Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Genes Dev ; 33(23-24): 1657-1672, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31727774

RESUMEN

In obesity, adipose tissue undergoes dynamic remodeling processes such as adipocyte hypertrophy, hypoxia, immune responses, and adipocyte death. However, whether and how invariant natural killer T (iNKT) cells contribute to adipose tissue remodeling are elusive. In this study, we demonstrate that iNKT cells remove unhealthy adipocytes and stimulate the differentiation of healthy adipocytes. In obese adipose tissue, iNKT cells were abundantly found nearby dead adipocytes. FasL-positive adipose iNKT cells exerted cytotoxic effects to eliminate hypertrophic and pro-inflammatory Fas-positive adipocytes. Furthermore, in vivo adipocyte-lineage tracing mice model showed that activation of iNKT cells by alpha-galactosylceramide promoted adipocyte turnover, eventually leading to potentiation of the insulin-dependent glucose uptake ability in adipose tissue. Collectively, our data propose a novel role of adipose iNKT cells in the regulation of adipocyte turnover in obesity.


Asunto(s)
Adipocitos/citología , Tejido Adiposo/citología , Tejido Adiposo/inmunología , Muerte Celular/fisiología , Activación de Linfocitos/fisiología , Células T Asesinas Naturales/fisiología , Obesidad/fisiopatología , Células 3T3 , Adipocitos/inmunología , Adipocitos/metabolismo , Animales , Proliferación Celular , Proteína Ligando Fas/metabolismo , Ratones , Ratones Endogámicos C57BL , Receptor fas/metabolismo
2.
Proc Natl Acad Sci U S A ; 116(24): 11936-11945, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31160440

RESUMEN

Accumulating evidence suggests that subcutaneous and visceral adipose tissues are differentially associated with metabolic disorders. In obesity, subcutaneous adipose tissue is beneficial for metabolic homeostasis because of repressed inflammation. However, the underlying mechanism remains unclear. Here, we demonstrate that γ-aminobutyric acid (GABA) sensitivity is crucial in determining fat depot-selective adipose tissue macrophage (ATM) infiltration in obesity. In diet-induced obesity, GABA reduced monocyte migration in subcutaneous inguinal adipose tissue (IAT), but not in visceral epididymal adipose tissue (EAT). Pharmacological modulation of the GABAB receptor affected the levels of ATM infiltration and adipose tissue inflammation in IAT, but not in EAT, and GABA administration ameliorated systemic insulin resistance and enhanced insulin-dependent glucose uptake in IAT, accompanied by lower inflammatory responses. Intriguingly, compared with adipose-derived stem cells (ADSCs) from EAT, IAT-ADSCs played key roles in mediating GABA responses that repressed ATM infiltration in high-fat diet-fed mice. These data suggest that selective GABA responses in IAT contribute to fat depot-selective suppression of inflammatory responses and protection from insulin resistance in obesity.


Asunto(s)
Tejido Adiposo/metabolismo , Inflamación/metabolismo , Obesidad/metabolismo , Células Madre/metabolismo , Tejido Subcutáneo/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Adipocitos/metabolismo , Adiposidad/genética , Animales , Dieta Alta en Grasa/efectos adversos , Femenino , Humanos , Insulina/metabolismo , Grasa Intraabdominal/metabolismo , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
3.
J Biol Chem ; 293(36): 13974-13988, 2018 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-30042231

RESUMEN

Lipid droplets are specialized cellular organelles that contain neutral lipid metabolites and play dynamic roles in energy homeostasis. Perilipin 1 (Plin1), one of the major lipid droplet-binding proteins, is highly expressed in adipocytes. In mice, Plin1 deficiency impairs peripheral insulin sensitivity, accompanied with reduced fat mass. However, the mechanisms underlying insulin resistance in lean Plin1 knockout (Plin1-/-) mice are largely unknown. The current study demonstrates that Plin1 deficiency promotes inflammatory responses and lipolysis in adipose tissue, resulting in insulin resistance. M1-type adipose tissue macrophages (ATMs) were higher in Plin1-/- than in Plin1+/+ mice on normal chow diet. Moreover, using lipidomics analysis, we discovered that Plin1-/- adipocytes promoted secretion of pro-inflammatory lipid metabolites such as prostaglandins, which potentiated monocyte migration. In lean Plin1-/- mice, insulin resistance was relieved by macrophage depletion with clodronate, implying that elevated pro-inflammatory ATMs might be attributable for insulin resistance under Plin1 deficiency. Together, these data suggest that Plin1 is required to restrain fat loss and pro-inflammatory responses in adipose tissue by reducing futile lipolysis to maintain metabolic homeostasis.


