have demonstrated that core clock genes are rhythmically expressed in peripheral tissues, including the liver, skeletal muscle, pancreatic islets, and white and brown adipose tissues. These peripheral clocks are entrained by physiological cues, thereby aligning the circadian pacemaker to tissue functions. The mechanisms that regulate brown adipose tissue clock in response to physiological signals remain poorly understood. Here we found that the expression of core clock genes is highly responsive to cold exposure in brown fat, but not in white fat. This cold-inducible regulation of the clock network is mediated by adrenergic receptor activation and the transcriptional coactivator PGC-1α. Brown adipocytes in mice lacking a functional clock contain large lipid droplets accompanied by dysregulation of genes involved in lipid metabolism and adaptive thermogenesis. Paradoxically, the "clockless" mice were competent in maintaining core body temperature during cold exposure. These studies elucidated the presence of adrenergic receptor/clock crosstalk that appears to be required for normal thermogenic gene expression in brown fat.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Relojes Biológicos , Regulación de la Expresión Génica , Receptores Adrenérgicos/metabolismo , Transducción de Señal , Termogénesis , Factores de Transcripción ARNTL/metabolismo , Adipocitos/citología , Animales , Temperatura Corporal , Diferenciación Celular , Ritmo Circadiano , Frío , Dioxoles/farmacología , Perfilación de la Expresión Génica , Lípidos/química , Masculino , Ratones , Ratones Noqueados , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Reacción en Cadena de la Polimerasa , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda