Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 11(1): 2117, 2020 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-32355218

RESUMEN

White adipose tissue plays an important role in physiological homeostasis and metabolic disease. Different fat depots have distinct metabolic and inflammatory profiles and are differentially associated with disease risk. It is unclear whether these differences are intrinsic to the pre-differentiated stage. Using single-cell RNA sequencing, a unique network methodology and a data integration technique, we predict metabolic phenotypes in differentiating cells. Single-cell RNA-seq profiles of human preadipocytes during adipogenesis in vitro identifies at least two distinct classes of subcutaneous white adipocytes. These differences in gene expression are separate from the process of browning and beiging. Using a systems biology approach, we identify a new network of zinc-finger proteins that are expressed in one class of preadipocytes and is potentially involved in regulating adipogenesis. Our findings gain a deeper understanding of both the heterogeneity of white adipocytes and their link to normal metabolism and disease.


Asunto(s)
Adipocitos Blancos/citología , Adipogénesis , Diferenciación Celular/genética , Análisis de la Célula Individual , Transcripción Genética , Células Cultivadas , Análisis por Conglomerados , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Glucosa/metabolismo , Humanos , Consumo de Oxígeno , Fenotipo , Reacción en Cadena de la Polimerasa , Mapeo de Interacción de Proteínas , Análisis de Secuencia de ARN , Biología de Sistemas
2.
Nat Commun ; 11(1): 1421, 2020 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-32184391

RESUMEN

Uncoupling protein-1 (UCP1) plays a central role in energy dissipation in brown adipose tissue (BAT). Using high-throughput library screening of secreted peptides, we identify two fibroblast growth factors (FGF), FGF6 and FGF9, as potent inducers of UCP1 expression in adipocytes and preadipocytes. Surprisingly, this occurs through a mechanism independent of adipogenesis and involves FGF receptor-3 (FGFR3), prostaglandin-E2 and interaction between estrogen receptor-related alpha, flightless-1 (FLII) and leucine-rich-repeat-(in FLII)-interacting-protein-1 as a regulatory complex for UCP1 transcription. Physiologically, FGF6/9 expression in adipose is upregulated by exercise and cold in mice, and FGF9/FGFR3 expression in human neck fat is significantly associated with UCP1 expression. Loss of FGF9 impairs BAT thermogenesis. In vivo administration of FGF9 increases UCP1 expression and thermogenic capacity. Thus, FGF6 and FGF9 are adipokines that can regulate UCP1 through a transcriptional network that is dissociated from brown adipogenesis, and act to modulate systemic energy metabolism.


Asunto(s)
Adipocitos Marrones/metabolismo , Adipogénesis , Factor 6 de Crecimiento de Fibroblastos/metabolismo , Factor 9 de Crecimiento de Fibroblastos/metabolismo , Obesidad/metabolismo , Proteína Desacopladora 1/metabolismo , Adipocitos Marrones/citología , Tejido Adiposo Pardo/citología , Tejido Adiposo Pardo/metabolismo , Animales , Factor 6 de Crecimiento de Fibroblastos/genética , Factor 9 de Crecimiento de Fibroblastos/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/genética , Obesidad/fisiopatología , Termogénesis , Proteína Desacopladora 1/genética
3.
Nat Metab ; 1(2): 291-303, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-31032475

RESUMEN

Exercise improves health and well-being across diverse organ systems, and elucidating mechanisms underlying the beneficial effects of exercise can lead to new therapies. Here, we show that transforming growth factor-ß2 (TGF-ß2) is secreted from adipose tissue in response to exercise and improves glucose tolerance in mice. We identify TGF-ß2 as an exercise-induced adipokine in a gene expression analysis of human subcutaneous adipose tissue biopsies after exercise training. In mice, exercise training increases TGF-ß2 in scWAT, serum, and its secretion from fat explants. Transplanting scWAT from exercise-trained wild type mice, but not from adipose tissue-specific Tgfb2-/- mice, into sedentary mice improves glucose tolerance. TGF-ß2 treatment reverses the detrimental metabolic effects of high fat feeding in mice. Lactate, a metabolite released from muscle during exercise, stimulates TGF-ß2 expression in human adipocytes. Administration of the lactate-lowering agent dichloroacetate during exercise training in mice decreases circulating TGF-ß2 levels and reduces exercise-stimulated improvements in glucose tolerance. Thus, exercise training improves systemic metabolism through inter-organ communication with fat via a lactate-TGF-ß2-signaling cycle.


Asunto(s)
Adipoquinas/metabolismo , Ácidos Grasos/metabolismo , Glucosa/metabolismo , Condicionamiento Físico Animal , Factor de Crecimiento Transformador beta2/metabolismo , Tejido Adiposo/metabolismo , Animales , Ratones
4.
Cell Rep ; 21(11): 3040-3048, 2017 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-29241534

RESUMEN

White adipocytes are specialized for energy storage, whereas brown adipocytes are specialized for energy expenditure. Explicating this difference can help identify therapeutic targets for obesity. A common tool to assess metabolic differences between such cells is the Seahorse Extracellular Flux (XF) Analyzer, which measures oxygen consumption and media acidification in the presence of different substrates and perturbagens. Here, we integrate the Analyzer's metabolic profile from human white and brown adipocytes with a genome-scale metabolic model to predict flux differences across the metabolic map. Predictions matched experimental data for the metabolite 4-aminobutyrate, the protein ABAT, and the fluxes for glucose, glutamine, and palmitate. We also uncovered a difference in how adipocytes dispose of nitrogenous waste, with brown adipocytes secreting less ammonia and more urea than white adipocytes. Thus, the method and software we developed allow for broader metabolic phenotyping and provide a distinct approach to uncovering metabolic differences.


Asunto(s)
Adipocitos Marrones/metabolismo , Adipocitos Blancos/metabolismo , Metabolismo Energético/genética , Genoma Humano , Redes y Vías Metabólicas/genética , 4-Aminobutirato Transaminasa/metabolismo , Adipocitos Marrones/citología , Adipocitos Blancos/citología , Amoníaco/metabolismo , Línea Celular Transformada , Glucosa/metabolismo , Glutamina/metabolismo , Homeostasis , Humanos , Metaboloma/genética , Especificidad de Órganos , Consumo de Oxígeno/genética , Ácido Palmítico/metabolismo , Cultivo Primario de Células , Programas Informáticos , Urea/metabolismo , Ácido gamma-Aminobutírico/metabolismo
5.
Sci Rep ; 7(1): 1383, 2017 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-28469146

RESUMEN

The identification of brown adipose deposits in adults has led to significant interest in targeting this metabolically active tissue for treatment of obesity and diabetes. Improved methods for the direct measurement of heat production as the signature function of brown adipocytes (BAs), particularly at the single cell level, would be of substantial benefit to these ongoing efforts. Here, we report the first application of a small molecule-type thermosensitive fluorescent dye, ERthermAC, to monitor thermogenesis in BAs derived from murine brown fat precursors and in human brown fat cells differentiated from human neck brown preadipocytes. ERthermAC accumulated in the endoplasmic reticulum of BAs and displayed a marked change in fluorescence intensity in response to adrenergic stimulation of cells, which corresponded to temperature change. ERthermAC fluorescence intensity profiles were congruent with mitochondrial depolarisation events visualised by the JC-1 probe. Moreover, the averaged fluorescence intensity changes across a population of cells correlated well with dynamic changes such as thermal power, oxygen consumption, and extracellular acidification rates. These findings suggest ERthermAC as a promising new tool for studying thermogenic function in brown adipocytes of both murine and human origins.


Asunto(s)
Adipocitos Marrones/metabolismo , Tejido Adiposo Pardo/metabolismo , Termogénesis , Termografía/métodos , Animales , Células Cultivadas , Retículo Endoplásmico/metabolismo , Colorantes Fluorescentes , Humanos , Ratones , Análisis de la Célula Individual , Termografía/instrumentación
6.
Endocrinology ; 158(3): 578-591, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-27967236

RESUMEN

Obesity has become the most common metabolic disorder worldwide. Promoting brown adipose tissue (BAT) and beige adipose tissue formation, and therefore, a functional increase in energy expenditure, may counteract obesity. Mice lacking type IIß regulatory subunit of adenosine 3',5' cyclic monophosphate (cAMP)-dependent protein kinase A (PKA-RIIB) display reduced adiposity and resistance to diet-induced obesity. PKA-RIIB, encoded by the Prkar2b gene, is most abundant in BAT and white adipose tissue (WAT) and in the brain. In this study, we show that mice lacking PKA-RIIB have increased energy expenditure, limited weight gain, and improved glucose metabolism. PKA-RIIB deficiency induces brownlike adipocyte in inguinal WAT (iWAT). PKA-RIIB deficiency also increases the expression of uncoupling protein 1 and other thermogenic genes in iWAT and primary preadipocytes from iWAT through a mechanism involving increased PKA activity, which is represented by increased phosphorylation of PKA substrate, cAMP response element binding protein, and P38 mitogen-activated protein kinase. Our study provides evidence for the role of PKA-RIIB deficiency in regulating thermogenesis in WAT, which may potentially have therapeutic implications for the treatment of obesity and related metabolic disorders.


Asunto(s)
Adipocitos Marrones , Tejido Adiposo Blanco/citología , Subunidad RIIbeta de la Proteína Quinasa Dependiente de AMP Cíclico/deficiencia , Metabolismo Energético , Termogénesis , Animales , Metabolismo de los Hidratos de Carbono , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Epidídimo/citología , Glucosa/metabolismo , Metabolismo de los Lípidos , Masculino , Ratones , Obesidad/metabolismo , Proteína Desacopladora 1/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
7.
Diabetologia ; 59(8): 1769-77, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27209464

RESUMEN

AIMS/HYPOTHESIS: Adipose tissue dysfunction is a prime risk factor for the development of metabolic disease. Bone morphogenetic proteins (BMPs) have previously been implicated in adipocyte formation. Here, we investigate the role of BMP signalling in adipose tissue health and systemic glucose homeostasis. METHODS: We employed the Cre/loxP system to generate mouse models with conditional ablation of BMP receptor 1A in differentiating and mature adipocytes, as well as tissue-resident myeloid cells. Metabolic variables were assessed by glucose and insulin tolerance testing, insulin-stimulated glucose uptake and gene expression analysis. RESULTS: Conditional deletion of Bmpr1a using the aP2 (also known as Fabp4)-Cre strain resulted in a complex phenotype. Knockout mice were clearly resistant to age-related impairment of insulin sensitivity during normal and high-fat-diet feeding and showed significantly improved insulin-stimulated glucose uptake in brown adipose tissue and skeletal muscle. Moreover, knockouts displayed significant reduction of variables of adipose tissue inflammation. Deletion of Bmpr1a in myeloid cells had no impact on insulin sensitivity, while ablation of Bmpr1a in mature adipocytes partially recapitulated the initial phenotype from aP2-Cre driven deletion. Co-cultivation of macrophages with pre-adipocytes lacking Bmpr1a markedly reduced expression of proinflammatory genes. CONCLUSIONS/INTERPRETATION: Our findings show that altered BMP signalling in adipose tissue affects the tissue's metabolic properties and systemic insulin resistance by altering the pattern of immune cell infiltration. The phenotype is due to ablation of Bmpr1a specifically in pre-adipocytes and maturing adipocytes rather than an immune cell-autonomous effect. Mechanistically, we provide evidence for a BMP-mediated direct crosstalk between pre-adipocytes and macrophages.


Asunto(s)
Tejido Adiposo/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Resistencia a la Insulina/fisiología , Adipocitos/metabolismo , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Dieta Alta en Grasa/efectos adversos , Ácidos Grasos no Esterificados/sangre , Glucosa/metabolismo , Insulina/sangre , Resistencia a la Insulina/genética , Interleucina-6/sangre , Ratones , Ratones Noqueados , Factor de Necrosis Tumoral alfa/sangre
8.
EMBO Rep ; 16(10): 1378-93, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26303948

RESUMEN

Brown adipose tissue (BAT) dissipates chemical energy as heat and can counteract obesity. MicroRNAs are emerging as key regulators in development and disease. Combining microRNA and mRNA microarray profiling followed by bioinformatic analyses, we identified miR-455 as a new regulator of brown adipogenesis. miR-455 exhibits a BAT-specific expression pattern and is induced by cold and the browning inducer BMP7. In vitro gain- and loss-of-function studies show that miR-455 regulates brown adipocyte differentiation and thermogenesis. Adipose-specific miR-455 transgenic mice display marked browning of subcutaneous white fat upon cold exposure. miR-455 activates AMPKα1 by targeting HIF1an, and AMPK promotes the brown adipogenic program and mitochondrial biogenesis. Concomitantly, miR-455 also targets the adipogenic suppressors Runx1t1 and Necdin, initiating adipogenic differentiation. Taken together, the data reveal a novel microRNA-regulated signaling network that controls brown adipogenesis and may be a potential therapeutic target for human metabolic disorders.


Asunto(s)
Adipocitos Marrones/metabolismo , Adipogénesis/genética , MicroARNs/genética , Transducción de Señal , Proteínas Quinasas Activadas por AMP/metabolismo , Tejido Adiposo Blanco , Animales , Diferenciación Celular/genética , Células Cultivadas , Frío , Humanos , Ratones , Ratones Transgénicos , Oxigenasas de Función Mixta/genética , Oxigenasas de Función Mixta/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Biogénesis de Organelos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Proteínas Represoras/metabolismo , Termogénesis/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
9.
Nat Med ; 21(7): 760-8, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26076036

RESUMEN

Targeting brown adipose tissue (BAT) content or activity has therapeutic potential for treating obesity and the metabolic syndrome by increasing energy expenditure. However, both inter- and intra-individual differences contribute to heterogeneity in human BAT and potentially to differential thermogenic capacity in human populations. Here we generated clones of brown and white preadipocytes from human neck fat and characterized their adipogenic and thermogenic differentiation. We combined an uncoupling protein 1 (UCP1) reporter system and expression profiling to define novel sets of gene signatures in human preadipocytes that could predict the thermogenic potential of the cells once they were maturated. Knocking out the positive UCP1 regulators, PREX1 and EDNRB, in brown preadipocytes using CRISPR-Cas9 markedly abolished the high level of UCP1 in brown adipocytes differentiated from the preadipocytes. Finally, we were able to prospectively isolate adipose progenitors with great thermogenic potential using the cell surface marker CD29. These data provide new insights into the cellular heterogeneity in human fat and offer potential biomarkers for identifying thermogenically competent preadipocytes.


Asunto(s)
Adipocitos Marrones/metabolismo , Adipocitos Blancos/metabolismo , Biomarcadores/metabolismo , Perfilación de la Expresión Génica , Termogénesis/genética , Adipocitos Marrones/citología , Adipocitos Blancos/citología , Diferenciación Celular , Línea Celular Transformada , Membrana Celular/metabolismo , Células Clonales , Genes Reporteros , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Integrina beta1/metabolismo , Canales Iónicos/genética , Canales Iónicos/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor de Endotelina B , Receptores de Endotelina/metabolismo , Células Madre/citología , Células Madre/metabolismo , Proteína Desacopladora 1
10.
Nat Med ; 21(4): 389-94, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25774848

RESUMEN

Brown adipose tissue (BAT) acts in mammals as a natural defense system against hypothermia, and its activation to a state of increased energy expenditure is believed to protect against the development of obesity. Even though the existence of BAT in adult humans has been widely appreciated, its cellular origin and molecular identity remain elusive largely because of high cellular heterogeneity within various adipose tissue depots. To understand the nature of adult human brown adipocytes at single cell resolution, we isolated clonally derived adipocytes from stromal vascular fractions of adult human BAT from two individuals and globally analyzed their molecular signatures. We used RNA sequencing followed by unbiased genome-wide expression analyses and found that a population of uncoupling protein 1 (UCP1)-positive human adipocytes possessed molecular signatures resembling those of a recruitable form of thermogenic adipocytes (that is, beige adipocytes). In addition, we identified molecular markers that were highly enriched in UCP1-positive human adipocytes, a set that included potassium channel K3 (KCNK3) and mitochondrial tumor suppressor 1 (MTUS1). Further, we functionally characterized these two markers using a loss-of-function approach and found that KCNK3 and MTUS1 were required for beige adipocyte differentiation and thermogenic function. The results of this study present new opportunities for human BAT research, such as facilitating cell-based disease modeling and unbiased screens for thermogenic regulators.


Asunto(s)
Adipocitos Marrones/metabolismo , Tejido Adiposo Pardo/citología , Canales Iónicos/genética , Proteínas Mitocondriales/genética , Proteínas del Tejido Nervioso/genética , Canales de Potasio de Dominio Poro en Tándem/genética , Proteínas Supresoras de Tumor/genética , Adulto , Animales , Proteínas Portadoras/genética , Análisis por Conglomerados , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Interferencia de ARN , Termogénesis/fisiología , Proteína Desacopladora 1
11.
Peptides ; 63: 126-33, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25451330

RESUMEN

Despite its wide distribution in the central nervous system, the presence of Neuropeptide Y (NPY) in peripheral tissues has been detected. White adipose tissue (WAT) is a new site of NPY synthesis and secretion. The development of brown-like adipocytes in WAT is controlled by hypothalamic NPY neurons through interaction with sympathetic nervous system (SNS). However, whether peripheral NPY has a direct effect on induction of the Uncoupling protein1 (UCP1)-positive adipocytes is unknown. We have used adipocytes derived from C3H10T1/2 stem cells as a model of brown-like adipocyte, and investigated the role of NPY in their differentiation and activation. In general, NPY had no effect on brown adipogenesis of C3H10T1/2 stem cell, but suppressed db-cAMP activation of brown-like adipocytes, which was due to blunting brown fat-relevant gene expression and mitochondrial function. NPY showed suppression in a receptor-dependent manner, inhibition of endogenous cAMP production and cAMP-PKA-dependent pathways p38 MAPK and CREB phosphorylation were involved in the downstream mechanisms. A novel role of NPY in the peripheral is presented, which may help decrease energy expenditure in WAT of obese subjects.


Asunto(s)
Adipocitos Marrones/fisiología , Neuropéptido Y/fisiología , Adipocitos Marrones/efectos de los fármacos , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Diferenciación Celular , Línea Celular , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Expresión Génica , Silenciador del Gen , Yoduro Peroxidasa/genética , Yoduro Peroxidasa/metabolismo , Canales Iónicos/genética , Canales Iónicos/metabolismo , Ratones , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Sistemas de Mensajero Secundario , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína Desacopladora 1 , Yodotironina Deyodinasa Tipo II
12.
Sci Transl Med ; 6(247): 247ra103, 2014 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-25080478

RESUMEN

White, beige, and brown adipocytes are developmentally and functionally distinct but often occur mixed together within individual depots. To target white, beige, and brown adipocytes for diagnostic or therapeutic purposes, a better understanding of the cell surface properties of these cell types is essential. Using a combination of in silico, in vitro, and in vivo methods, we have identified three new cell surface markers of adipose cell types. The amino acid transporter ASC-1 is a white adipocyte-specific cell surface protein, with little or no expression in brown adipocytes, whereas the amino acid transporter PAT2 and the purinergic receptor P2RX5 are cell surface markers expressed in classical brown and beige adipocytes in mice. These markers also selectively mark brown/beige and white adipocytes in human tissue. Thus, ASC-1, PAT2, and P2RX5 are membrane surface proteins that may serve as tools to identify and target white and brown/beige adipocytes for therapeutic purposes.


Asunto(s)
Adipocitos Marrones/metabolismo , Adipocitos Blancos/metabolismo , Sistema de Transporte de Aminoácidos y+/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Membrana Celular/metabolismo , Receptores Purinérgicos P2X5/metabolismo , Simportadores/metabolismo , Adipocitos Marrones/efectos de los fármacos , Adipocitos Blancos/efectos de los fármacos , Agonistas de Receptores Adrenérgicos beta 3/farmacología , Sistema de Transporte de Aminoácidos y+/genética , Sistemas de Transporte de Aminoácidos Neutros/genética , Animales , Biomarcadores/metabolismo , Membrana Celular/efectos de los fármacos , Frío , Biología Computacional , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Obesidad/genética , Obesidad/metabolismo , Receptores Purinérgicos P2X5/genética , Simportadores/genética , Factores de Tiempo
13.
Cell ; 158(1): 69-83, 2014 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-24995979

RESUMEN

Brown fat can reduce obesity through the dissipation of calories as heat. Control of thermogenic gene expression occurs via the induction of various coactivators, most notably PGC-1α. In contrast, the transcription factor partner(s) of these cofactors are poorly described. Here, we identify interferon regulatory factor 4 (IRF4) as a dominant transcriptional effector of thermogenesis. IRF4 is induced by cold and cAMP in adipocytes and is sufficient to promote increased thermogenic gene expression, energy expenditure, and cold tolerance. Conversely, knockout of IRF4 in UCP1(+) cells causes reduced thermogenic gene expression and energy expenditure, obesity, and cold intolerance. IRF4 also induces the expression of PGC-1α and PRDM16 and interacts with PGC-1α, driving Ucp1 expression. Finally, cold, ß-agonists, or forced expression of PGC-1α are unable to cause thermogenic gene expression in the absence of IRF4. These studies establish IRF4 as a transcriptional driver of a program of thermogenic gene expression and energy expenditure.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Factores Reguladores del Interferón/metabolismo , Termogénesis , Factores de Transcripción/metabolismo , Activación Transcripcional , Adipocitos/metabolismo , Tejido Adiposo Pardo/citología , Agonistas de Receptores Adrenérgicos beta 3/farmacología , Animales , Frío , AMP Cíclico/metabolismo , Metabolismo Energético , Humanos , Canales Iónicos/genética , Ratones , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Obesidad/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Delgadez/metabolismo , Activación Transcripcional/efectos de los fármacos , Proteína Desacopladora 1
14.
Am J Physiol Endocrinol Metab ; 306(4): E363-72, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24347060

RESUMEN

There are two different types of fat present in mammals: white adipose tissue, the primary site of energy storage, and brown adipose tissue, which is specializes in energy expenditure. Factors that specify the developmental fate and function of brown fat are poorly understood. Bone morphogenic proteins (BMPs) play an important role in adipogenesis. While BMP4 is capable of triggering commitment of stem cells to the white adipocyte lineage, BMP7 triggers commitment of progenitor cells to a brown adipocyte lineage and activates brown adipogenesis. To investigate the differential effects of BMPs on the development of adipocytes, C3H10T1/2 pluripotent cells were pretreated with BMP4 and BMP7, followed by different adipogenic induction cocktails. Both BMP4 and BMP7 unexpectedly activated a full program of brown adipogenesis, including induction of the brown fat-defining marker uncoupling protein-1 (UCP1), increasing the expression of early regulators of brown fat fate PRDM16 (PR domain-containing 16) and induction of mitochondrial biogenesis and function. Implantation of BMP4-pretreated C3H10T1/2 cells into nude mice resulted in the development of adipose tissue depots containing UCP1-positive brown adipocytes. Interestingly, BMP4 could also induce brown fat-like adipocytes in both white and brown preadipocytes, thereby decreasing the classical brown adipocyte marker Zic1 and increasing the recently identified beige adipocyte marker TMEM26. The data indicate an important role for BMP4 in promoting brown adipocyte differentiation and thermogenesis in vivo and in vitro and offers a potentially new therapeutic approach for the treatment of obesity.


Asunto(s)
Adipocitos Marrones/efectos de los fármacos , Adipogénesis/efectos de los fármacos , Proteína Morfogenética Ósea 4/farmacología , Adipocitos Marrones/citología , Adipocitos Marrones/metabolismo , Adipogénesis/fisiología , Animales , Proteína Morfogenética Ósea 7/farmacología , Línea Celular , Proteínas de Unión al ADN/metabolismo , Canales Iónicos/metabolismo , Masculino , Ratones , Ratones Desnudos , Proteínas Mitocondriales/metabolismo , Factores de Transcripción/metabolismo , Proteína Desacopladora 1
15.
Eur J Endocrinol ; 170(3): 359-66, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24288355

RESUMEN

OBJECTIVE: Previous studies have shown that active brown adipose tissue (BAT) is present in adults and may play important roles in the regulation of energy homeostasis. However, nearly every study has been carried out in patients undergoing scanning for cancer surveillance (CS), whose metabolism and BAT activity may not reflect those of healthy individuals. The objective of this study was to investigate the prevalence and predictors of active BAT in Chinese adults, particularly in healthy individuals. DESIGN: A total of 31,088 consecutive subjects aged ≥18 years who had undergone positron emission tomography/computed tomography (PET/CT) scanning of BAT were evaluated in this study. METHODS: We measured BAT activity via (18)F-fluorodeoxyglucose PET/CT in subjects who had undergone scanning for either a routine medical checkup (MC) or CS in Shanghai. Then, we investigated the predictors of active BAT, particularly in healthy individuals. RESULTS: In both groups, the prevalence of BAT was higher in women than in men. Using a multivariate logistic analysis, we found age, sex, BMI, and high thyroid glucose uptake to be significant predictors of BAT activity in the MC group. Similarly, we found age, sex, and BMI to be significant predictors of BAT activity, but not thyroid high glucose uptake, in the CS group. CONCLUSIONS: In Chinese adults, BAT activity inversely correlates with BMI and thyroid high glucose uptake, which reinforces the central role of brown fat in adult metabolism and provides clues to a potential means for treating the metabolic syndrome.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Adolescente , Adulto , Pueblo Asiatico , Índice de Masa Corporal , China/epidemiología , Femenino , Fluorodesoxiglucosa F18 , Glucosa/metabolismo , Humanos , Masculino , Tomografía de Emisión de Positrones , Prevalencia , Estudios Retrospectivos , Glándula Tiroides/metabolismo , Tomografía Computarizada por Rayos X
16.
Wien Klin Wochenschr ; 125(21-22): 687-95, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24146327

RESUMEN

BACKGROUND: Previous studies have proven the existence of active brown adipose tissue (BAT) in adults; however, its effect on systematic metabolism remains unclear. AIM: The current study was designed to investigate the differences in the metabolic profiles of healthy adults with and without active BAT using positron emission tomography-computed tomography (PET-CT) scans in the un-stimulated state. METHODS: A cross-sectional analysis was performed to assess the health of adults using PET-CT whole-body scans at Huashan Hospital Medical Centre between November 2009 and May 2010. A total of 62 healthy adults with active BAT were enrolled in the BAT-positive group. For each positive subject, a same-gender individual who underwent PET-CT the same day and who had no detectable BAT was chosen as the negative control. Body composition was measured, and blood samples were collected for assays of metabolic profiles and other biomarkers. RESULTS: In both the male and female groups, BAT-positive individuals were younger and had lower body mass indexes, fasting insulin, insulin resistance, and leptin, but a greater level of high-density lipoprotein cholesterol compared with the negative controls. In the male group, body fat content and levels of tumor necrosis factor-α were significantly lower in the BAT-positive than in the negative control group. CONCLUSIONS: The healthy adults with active BAT in an un-stimulated state had favorable metabolic profiles suggesting that active BAT may be a potential target for preventing and treating obesity and other metabolic disorders.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Glucemia/metabolismo , Composición Corporal/fisiología , Metabolismo Energético/fisiología , Adulto , Femenino , Humanos , Masculino , Valores de Referencia
17.
Nat Med ; 19(5): 635-9, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23603815

RESUMEN

The imbalance between energy intake and expenditure is the underlying cause of the current obesity and diabetes pandemics. Central to these pathologies is the fat depot: white adipose tissue (WAT) stores excess calories, and brown adipose tissue (BAT) consumes fuel for thermogenesis using tissue-specific uncoupling protein 1 (UCP1). BAT was once thought to have a functional role in rodents and human infants only, but it has been recently shown that in response to mild cold exposure, adult human BAT consumes more glucose per gram than any other tissue. In addition to this nonshivering thermogenesis, human BAT may also combat weight gain by becoming more active in the setting of increased whole-body energy intake. This phenomenon of BAT-mediated diet-induced thermogenesis has been observed in rodents and suggests that activation of human BAT could be used as a safe treatment for obesity and metabolic dysregulation. In this study, we isolated anatomically defined neck fat from adult human volunteers and compared its gene expression, differentiation capacity and basal oxygen consumption to different mouse adipose depots. Although the properties of human neck fat vary substantially between individuals, some human samples share many similarities with classical, also called constitutive, rodent BAT.


Asunto(s)
Tejido Adiposo Pardo/anatomía & histología , Tejido Adiposo Pardo/fisiología , Perfilación de la Expresión Génica , Adipocitos/citología , Tejido Adiposo , Tejido Adiposo Pardo/metabolismo , Adulto , Animales , Diferenciación Celular , Linaje de la Célula , Análisis por Conglomerados , Expresión Génica , Regulación de la Expresión Génica , Humanos , Canales Iónicos/metabolismo , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Mitocondriales/metabolismo , Consumo de Oxígeno , Termogénesis , Proteína Desacopladora 1
18.
Nature ; 495(7441): 379-83, 2013 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-23485971

RESUMEN

Maintenance of body temperature is essential for the survival of homeotherms. Brown adipose tissue (BAT) is a specialized fat tissue that is dedicated to thermoregulation. Owing to its remarkable capacity to dissipate stored energy and its demonstrated presence in adult humans, BAT holds great promise for the treatment of obesity and metabolic syndrome. Rodent data suggest the existence of two types of brown fat cells: constitutive BAT (cBAT), which is of embryonic origin and anatomically located in the interscapular region of mice; and recruitable BAT (rBAT), which resides within white adipose tissue (WAT) and skeletal muscle, and has alternatively been called beige, brite or inducible BAT. Bone morphogenetic proteins (BMPs) regulate the formation and thermogenic activity of BAT. Here we use mouse models to provide evidence for a systemically active regulatory mechanism that controls whole-body BAT activity for thermoregulation and energy homeostasis. Genetic ablation of the type 1A BMP receptor (Bmpr1a) in brown adipogenic progenitor cells leads to a severe paucity of cBAT. This in turn increases sympathetic input to WAT, thereby promoting the formation of rBAT within white fat depots. This previously unknown compensatory mechanism, aimed at restoring total brown-fat-mediated thermogenic capacity in the body, is sufficient to maintain normal temperature homeostasis and resistance to diet-induced obesity. These data suggest an important physiological cross-talk between constitutive and recruitable brown fat cells. This sophisticated regulatory mechanism of body temperature may participate in the control of energy balance and metabolic disease.


Asunto(s)
Tejido Adiposo Pardo/citología , Tejido Adiposo Blanco/citología , Proteínas Morfogenéticas Óseas/metabolismo , Transducción de Señal , Tejido Adiposo Pardo/inervación , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Temperatura Corporal , Regulación de la Temperatura Corporal , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Proteínas Morfogenéticas Óseas/genética , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Metabolismo Energético , Ratones , Células Madre/citología
19.
Proc Natl Acad Sci U S A ; 110(9): E798-807, 2013 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-23388637

RESUMEN

Expression of bone morphogenetic protein 4 (BMP4) in adipocytes of white adipose tissue (WAT) produces "white adipocytes" with characteristics of brown fat and leads to a reduction of adiposity and its metabolic complications. Although BMP4 is known to induce commitment of pluripotent stem cells to the adipocyte lineage by producing cells that possess the characteristics of preadipocytes, its effects on the mature white adipocyte phenotype and function were unknown. Forced expression of a BMP4 transgene in white adipocytes of mice gives rise to reduced WAT mass and white adipocyte size along with an increased number of a white adipocyte cell types with brown adipocyte characteristics comparable to those of beige or brite adipocytes. These changes correlate closely with increased energy expenditure, improved insulin sensitivity, and protection against diet-induced obesity and diabetes. Conversely, BMP4-deficient mice exhibit enlarged white adipocyte morphology and impaired insulin sensitivity. We identify peroxisome proliferator-activated receptor gamma coactivator 1-α (PGC1α) as the target of BMP signaling required for these brown fat-like changes in WAT. This effect of BMP4 on WAT appears to extend to human adipose tissue, because the level of expression of BMP4 in WAT correlates inversely with body mass index. These findings provide a genetic and metabolic basis for BMP4's role in altering insulin sensitivity by affecting WAT development.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Metabolismo Energético , Glucosa/metabolismo , Homeostasis , Células 3T3-L1 , Factor de Transcripción Activador 2/metabolismo , Adipocitos Marrones/efectos de los fármacos , Adipocitos Marrones/metabolismo , Adipocitos Marrones/patología , Adipocitos Blancos/efectos de los fármacos , Adipocitos Blancos/enzimología , Adipocitos Blancos/patología , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/patología , Tejido Adiposo Pardo/ultraestructura , Tejido Adiposo Blanco/efectos de los fármacos , Tejido Adiposo Blanco/enzimología , Tejido Adiposo Blanco/ultraestructura , Animales , Dieta Alta en Grasa , Metabolismo Energético/efectos de los fármacos , Proteínas de Unión a Ácidos Grasos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Homeostasis/efectos de los fármacos , Humanos , Insulina/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Tamaño de los Órganos/efectos de los fármacos , Consumo de Oxígeno/efectos de los fármacos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Fenotipo , Delgadez/metabolismo , Delgadez/patología , Transactivadores/metabolismo , Factores de Transcripción , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...