Asunto(s)
Tejido Adiposo/patología , Inflamación/etiología , Metabolismo de los Lípidos , Perilipina-1/deficiencia , Adipocitos/metabolismo , Animales , Resistencia a la Insulina , Lipólisis , Macrófagos/patología , Ratones , Ratones Noqueados
4.
Hepatology ; 60(3): 844-57, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24425205

RESUMEN

UNLABELLED: Sterol regulatory element binding protein1c (SREBP1c) is a key transcription factor for de novo lipogenesis during the postprandial state. During nutritional deprivation, hepatic SREBP1c is rapidly suppressed by fasting signals to prevent lipogenic pathways. However, the molecular mechanisms that control SREBP1c turnover in response to fasting status are not thoroughly understood. To elucidate which factors are involved in the inactivation of SREBP1c, we attempted to identify SREBP1c-interacting proteins by mass spectrometry analysis. Since we observed that ring finger protein20 (RNF20) ubiquitin ligase was identified as one of SREBP1c-interacting proteins, we hypothesized that fasting signaling would promote SREBP1c degradation in an RNF20-dependent manner. In this work, we demonstrate that RNF20 physically interacts with SREBP1c, leading to degradation of SREBP1c via ubiquitination. In accordance with these findings, RNF20 represses the transcriptional activity of SREBP1c and turns off the expression of lipogenic genes that are targets of SREBP1c. In contrast, knockdown of RNF20 stimulates the expression of SREBP1c and lipogenic genes and induces lipogenic activity in primary hepatocytes. Furthermore, activation of protein kinase A (PKA) with glucagon or forskolin enhances the expression of RNF20 and potentiates the ubiquitination of SREBP1c via RNF20. In wild-type and db/db mice, adenoviral overexpression of RNF20 markedly suppresses FASN promoter activity and reduces the level of hepatic triglycerides, accompanied by a decrease in the hepatic lipogenic program. Here, we reveal that RNF20-induced SREBP1c ubiquitination down-regulates hepatic lipogenic activity upon PKA activation. CONCLUSION: RNF20 acts as a negative regulator of hepatic fatty acid metabolism through degradation of SREBP1c upon PKA activation. Knowledge regarding this process enhances our understanding of how SREBP1c is able to turn off hepatic lipid metabolism during nutritional deprivation.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Metabolismo de los Lípidos/fisiología , Hígado/química , Hígado/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Ubiquitina-Proteína Ligasas/fisiología , Animales , Células COS , Chlorocebus aethiops , Hígado Graso/metabolismo , Regulación de la Expresión Génica , Células HeLa , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Estado Nutricional , Estabilidad Proteica , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/antagonistas & inhibidores
6.
Hepatology ; 57(4): 1366-77, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23152128

RESUMEN

UNLABELLED: Recent evidence suggests that obese animals exhibit increased endoplasmic reticulum (ER) stress in the liver and adipose tissue. Although ER stress is closely associated with lipid homeostasis, it is largely unknown how ER stress contributes to hepatic steatosis. In this study, we demonstrate that the induction of ER stress stimulates hepatic steatosis through increased expression of the hepatic very low-density lipoprotein receptor (VLDLR). Among the unfolded protein response sensors, the protein kinase RNA-like ER kinase-activating transcription factor 4 signaling pathway was required for hepatic VLDLR up-regulation. In primary hepatocytes, ER stress-dependent VLDLR expression induced intracellular triglyceride accumulation in the presence of very low-density lipoprotein. Moreover, ER stress-dependent hepatic steatosis was diminished in the livers of VLDLR-deficient and apolipoprotein E-deficient mice compared with wild-type mice. In addition, the VLDLR-deficient mice exhibited decreased hepatic steatosis upon high-fat diet feeding. CONCLUSION: These data suggest that ER stress-dependent expression of hepatic VLDLR leads to hepatic steatosis by increasing lipoprotein delivery to the liver, which might be a novel mechanism explaining ER stress-induced hepatic steatosis.


Asunto(s)
Estrés del Retículo Endoplásmico/fisiología , Hígado Graso/fisiopatología , Receptores de LDL/fisiología , Regulación hacia Arriba/fisiología , Factor de Transcripción Activador 4/metabolismo , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Modelos Animales de Enfermedad , Hígado Graso/metabolismo , Lipoproteínas/metabolismo , Hígado/metabolismo , Ratones , Ratones Noqueados , Receptores de LDL/deficiencia , Receptores de LDL/genética , Triglicéridos/metabolismo
7.
J Lipid Res ; 53(7): 1277-86, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22493094

RESUMEN

In this study, we demonstrate that activation of AMP-activated protein kinase (AMPK) with glabridin alleviates adiposity and hyperlipidemia in obesity. In several obese rodent models, glabridin decreased body weight and adiposity with a concomitant reduction in fat cell size. Further, glabridin ameliorated fatty liver and plasma levels of triglyceride and cholesterol. In accordance with these findings, glabridin suppressed the expression of lipogenic genes such as sterol regulatory element binding transcription factor (SREBP)-1c, fatty acid synthase (FAS), acetyl-CoA carboxylase (ACC), and stearoyl-CoA desaturase (SCD)-1 in white adipose tissues and liver, whereas it elevated the expression of fatty acid oxidation genes such as carnitine palmitoyl transferase (CPT)1, acyl-CoA oxidase (ACO), and peroxisome proliferator-activated receptor (PPAR)α in muscle. Moreover, glabridin enhanced phosphorylation of AMPK in muscle and liver and promoted fatty acid oxidation by modulating mitochondrial activity. Together, these data suggest that glabridin is a novel AMPK activator that would exert therapeutic effects in obesity-related metabolic disorders.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Adiposidad/efectos de los fármacos , Isoflavonas/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Obesidad/tratamiento farmacológico , Fenoles/farmacología , Animales , Peso Corporal/efectos de los fármacos , Ácidos Grasos/metabolismo , Ratones , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Obesidad/metabolismo , Fosforilación/efectos de los fármacos
8.
Biochem Biophys Res Commun ; 417(2): 800-6, 2012 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-22197824

RESUMEN

We have previously reported anti-obesity effects of Lysimachia foenum-graecum in high-fat diet (HFD)-induced obesity model. Here we isolated a triterpene saponin foenumoside B as an active component of L. foenum-graecum. Foenumoside B blocked the differentiation of 3T3-L1 preadipocytes in a dose-dependent manner with an IC50 of 0.2 µg/ml in adipogenesis assay and suppressed the induction of PPARγ, the master regulator of adipogenesis. Foenumoside B induced the activation of AMP-activated protein kinase (AMPK), and modulated the expression of genes involved in lipid metabolism towards lipid breakdown in differentiated adipocytes. In mouse model, oral administration of foenumoside B (10mg/kg/day for 6 weeks) reduced HFD-induced body weight gain significantly without affecting food intake. Treatment of foenumoside B suppressed lipid accumulation in white adipose tissues and the liver, and lowered blood levels of glucose, triglycerides, ALT, and AST in HFD-induced obese mice. Consistent with the in vitro results, foenumoside B activated AMPK signaling, suppressed the expression of lipogenic genes, and enhanced the expression of lipolytic genes in vivo. Foenumoside B also blocked HFD-induced proinflammatory cytokine production in adipose tissue, suggesting its protective role against insulin resistance. Taken together, these findings demonstrate that foenumoside B represents the anti-obesity effects of L. foenum-graecum, and suggest therapeutic potential of foenumoside B in obesity and obesity-related metabolic diseases.


Asunto(s)
Adipocitos/efectos de los fármacos , Adipogénesis/efectos de los fármacos , Fármacos Antiobesidad/uso terapéutico , Dieta Alta en Grasa/efectos adversos , Obesidad/tratamiento farmacológico , Primulaceae/química , Saponinas/uso terapéutico , Células 3T3-L1 , Quinasas de la Proteína-Quinasa Activada por el AMP , Adipocitos/citología , Animales , Fármacos Antiobesidad/farmacología , Activación Enzimática , Expresión Génica/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología , Proteínas Quinasas/biosíntesis , Saponinas/farmacología
9.
Cell Rep ; 41(11): 111806, 2022 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-36516764

RESUMEN

In mammals, brown adipose tissue (BAT) is specialized to conduct non-shivering thermogenesis for survival under cold acclimation. Although emerging evidence suggests that lipid metabolites are essential for heat generation in cold-activated BAT, the underlying mechanisms of lipid uptake in BAT have not been thoroughly understood. Here, we show that very-low-density lipoprotein (VLDL) uptaken by VLDL receptor (VLDLR) plays important roles in thermogenic execution in BAT. Compared with wild-type mice, VLDLR knockout mice exhibit impaired thermogenic features. Mechanistically, VLDLR-mediated VLDL uptake provides energy sources for mitochondrial oxidation via lysosomal processing, subsequently enhancing thermogenic activity in brown adipocytes. Moreover, the VLDL-VLDLR axis potentiates peroxisome proliferator activated receptor (PPAR)ß/δ activity with thermogenic gene expression in BAT. Accordingly, VLDL-induced thermogenic capacity is attenuated in brown-adipocyte-specific PPARß/δ knockout mice. Collectively, these data suggest that the VLDL-VLDLR axis in brown adipocytes is a key factor for thermogenic execution during cold exposure.


Asunto(s)
Tejido Adiposo Pardo , PPAR-beta , Ratones , Animales , Tejido Adiposo Pardo/metabolismo , PPAR-beta/metabolismo , Lipoproteínas VLDL/metabolismo , Termogénesis/genética , Adipocitos Marrones/metabolismo , Ratones Noqueados , Mamíferos
10.
Nat Commun ; 13(1): 3268, 2022 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-35672324

RESUMEN

Thermogenic adipocytes generate heat to maintain body temperature against hypothermia in response to cold. Although tight regulation of thermogenesis is required to prevent energy sources depletion, the molecular details that tune thermogenesis are not thoroughly understood. Here, we demonstrate that adipocyte hypoxia-inducible factor α (HIFα) plays a key role in calibrating thermogenic function upon cold and re-warming. In beige adipocytes, HIFα attenuates protein kinase A (PKA) activity, leading to suppression of thermogenic activity. Mechanistically, HIF2α suppresses PKA activity by inducing miR-3085-3p expression to downregulate PKA catalytic subunit α (PKA Cα). Ablation of adipocyte HIF2α stimulates retention of beige adipocytes, accompanied by increased PKA Cα during re-warming after cold stimuli. Moreover, administration of miR-3085-3p promotes beige-to-white transition via downregulation of PKA Cα and mitochondrial abundance in adipocyte HIF2α deficient mice. Collectively, these findings suggest that HIF2α-dependent PKA regulation plays an important role as a thermostat through dynamic remodeling of beige adipocytes.


Asunto(s)
Adipocitos Beige , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Subunidades Catalíticas de Proteína Quinasa Dependientes de AMP Cíclico/metabolismo , MicroARNs , Adipocitos , Adipocitos Beige/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Frío , Ratones , MicroARNs/metabolismo , Termogénesis/genética
11.
Cell Metab ; 34(3): 458-472.e6, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35021043

RESUMEN

In mammals, white adipose tissues are largely divided into visceral epididymal adipose tissue (EAT) and subcutaneous inguinal adipose tissue (IAT) with distinct metabolic properties. Although emerging evidence suggests that subpopulations of adipose stem cells (ASCs) would be important to explain fat depot differences, ASCs of two fat depots have not been comparatively investigated. Here, we characterized heterogeneous ASCs and examined the effects of intrinsic and tissue micro-environmental factors on distinct ASC features. We demonstrated that ASC subpopulations in EAT and IAT exhibited different molecular features with three adipogenic stages. ASC transplantation experiments revealed that intrinsic ASC features primarily determined their adipogenic potential. Upon obesogenic stimuli, EAT-specific SDC1+ ASCs promoted fibrotic remodeling, whereas IAT-specific CXCL14+ ASCs suppressed macrophage infiltration. Moreover, IAT-specific BST2high ASCs exhibited a high potential to become beige adipocytes. Collectively, our data broaden the understanding of ASCs with new insights into the origin of white fat depot differences.


Asunto(s)
Adipocitos , Tejido Adiposo , Adipocitos/metabolismo , Adipogénesis , Tejido Adiposo/metabolismo , Animales , Mamíferos , Células Madre/metabolismo , Grasa Subcutánea/metabolismo
12.
Diabetes ; 70(1): 182-195, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33046512

RESUMEN

Becn1/Beclin-1 is a core component of the class III phosphatidylinositol 3-kinase required for autophagosome formation and vesicular trafficking. Although Becn1 has been implicated in numerous diseases such as cancer, aging, and neurodegenerative disease, the role of Becn1 in white adipose tissue and related metabolic diseases remains elusive. In this study, we show that adipocyte-specific Becn1 knockout mice develop severe lipodystrophy, leading to adipose tissue inflammation, hepatic steatosis, and insulin resistance. Ablation of Becn1 in adipocytes stimulates programmed cell death in a cell-autonomous manner, accompanied by elevated endoplasmic reticulum (ER) stress gene expression. Furthermore, we observed that Becn1 depletion sensitized mature adipocytes to ER stress, leading to accelerated cell death. Taken together, these data suggest that adipocyte Becn1 would serve as a crucial player for adipocyte survival and adipose tissue homeostasis.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo Blanco/metabolismo , Beclina-1/metabolismo , Resistencia a la Insulina/genética , Lipodistrofia/metabolismo , Enfermedades Metabólicas/metabolismo , Animales , Beclina-1/genética , Hígado Graso/genética , Hígado Graso/metabolismo , Homeostasis/genética , Inflamación/genética , Inflamación/metabolismo , Lipodistrofia/genética , Enfermedades Metabólicas/genética , Ratones , Ratones Noqueados
13.
Mol Pharmacol ; 78(5): 877-85, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20724462

RESUMEN

Activation of peroxisome proliferator-activated receptors (PPARs) have been implicated in the treatment of metabolic disorders with different mechanisms; PPARα agonists promote fatty acid oxidation and reduce hyperlipidemia, whereas PPARγ agonists regulate lipid redistribution from visceral fat to subcutaneous fat and enhance insulin sensitivity. To achieve combined benefits from activated PPARs on lipid metabolism and insulin sensitivity, a number of PPARα/γ dual agonists have been developed. However, several adverse effects such as weight gain and organ failure of PPARα/γ dual agonists have been reported. By use of virtual ligand screening, we identified and characterized a novel PPARα/γ dual agonist, (R)-1-(4-(2-(5-methyl-2-p-tolyloxazol-4-yl)ethoxy)benzyl)piperidine-2-carboxylic acid (CG301360), exhibiting the improvement in insulin sensitivity and lipid metabolism. CG301360 selectively stimulated transcriptional activities of PPARα and PPARγ and induced expression of their target genes in a PPARα- and PPARγ-dependent manner. In cultured cells, CG301360 enhanced fatty acid oxidation and glucose uptake and it reduced pro-inflammatory gene expression. In db/db mice, CG301360 also restored insulin sensitivity and lipid homeostasis. Collectively, these data suggest that CG301360 would be a novel PPARα/γ agonist, which might be a potential lead compound to develop against insulin resistance and hyperlipidemia.


Asunto(s)
Resistencia a la Insulina , Metabolismo de los Lípidos/efectos de los fármacos , Oxazoles/farmacología , PPAR alfa/agonistas , PPAR delta/agonistas , Ácidos Pipecólicos/farmacología , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Animales , Células Cultivadas , Ciclooxigenasa 2/biosíntesis , Citocinas/biosíntesis , Ácidos Grasos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Metaloproteinasa 9 de la Matriz/biosíntesis , Ratones , Ratones Obesos , Oxidación-Reducción , PPAR alfa/fisiología , PPAR delta/fisiología , Estereoisomerismo , Transcripción Genética
14.
Front Immunol ; 11: 598566, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33584664

RESUMEN

Accumulating evidence reveals that adipose tissue is an immunologically active organ that exerts multiple impacts on the regulation of systemic energy metabolism. Adipose tissue immunity is modulated by the interactions between adipocytes and various immune cells. Nevertheless, the underlying mechanisms that control inter-cellular interactions between adipocytes and immune cells in adipose tissue have not been thoroughly elucidated. Recently, it has been demonstrated that adipocytes utilize lipid metabolites as a key mediator to initiate and mediate diverse adipose tissue immune responses. Adipocytes present lipid antigens and secrete lipid metabolites to determine adipose immune tones. In addition, the interactions between adipocytes and adipose immune cells are engaged in the control of adipocyte fate and functions upon metabolic stimuli. In this review, we discuss an integrated view of how adipocytes communicate with adipose immune cells using lipid metabolites. Also, we briefly discuss the newly discovered roles of adipose stem cells in the regulation of adipose tissue immunity.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo/inmunología , Tejido Adiposo/metabolismo , Metabolismo de los Lípidos , Animales , Presentación de Antígeno , Biomarcadores , Susceptibilidad a Enfermedades , Metabolismo Energético , Humanos , Inmunidad Innata , Inmunomodulación , Lípidos/inmunología , Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/metabolismo , Paniculitis/etiología , Paniculitis/metabolismo , Paniculitis/patología , Células Madre/metabolismo
15.
Nat Commun ; 11(1): 578, 2020 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-31996685

RESUMEN

Lipid droplets (LDs) are key subcellular organelles for regulating lipid metabolism. Although several subcellular organelles participate in lipid metabolism, it remains elusive whether physical contacts between subcellular organelles and LDs might be involved in lipolysis upon nutritional deprivation. Here, we demonstrate that peroxisomes and peroxisomal protein PEX5 mediate fasting-induced lipolysis by stimulating adipose triglyceride lipase (ATGL) translocation onto LDs. During fasting, physical contacts between peroxisomes and LDs are increased by KIFC3-dependent movement of peroxisomes toward LDs, which facilitates spatial translocations of ATGL onto LDs. In addition, PEX5 could escort ATGL to contact points between peroxisomes and LDs in the presence of fasting cues. Moreover, in adipocyte-specific PEX5-knockout mice, the recruitment of ATGL onto LDs was defective and fasting-induced lipolysis is attenuated. Collectively, these data suggest that physical contacts between peroxisomes and LDs are required for spatiotemporal translocation of ATGL, which is escorted by PEX5 upon fasting, to maintain energy homeostasis.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Ayuno/efectos adversos , Gotas Lipídicas/metabolismo , Lipólisis/fisiología , Receptor de la Señal 1 de Direccionamiento al Peroxisoma/metabolismo , Peroxisomas/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Análisis Espacio-Temporal , Células 3T3-L1/metabolismo , Adipocitos/metabolismo , Animales , Caenorhabditis elegans , Señales (Psicología) , Citoesqueleto , Cinesinas/metabolismo , Metabolismo de los Lípidos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nutrientes , Receptor de la Señal 1 de Direccionamiento al Peroxisoma/genética , Peroxisomas/genética , Transducción de Señal
16.
Diabetes ; 69(1): 20-34, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31604693

RESUMEN

Adipose tissue is the key organ coordinating whole-body energy homeostasis. Although it has been reported that ring finger protein 20 (RNF20) regulates lipid metabolism in the liver and kidney, the roles of RNF20 in adipose tissue have not been explored. Here, we demonstrate that RNF20 promotes adipogenesis by potentiating the transcriptional activity of peroxisome proliferator-activated receptor-γ (PPARγ). Under normal chow diet feeding, Rnf20 defective (Rnf20 +/- ) mice exhibited reduced fat mass with smaller adipocytes compared with wild-type littermates. In addition, high-fat diet-fed Rnf20 +/- mice alleviated systemic insulin resistance accompanied by a reduced expansion of fat tissue. Quantitative proteomic analyses revealed significantly decreased levels of PPARγ target proteins in adipose tissue of Rnf20 +/- mice. Mechanistically, RNF20 promoted proteasomal degradation of nuclear corepressor 1 (NCoR1), which led to stimulation of the transcriptional activity of PPARγ. Collectively, these data suggest that RNF20-NCoR1 is a novel axis in adipocyte biology through fine-tuning the transcriptional activity of PPARγ.


Asunto(s)
Adipocitos/metabolismo , Co-Represor 1 de Receptor Nuclear/metabolismo , PPAR gamma/metabolismo , Ubiquitina-Proteína Ligasas/fisiología , Animales , Dieta Alta en Grasa , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Ratones Transgénicos , Obesidad/etiología , Obesidad/genética , Obesidad/metabolismo , Obesidad/patología , PPAR gamma/fisiología , Proteolisis , Transactivadores/genética , Transactivadores/fisiología , Ubiquitina-Proteína Ligasas/genética
17.
Mol Cell Biol ; 39(2)2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30397073

RESUMEN

Oxygen is a key molecule for efficient energy production in living organisms. Although aerobic organisms have adaptive processes to survive in low-oxygen environments, it is poorly understood how lipolysis, the first step of energy production from stored lipid metabolites, would be modulated during hypoxia. Here, we demonstrate that fasting-induced lipolysis is downregulated by hypoxia through the hypoxia-inducible factor (HIF) signaling pathway. In Caenorhabditis elegans and mammalian adipocytes, hypoxia suppressed protein kinase A (PKA)-stimulated lipolysis, which is evolutionarily well conserved. During hypoxia, the levels of PKA activity and adipose triglyceride lipase (ATGL) protein were downregulated, resulting in attenuated fasting-induced lipolysis. In worms, HIF stabilization was sufficient to moderate the suppressive effect of hypoxia on lipolysis through ATGL and PKA inhibition. These data suggest that HIF activation under hypoxia plays key roles in the suppression of lipolysis, which might preserve energy resources in both C. elegans and mammalian adipocytes.


Asunto(s)
Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia/metabolismo , Lipasa/metabolismo , Células 3T3 , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Animales , Caenorhabditis elegans , Proteínas Portadoras/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Regulación hacia Abajo , Lípidos/fisiología , Lipólisis/efectos de los fármacos , Lipólisis/fisiología , Ratones , Nematodos , Fosforilación , Transducción de Señal
18.
Diabetes ; 68(1): 81-94, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30352876

RESUMEN

SREBP1c is a key transcription factor for de novo lipogenesis. Although SREBP1c is expressed in pancreatic islets, its physiological roles in pancreatic ß-cells are largely unknown. In this study, we demonstrate that SREBP1c regulates ß-cell compensation under metabolic stress. SREBP1c expression level was augmented in pancreatic islets from obese and diabetic animals. In pancreatic ß-cells, SREBP1c activation promoted the expression of cell cycle genes and stimulated ß-cell proliferation through its novel target gene, PAX4 Compared with SREBP1c+/+ mice, SREBP1c-/- mice showed glucose intolerance with low insulin levels. Moreover, ß-cells from SREBP1c-/- mice exhibited reduced capacity to proliferate and secrete insulin. Conversely, transplantation of SREBP1c-overexpressing islets restored insulin levels and relieved hyperglycemia in streptozotocin-induced diabetic animals. Collectively, these data suggest that pancreatic SREBP1c is a key player in mediating ß-cell compensatory responses in obesity.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Células Secretoras de Insulina/metabolismo , Factores de Transcripción Paired Box/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Animales , Apoptosis/genética , Apoptosis/fisiología , Ciclo Celular/genética , Ciclo Celular/fisiología , Línea Celular , Proliferación Celular/genética , Proliferación Celular/fisiología , Inmunoprecipitación de Cromatina , Proteínas de Homeodominio/genética , Inmunohistoquímica , Masculino , Ratones , Factores de Transcripción Paired Box/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética
19.
Mol Cell Biol ; 39(20)2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31308132

RESUMEN

Adipocytes have unique morphological traits in insulin sensitivity control. However, how the appearance of adipocytes can determine insulin sensitivity has not been understood. Here, we demonstrate that actin cytoskeleton reorganization upon lipid droplet (LD) configurations in adipocytes plays important roles in insulin-dependent glucose uptake by regulating GLUT4 trafficking. Compared to white adipocytes, brown/beige adipocytes with multilocular LDs exhibited well-developed filamentous actin (F-actin) structure and potentiated GLUT4 translocation to the plasma membrane in the presence of insulin. In contrast, LD enlargement and unilocularization in adipocytes downregulated cortical F-actin formation, eventually leading to decreased F-actin-to-globular actin (G-actin) ratio and suppression of insulin-dependent GLUT4 trafficking. Pharmacological inhibition of actin polymerization accompanied with impaired F/G-actin dynamics reduced glucose uptake in adipose tissue and conferred systemic insulin resistance in mice. Thus, our study reveals that adipocyte remodeling with different LD configurations could be an important factor to determine insulin sensitivity by modulating F/G-actin dynamics.


Asunto(s)
Actinas/metabolismo , Adipocitos/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Resistencia a la Insulina , Gotas Lipídicas/metabolismo , Citoesqueleto de Actina/metabolismo , Adipocitos/efectos de los fármacos , Adipocitos/patología , Adipocitos Blancos/metabolismo , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Respuesta al Choque por Frío , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Glucosa/metabolismo , Masculino , Ratones Endogámicos C57BL , Obesidad/metabolismo , Obesidad/patología , Transporte de Proteínas
20.
Mol Cell Biol ; 25(12): 5146-57, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15923630

RESUMEN

Glucose-6-phosphate dehydrogenase (G6PD) produces cellular NADPH, which is required for the biosynthesis of fatty acids and cholesterol. Although G6PD is required for lipogenesis, it is poorly understood whether G6PD in adipocytes is involved in energy homeostasis, such as lipid and glucose metabolism. We report here that G6PD plays a role in adipogenesis and that its increase is tightly associated with the dysregulation of lipid metabolism and insulin resistance in obesity. We observed that the enzymatic activity and expression levels of G6PD were significantly elevated in white adipose tissues of obese models, including db/db, ob/ob, and diet-induced obesity mice. In 3T3-L1 cells, G6PD overexpression stimulated the expression of most adipocyte marker genes and elevated the levels of cellular free fatty acids, triglyceride, and FFA release. Consistently, G6PD knockdown via small interfering RNA attenuated adipocyte differentiation with less lipid droplet accumulation. Surprisingly, the expression of certain adipocytokines such as tumor necrosis factor alpha and resistin was increased, whereas that of adiponectin was decreased in G6PD overexpressed adipocytes. In accordance with these results, overexpression of G6PD impaired insulin signaling and suppressed insulin-dependent glucose uptake in adipocytes. Taken together, these data strongly suggest that aberrant increase of G6PD in obese and/or diabetic subjects would alter lipid metabolism and adipocytokine expression, thereby resulting in failure of lipid homeostasis and insulin resistance in adipocytes.


Asunto(s)
Glucosafosfato Deshidrogenasa/metabolismo , Resistencia a la Insulina , Metabolismo de los Lípidos , Obesidad/fisiopatología , Células 3T3-L1 , Adiponectina , Tejido Adiposo/citología , Tejido Adiposo/crecimiento & desarrollo , Tejido Adiposo/metabolismo , Animales , Biomarcadores , Diferenciación Celular , Regulación Enzimológica de la Expresión Génica , Glucosafosfato Deshidrogenasa/genética , Homeostasis , Hormonas Ectópicas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Mutagénesis Sitio-Dirigida , Obesidad/enzimología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Resistina , Transducción de Señal/fisiología , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA