Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 185(3): 419-446, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-35120662

RESUMEN

Adipose tissue, colloquially known as "fat," is an extraordinarily flexible and heterogeneous organ. While historically viewed as a passive site for energy storage, we now appreciate that adipose tissue regulates many aspects of whole-body physiology, including food intake, maintenance of energy levels, insulin sensitivity, body temperature, and immune responses. A crucial property of adipose tissue is its high degree of plasticity. Physiologic stimuli induce dramatic alterations in adipose-tissue metabolism, structure, and phenotype to meet the needs of the organism. Limitations to this plasticity cause diminished or aberrant responses to physiologic cues and drive the progression of cardiometabolic disease along with other pathological consequences of obesity.


Asunto(s)
Adaptación Fisiológica , Tejido Adiposo/fisiología , Enfermedad , Salud , Adipocitos Blancos/metabolismo , Animales , Humanos , Termogénesis
2.
Cell ; 182(3): 537-539, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32763184

RESUMEN

Oguri and colleagues use single-cell RNA sequencing to identify a beige adipocyte precursor cell that gives rise to thermogenic adipocytes in subcutaneous adipose tissue. These beige fat progenitors are marked by PDGFRα, Sca1, and CD81 and proliferate upon activation of FAK-signaling in response to the cold and irisin.


Asunto(s)
Adipocitos Beige , Tejido Adiposo Beige , Transducción de Señal , Células Madre , Termogénesis
3.
Genes Dev ; 33(13-14): 747-762, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31123067

RESUMEN

Prolonged cold exposure stimulates the recruitment of beige adipocytes within white adipose tissue. Beige adipocytes depend on mitochondrial oxidative phosphorylation to drive thermogenesis. The transcriptional mechanisms that promote remodeling in adipose tissue during the cold are not well understood. Here we demonstrate that the transcriptional coregulator transducin-like enhancer of split 3 (TLE3) inhibits mitochondrial gene expression in beige adipocytes. Conditional deletion of TLE3 in adipocytes promotes mitochondrial oxidative metabolism and increases energy expenditure, thereby improving glucose control. Using chromatin immunoprecipitation and deep sequencing, we found that TLE3 occupies distal enhancers in proximity to nuclear-encoded mitochondrial genes and that many of these binding sites are also enriched for early B-cell factor (EBF) transcription factors. TLE3 interacts with EBF2 and blocks its ability to promote the thermogenic transcriptional program. Collectively, these studies demonstrate that TLE3 regulates thermogenic gene expression in beige adipocytes through inhibition of EBF2 transcriptional activity. Inhibition of TLE3 may provide a novel therapeutic approach for obesity and diabetes.


Asunto(s)
Adipocitos Beige/metabolismo , Proteínas Co-Represoras/genética , Proteínas Co-Represoras/metabolismo , Glucosa/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Células Cultivadas , Dieta Alta en Grasa , Metabolismo Energético/genética , Eliminación de Gen , Regulación de la Expresión Génica/genética , Estudio de Asociación del Genoma Completo , Resistencia a la Insulina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/genética , Mitocondrias/metabolismo , Termogénesis/genética
4.
J Lipid Res ; 65(2): 100434, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37640283

RESUMEN

Adipose tissue is the site of long-term energy storage. During the fasting state, exercise, and cold exposure, the white adipose tissue mobilizes energy for peripheral tissues through lipolysis. The mobilization of lipids from white adipose tissue to the liver can lead to excess triglyceride accumulation and fatty liver disease. Although the white adipose tissue is known to release free fatty acids, a comprehensive analysis of lipids mobilized from white adipocytes in vivo has not been completed. In these studies, we provide a comprehensive quantitative analysis of the adipocyte-secreted lipidome and show that there is interorgan crosstalk with liver. Our analysis identifies multiple lipid classes released by adipocytes in response to activation of lipolysis. Time-dependent analysis of the serum lipidome showed that free fatty acids increase within 30 min of ß3-adrenergic receptor activation and subsequently decrease, followed by a rise in serum triglycerides, liver triglycerides, and several ceramide species. The triglyceride composition of liver is enriched for linoleic acid despite higher concentrations of palmitate in the blood. To further validate that these findings were a specific consequence of lipolysis, we generated mice with conditional deletion of adipose tissue triglyceride lipase exclusively in adipocytes. This loss of in vivo adipocyte lipolysis prevented the rise in serum free fatty acids and hepatic triglycerides. Furthermore, conditioned media from adipocytes promotes lipid remodeling in hepatocytes with concomitant changes in genes/pathways mediating lipid utilization. Together, these data highlight critical role of adipocyte lipolysis in interorgan crosstalk between adipocytes and liver.


Asunto(s)
Ácidos Grasos no Esterificados , Lipólisis , Ratones , Animales , Lipólisis/fisiología , Ácidos Grasos no Esterificados/metabolismo , Lipidómica , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Hígado/metabolismo , Triglicéridos/metabolismo
5.
PLoS Genet ; 15(2): e1007970, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30768595

RESUMEN

Identifying regulatory mechanisms that influence inflammation in metabolic tissues is critical for developing novel metabolic disease treatments. Here, we investigated the role of microRNA-146a (miR-146a) during diet-induced obesity in mice. miR-146a is reduced in obese and type 2 diabetic patients and our results reveal that miR-146a-/- mice fed a high-fat diet (HFD) have exaggerated weight gain, increased adiposity, hepatosteatosis, and dysregulated blood glucose levels compared to wild-type controls. Pro-inflammatory genes and NF-κB activation increase in miR-146a-/- mice, indicating a role for this miRNA in regulating inflammatory pathways. RNA-sequencing of adipose tissue macrophages demonstrated a role for miR-146a in regulating both inflammation and cellular metabolism, including the mTOR pathway, during obesity. Further, we demonstrate that miR-146a regulates inflammation, cellular respiration and glycolysis in macrophages through a mechanism involving its direct target Traf6. Finally, we found that administration of rapamycin, an inhibitor of mTOR, was able to rescue the obesity phenotype in miR-146a-/- mice. Altogether, our study provides evidence that miR-146a represses inflammation and diet-induced obesity and regulates metabolic processes at the cellular and organismal levels, demonstrating how the combination of diet and miRNA genetics influences obesity and diabetic phenotypes.


Asunto(s)
Inflamación/prevención & control , Enfermedades Metabólicas/prevención & control , MicroARNs/genética , MicroARNs/metabolismo , Animales , Glucemia/metabolismo , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Humanos , Hiperglucemia/genética , Hiperglucemia/metabolismo , Hiperglucemia/prevención & control , Inflamación/genética , Inflamación/metabolismo , Insulina/sangre , Grasa Intraabdominal/metabolismo , Grasa Intraabdominal/patología , Macrófagos/metabolismo , Masculino , Enfermedades Metabólicas/genética , Enfermedades Metabólicas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/antagonistas & inhibidores , FN-kappa B/metabolismo , Obesidad/genética , Obesidad/metabolismo , Obesidad/prevención & control , Proteínas Proto-Oncogénicas c-akt/genética , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/genética , Aumento de Peso/efectos de los fármacos , Aumento de Peso/genética
6.
J Cell Biochem ; 120(3): 3599-3610, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30272815

RESUMEN

Adipocyte differentiation is controlled by multiple signaling pathways. To identify new adipogenic factors, C3H10T1/2 adipocytes were treated with previously known antiadipogenic phytochemicals (resveratrol, butein, sulfuretin, and fisetin) for 24 hours. Commonly regulated genes were then identified by transcriptional profiling analysis. Three genes (chemokine (C-X-C motif) ligand 1 [ Cxcl1], heme oxygenase 1 [ Hmox1], and PHD (plant homeo domain) finger protein 16 [ Phf16]) were upregulated while two genes (G0/G1 switch gene 2 [ G0s2] and patatin-like phospholipase domain containing 3 [ Pnpla3]) were downregulated by these four antiadipogenic compounds. Tissue expression profiles showed that the G0s2 and Pnpla3 expressions were highly specific to adipose depots while the other three induced genes were ubiquitously expressed with significantly higher expression in adipose tissues. While Cxcl1 expression was decreased, expressions of the other four genes were significantly increased during adipogenic differentiation of C3H10T1/2 cells. Small interfering RNA-mediated knockdown including Phf16 and Pnpla3 indicated that these genes might play regulatory roles in lipid accumulation and adipocyte differentiation. Specifically, the silencing of two newly identified adipogenic genes, Phf16 or Pnpla3, suppressed lipid accumulation and expression of adipocyte markers in both 3T3-L1 and C3H10T1/2 cells. Taken together, these data showed previously uncovered roles of Phf16 and Pnpla3 in adipogenesis, highlighting the potential of using phytochemicals for further investigation of adipocyte biology.


Asunto(s)
Adipogénesis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas Oncogénicas/metabolismo , Fosfolipasas A2 Calcio-Independiente/metabolismo , Fitoquímicos/farmacología , Células 3T3-L1 , Animales , Quimiocina CXCL1/biosíntesis , Ratones , Proteínas Oncogénicas/genética , Fosfolipasas A2 Calcio-Independiente/genética
7.
Am J Physiol Endocrinol Metab ; 316(2): E293-E304, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30512991

RESUMEN

Mitochondrial dynamics refers to the constant remodeling of mitochondrial populations by multiple cellular pathways that help maintain mitochondrial health and function. Disruptions in mitochondrial dynamics often lead to mitochondrial dysfunction, which is frequently associated with disease in rodents and humans. Consistent with this, obesity is associated with reduced mitochondrial function in white adipose tissue, partly via alterations in mitochondrial dynamics. Several proteins, including the E3 ubiquitin ligase membrane-associated RING-CH-type finger 5 (MARCH5), are known to regulate mitochondrial dynamics; however, the role of these proteins in adipocytes has been poorly studied. Here, we show that MARCH5 is regulated by peroxisome proliferator-activated receptor-γ (PPARγ) during adipogenesis and is correlated with fat mass across a panel of genetically diverse mouse strains, in ob/ob mice, and in humans. Furthermore, manipulation of MARCH5 expression in vitro and in vivo alters mitochondrial function, affects cellular metabolism, and leads to differential regulation of several metabolic genes. Thus our data demonstrate an association between mitochondrial dynamics and metabolism that defines MARCH5 as a critical link between these interconnected pathways.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Proteínas de la Membrana/metabolismo , Síndrome Metabólico/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Obesidad/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Células 3T3-L1 , Adipogénesis , Adulto , Animales , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Proteínas Mitocondriales/genética , PPAR gamma/genética , PPAR gamma/metabolismo , Ubiquitina-Proteína Ligasas/genética
8.
Immunity ; 33(5): 647-9, 2010 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-21094459

RESUMEN

The mechanisms that direct cell-type-specific peroxisome proliferator-activated receptor (PPAR) gene programs are poorly understood. In this issue of Immunity, Szanto et al. (2010) identify signal transducer and activator of transcription 6 as a transcriptional switch that licenses PPARγ-dependent gene expression in macrophages and dendritic cells.

9.
J Mol Cell Cardiol ; 113: 22-32, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28962857

RESUMEN

Calcium (Ca2+) influx into the mitochondrial matrix stimulates ATP synthesis. Here, we investigate whether mitochondrial Ca2+ transport pathways are altered in the setting of deficient mitochondrial energy synthesis, as increased matrix Ca2+ may provide a stimulatory boost. We focused on mitochondrial cardiomyopathies, which feature such dysfunction of oxidative phosphorylation. We study a mouse model where the main transcription factor for mitochondrial DNA (transcription factor A, mitochondrial, Tfam) has been disrupted selectively in cardiomyocytes. By the second postnatal week (10-15day old mice), these mice have developed a dilated cardiomyopathy associated with impaired oxidative phosphorylation. We find evidence of increased mitochondrial Ca2+ during this period using imaging, electrophysiology, and biochemistry. The mitochondrial Ca2+ uniporter, the main portal for Ca2+ entry, displays enhanced activity, whereas the mitochondrial sodium-calcium (Na+-Ca2+) exchanger, the main portal for Ca2+ efflux, is inhibited. These changes in activity reflect changes in protein expression of the corresponding transporter subunits. While decreased transcription of Nclx, the gene encoding the Na+-Ca2+ exchanger, explains diminished Na+-Ca2+ exchange, the mechanism for enhanced uniporter expression appears to be post-transcriptional. Notably, such changes allow cardiac mitochondria from Tfam knockout animals to be far more sensitive to Ca2+-induced increases in respiration. In the absence of Ca2+, oxygen consumption declines to less than half of control values in these animals, but rebounds to control levels when incubated with Ca2+. Thus, we demonstrate a phenotype of enhanced mitochondrial Ca2+ in a mitochondrial cardiomyopathy model, and show that such Ca2+ accumulation is capable of rescuing deficits in energy synthesis capacity in vitro.


Asunto(s)
Señalización del Calcio/fisiología , Calcio/metabolismo , Cardiomiopatías/metabolismo , Mitocondrias Cardíacas/metabolismo , Animales , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/metabolismo , Fosforilación Oxidativa , Sodio/metabolismo , Intercambiador de Sodio-Calcio/metabolismo
10.
J Biol Chem ; 290(9): 5566-81, 2015 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-25468909

RESUMEN

Obesity is associated with increased breast cancer (BrCA) incidence. Considering that inactivation of estrogen receptor (ER)α promotes obesity and metabolic dysfunction in women and female mice, understanding the mechanisms and tissue-specific sites of ERα action to combat metabolic-related disease, including BrCA, is of clinical importance. To study the role of ERα in adipose tissue we generated fat-specific ERα knock-out (FERKO) mice. Herein we show that ERα deletion increased adipocyte size, fat pad weight, and tissue expression and circulating levels of the secreted glycoprotein, lipocalin 2 (Lcn2), an adipokine previously associated with BrCA development. Chromatin immunoprecipitation and luciferase reporter studies showed that ERα binds the Lcn2 promoter to repress its expression. Because adipocytes constitute an important cell type of the breast microenvironment, we examined the impact of adipocyte ERα deletion on cancer cell behavior. Conditioned medium from ERα-null adipocytes and medium containing pure Lcn2 increased proliferation and migration of a subset of BrCA cells in culture. The proliferative and promigratory effects of ERα-deficient adipocyte-conditioned medium on BrCA cells was reversed by Lcn2 deletion. BrCA cell responsiveness to exogenous Lcn2 was heightened in cell types where endogenous Lcn2 expression was minimal, but components of the Lcn2 signaling pathway were enriched, i.e. SLC22A17 and 3-hydroxybutyrate dehydrogenase (BDH2). In breast tumor biopsies from women diagnosed with BrCA we found that BDH2 expression was positively associated with adiposity and circulating Lcn2 levels. Collectively these data suggest that reduction of ERα expression in adipose tissue promotes adiposity and is linked with the progression and severity of BrCA via increased adipocyte-specific Lcn2 production and enhanced tumor cell Lcn2 sensitivity.


Asunto(s)
Proteínas de Fase Aguda/metabolismo , Tejido Adiposo/metabolismo , Receptor alfa de Estrógeno/metabolismo , Lipocalinas/metabolismo , Obesidad/metabolismo , Proteínas Oncogénicas/metabolismo , Células 3T3-L1 , Proteínas de Fase Aguda/genética , Adipocitos/citología , Adipocitos/metabolismo , Tejido Adiposo/citología , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Progresión de la Enfermedad , Receptor alfa de Estrógeno/genética , Femenino , Perfilación de la Expresión Génica , Células HEK293 , Humanos , Immunoblotting , Lipocalina 2 , Lipocalinas/sangre , Lipocalinas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Obesidad/genética , Proteínas Oncogénicas/sangre , Proteínas Oncogénicas/genética , Regiones Promotoras Genéticas/genética , Unión Proteica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
J Clin Invest ; 133(21)2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37909330

RESUMEN

Interplay between energy-storing white adipose cells and thermogenic beige adipocytes contributes to obesity and insulin resistance. Irrespective of specialized niche, adipocytes require the activity of the nuclear receptor PPARγ for proper function. Exposure to cold or adrenergic signaling enriches thermogenic cells though multiple pathways that act synergistically with PPARγ; however, the molecular mechanisms by which PPARγ licenses white adipose tissue to preferentially adopt a thermogenic or white adipose fate in response to dietary cues or thermoneutral conditions are not fully elucidated. Here, we show that a PPARγ/long noncoding RNA (lncRNA) axis integrates canonical and noncanonical thermogenesis to restrain white adipose tissue heat dissipation during thermoneutrality and diet-induced obesity. Pharmacologic inhibition or genetic deletion of the lncRNA Lexis enhances uncoupling protein 1-dependent (UCP1-dependent) and -independent thermogenesis. Adipose-specific deletion of Lexis counteracted diet-induced obesity, improved insulin sensitivity, and enhanced energy expenditure. Single-nuclei transcriptomics revealed that Lexis regulates a distinct population of thermogenic adipocytes. We systematically map Lexis motif preferences and show that it regulates the thermogenic program through the activity of the metabolic GWAS gene and WNT modulator TCF7L2. Collectively, our studies uncover a new mode of crosstalk between PPARγ and WNT that preserves white adipose tissue plasticity.


Asunto(s)
Resistencia a la Insulina , ARN Largo no Codificante , Animales , Ratones , Adipocitos/metabolismo , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Resistencia a la Insulina/genética , Obesidad/genética , Obesidad/metabolismo , PPAR gamma/genética , PPAR gamma/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Termogénesis/genética , Proteína Desacopladora 1/genética
12.
Sci Adv ; 9(8): eade7864, 2023 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-36827367

RESUMEN

Thermogenesis by uncoupling protein 1 (UCP1) is one of the primary mechanisms by which brown adipose tissue (BAT) increases energy expenditure. UCP1 resides in the inner mitochondrial membrane (IMM), where it dissipates membrane potential independent of adenosine triphosphate (ATP) synthase. Here, we provide evidence that phosphatidylethanolamine (PE) modulates UCP1-dependent proton conductance across the IMM to modulate thermogenesis. Mitochondrial lipidomic analyses revealed PE as a signature molecule whose abundance bidirectionally responds to changes in thermogenic burden. Reduction in mitochondrial PE by deletion of phosphatidylserine decarboxylase (PSD) made mice cold intolerant and insensitive to ß3 adrenergic receptor agonist-induced increase in whole-body oxygen consumption. High-resolution respirometry and fluorometry of BAT mitochondria showed that loss of mitochondrial PE specifically lowers UCP1-dependent respiration without compromising electron transfer efficiency or ATP synthesis. These findings were confirmed by a reduction in UCP1 proton current in PE-deficient mitoplasts. Thus, PE performs a previously unknown role as a temperature-responsive rheostat that regulates UCP1-dependent thermogenesis.


Asunto(s)
Fosfatidiletanolaminas , Protones , Ratones , Animales , Proteína Desacopladora 1/metabolismo , Fosfatidiletanolaminas/metabolismo , Mitocondrias/metabolismo , Termogénesis , Obesidad/metabolismo , Adenosina Trifosfato/metabolismo , Ratones Noqueados
13.
Nat Metab ; 5(1): 165-181, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36646756

RESUMEN

In cell models, changes in the 'accessible' pool of plasma membrane (PM) cholesterol are linked with the regulation of endoplasmic reticulum sterol synthesis and metabolism by the Aster family of nonvesicular transporters; however, the relevance of such nonvesicular transport mechanisms for lipid homeostasis in vivo has not been defined. Here we reveal two physiological contexts that generate accessible PM cholesterol and engage the Aster pathway in the liver: fasting and reverse cholesterol transport. During fasting, adipose-tissue-derived fatty acids activate hepatocyte sphingomyelinase to liberate sequestered PM cholesterol. Aster-dependent cholesterol transport during fasting facilitates cholesteryl ester formation, cholesterol movement into bile and very low-density lipoprotein production. During reverse cholesterol transport, high-density lipoprotein delivers excess cholesterol to the hepatocyte PM through scavenger receptor class B member 1. Loss of hepatic Asters impairs cholesterol movement into feces, raises plasma cholesterol levels and causes cholesterol accumulation in peripheral tissues. These results reveal fundamental mechanisms by which Aster cholesterol flux contributes to hepatic and systemic lipid homeostasis.


Asunto(s)
Colesterol , Hígado , Colesterol/metabolismo , Transporte Biológico/fisiología , Hígado/metabolismo , Homeostasis , Ácidos Grasos/metabolismo
14.
Cell Metab ; 33(8): 1505-1506, 2021 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-34348093

RESUMEN

In a new study, Zhang et al. (2021) show that reducing iron levels in adipose tissue improves metabolic function. This occurs through an interorgan communication system where signals from the adipocyte reduce intestinal lipid absorption.


Asunto(s)
Tejido Adiposo , Metabolismo de los Lípidos , Adipocitos , Tejido Adiposo/metabolismo
15.
Hepatology ; 50(2): 434-42, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19472314

RESUMEN

UNLABELLED: Nonalcoholic fatty liver disease, characterized by the accumulation of triacylglycerols (TGs) and other lipids in the liver, often accompanies obesity and is a risk factor for nonalcoholic steatohepatitis and fibrosis. To treat or prevent fatty liver, a thorough understanding of hepatic fatty acid and TG metabolism is crucial. To investigate the role of acyl CoA:diacylglycerol acyltransferase 1 (DGAT1), a key enzyme of TG synthesis, in fatty liver development, we studied mice with global and liver-specific knockout of Dgat1. DGAT1 was required for hepatic steatosis induced by a high-fat diet and prolonged fasting, which are both characterized by delivery of exogenous fatty acids to the liver. Studies in primary hepatocytes showed that DGAT1 deficiency protected against hepatic steatosis by reducing synthesis and increasing the oxidation of fatty acids. In contrast, lipodystrophy (aP2-SREBP-1c436) and liver X receptor activation (T0901317), which increase de novo fatty acid synthesis in liver, caused steatosis independently of DGAT1. Pharmacologic inhibition of Dgat1 with antisense oligonucleotides protected against fatty liver induced by a high-fat diet. CONCLUSION: Our findings identify a specific role for hepatic DGAT1 in esterification of exogenous fatty acids and indicate that DGAT1 contributes to hepatic steatosis induced by this mechanism.


Asunto(s)
Diacilglicerol O-Acetiltransferasa/deficiencia , Ácidos Grasos/efectos adversos , Hígado Graso/enzimología , Animales , Grasas de la Dieta/efectos adversos , Ayuno/efectos adversos , Ácidos Grasos/administración & dosificación , Ácidos Grasos/biosíntesis , Hígado Graso/etiología , Hígado Graso/prevención & control , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Oligonucleótidos Antisentido/farmacología
16.
Elife ; 92020 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-32795388

RESUMEN

Brown adipose tissue (BAT) is composed of thermogenic cells that convert chemical energy into heat to maintain a constant body temperature and counteract metabolic disease. The metabolic adaptations required for thermogenesis are not fully understood. Here, we explore how steady state levels of metabolic intermediates are altered in brown adipose tissue in response to cold exposure. Transcriptome and metabolome analysis revealed changes in pathways involved in amino acid, glucose, and TCA cycle metabolism. Using isotopic labeling experiments, we found that activated brown adipocytes increased labeling of pyruvate and TCA cycle intermediates from U13C-glucose. Although glucose oxidation has been implicated as being essential for thermogenesis, its requirement for efficient thermogenesis has not been directly tested. We show that mitochondrial pyruvate uptake is essential for optimal thermogenesis, as conditional deletion of Mpc1 in brown adipocytes leads to impaired cold adaptation. Isotopic labeling experiments using U13C-glucose showed that loss of MPC1 led to impaired labeling of TCA cycle intermediates. Loss of MPC1 in BAT increased 3-hydroxybutyrate levels in blood and BAT in response to the cold, suggesting that ketogenesis provides an alternative fuel source to compensate. Collectively, these studies highlight that complete glucose oxidation is essential for optimal brown fat thermogenesis.


Asunto(s)
Tejido Adiposo Pardo/fisiología , Proteínas de Transporte de Anión/genética , Frío , Glucosa/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/genética , Transportadores de Ácidos Monocarboxílicos/genética , Termogénesis , Adipocitos Marrones/metabolismo , Animales , Proteínas de Transporte de Anión/metabolismo , Masculino , Metabolómica , Ratones , Ratones Endogámicos C57BL , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Oxidación-Reducción , Suero/química
17.
Cell Metab ; 31(2): 284-300.e7, 2020 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-31813825

RESUMEN

Although metabolic adaptations have been demonstrated to be essential for tumor cell proliferation, the metabolic underpinnings of tumor initiation are poorly understood. We found that the earliest stages of colorectal cancer (CRC) initiation are marked by a glycolytic metabolic signature, including downregulation of the mitochondrial pyruvate carrier (MPC), which couples glycolysis and glucose oxidation through mitochondrial pyruvate import. Genetic studies in Drosophila suggest that this downregulation is required because hyperplasia caused by loss of the Apc or Notch tumor suppressors in intestinal stem cells can be completely blocked by MPC overexpression. Moreover, in two distinct CRC mouse models, loss of Mpc1 prior to a tumorigenic stimulus doubled the frequency of adenoma formation and produced higher grade tumors. MPC loss was associated with a glycolytic metabolic phenotype and increased expression of stem cell markers. These data suggest that changes in cellular pyruvate metabolism are necessary and sufficient to promote cancer initiation.


Asunto(s)
Adenoma/metabolismo , Carcinogénesis/metabolismo , Neoplasias Colorrectales/metabolismo , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Ácido Pirúvico/metabolismo , Animales , Transformación Celular Neoplásica/metabolismo , Drosophila , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL
18.
Mol Endocrinol ; 22(12): 2596-608, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18945812

RESUMEN

Members of the nuclear receptor 4A (NR4A) subgroup of nuclear receptors have been implicated in the regulation of glucose and lipid metabolism in insulin-sensitive tissues such as liver and skeletal muscle. However, their function in adipocytes is not well defined. Previous studies have reported that these receptors are rapidly up-regulated after treatment of 3T3-L1 preadipocytes with an adipogenic cocktail. We show here that although Nur77 expression is acutely induced by cAMP agonists in 3T3-L1 cells, it is not induced by other adipogenic stimuli, such as peroxisome proliferator-activated receptor-gamma ligands, nor is it induced during the differentiation of 3T3-F442A preadipocytes, suggesting that Nur77 induction is not an obligatory feature of preadipocyte differentiation. We further demonstrate that inflammatory signals that antagonize differentiation, such as TNFalpha and lipopolysaccharide, acutely induce Nur77 expression both in vitro and in vivo. We also show that NR4A expression in adipose tissue is responsive to fasting/refeeding. Retroviral transduction of each of the NR4A receptors (Nur77, Nurr1, and NOR1) into either 3T3-L1 or 3T3-F442A preadipocytes potently inhibits adipogenesis. Interestingly, NR4A-mediated inhibition of adipogenesis cannot be rescued by peroxisome proliferator-activated receptor-gamma overexpression or activation. Transcriptional profiling of Nur77-expressing preadipocytes led to the identification of gap-junction protein alpha1 (Gja1) and tolloid-like 1 (Tll1) as Nur77-responsive genes. Remarkably, retroviral expression of either Gja1 or Tll1 in 3T3-L1 preadipocytes also inhibited adipocyte differentiation, implicating these genes as potential mediators of Nur77's effects on adipogenesis. Finally, we show that Nur77 expression inhibits mitotic clonal expansion of preadipocytes, providing an additional mechanism by which Nur77 may inhibit adipogenesis.


Asunto(s)
Adipogénesis/genética , Diferenciación Celular/genética , Proteínas de Unión al ADN/fisiología , Proteínas del Tejido Nervioso/fisiología , Receptores de Esteroides/fisiología , Receptores de Hormona Tiroidea/fisiología , Factores de Transcripción/fisiología , Células 3T3-L1 , Adipocitos/metabolismo , Adipocitos/fisiología , Animales , Desdiferenciación Celular/genética , Línea Celular , Proliferación Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo/genética , Ayuno/metabolismo , Ayuno/fisiología , Inflamación/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo , Receptores de Hormona Tiroidea/genética , Receptores de Hormona Tiroidea/metabolismo , Transducción de Señal/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transfección
19.
Mol Endocrinol ; 22(9): 2038-48, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18562627

RESUMEN

We previously identified the small molecule harmine as a regulator of peroxisome proliferator activated-receptor gamma (PPARgamma) and adipocyte differentiation. In an effort to identify signaling pathways mediating harmine's effects, we performed transcriptional profiling of 3T3-F442A preadipocytes. Inhibitor of DNA biding 2 (Id2) was identified as a gene rapidly induced by harmine but not by PPARgamma agonists. Id2 is also induced in 3T3-L1 preadipocytes treated with dexamethasone, 3-isobutyl-1-methylxanthine, and insulin, suggesting that Id2 regulation is a common feature of the adipogenic program. Stable overexpression of Id2 in preadipocytes promotes expression of PPARgamma and enhances morphological differentiation and lipid accumulation. Conversely, small interfering RNA-mediated knockdown of Id2 antagonizes adipocyte differentiation. Mice lacking Id2 expression display reduced adiposity, and embryonic fibroblasts derived from these mice exhibit reduced PPARgamma expression and a diminished capacity for adipocyte differentiation. Finally, Id2 expression is elevated in adipose tissues of obese mice and humans. These results outline a role for Id2 in the modulation of PPARgamma expression and adipogenesis and underscore the utility of adipogenic small molecules as tools to dissect adipocyte biology.


Asunto(s)
Adipocitos/metabolismo , Proteína 2 Inhibidora de la Diferenciación/genética , Proteína 2 Inhibidora de la Diferenciación/metabolismo , PPAR gamma/genética , Células 3T3 , Células 3T3-L1 , Adipocitos/citología , Adipocitos/efectos de los fármacos , Adipogénesis/genética , Adipogénesis/fisiología , Adiposidad , Animales , Diferenciación Celular , Expresión Génica/efectos de los fármacos , Perfilación de la Expresión Génica , Harmina/farmacología , Humanos , Proteína 2 Inhibidora de la Diferenciación/antagonistas & inhibidores , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Interferencia de ARN , Transducción de Señal , Proteínas Wnt/metabolismo
20.
Dev Cell ; 48(2): 200-214.e6, 2019 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-30554999

RESUMEN

Animals must adjust their metabolism as they progress through development in order to meet the needs of each stage in the life cycle. Here, we show that the dHNF4 nuclear receptor acts at the onset of Drosophila adulthood to direct an essential switch in lipid metabolism. Lipid stores are consumed shortly after metamorphosis but contribute little to energy metabolism. Rather, dHNF4 directs their conversion to very long chain fatty acids and hydrocarbons, which waterproof the animal to preserve fluid homeostasis. Similarly, HNF4α is required in mouse hepatocytes for the expression of fatty acid elongases that contribute to a waterproof epidermis, suggesting that this pathway is conserved through evolution. This developmental switch in Drosophila lipid metabolism promotes lifespan and desiccation resistance in adults and suppresses hallmarks of diabetes, including elevated glucose levels and intolerance to dietary sugars. These studies establish dHNF4 as a regulator of the adult metabolic state.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Metabolismo Energético/fisiología , Factor Nuclear 4 del Hepatocito/metabolismo , Homeostasis/fisiología , Metabolismo de los Lípidos/fisiología , Animales , Drosophila melanogaster/crecimiento & desarrollo , Ácidos Grasos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA