Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 133
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Nat Metab ; 6(2): 273-289, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38286821

RESUMEN

Mitochondrial dysfunction is a characteristic trait of human and rodent obesity, insulin resistance and fatty liver disease. Here we show that high-fat diet (HFD) feeding causes mitochondrial fragmentation in inguinal white adipocytes from male mice, leading to reduced oxidative capacity by a process dependent on the small GTPase RalA. RalA expression and activity are increased in white adipocytes after HFD. Targeted deletion of RalA in white adipocytes prevents fragmentation of mitochondria and diminishes HFD-induced weight gain by increasing fatty acid oxidation. Mechanistically, RalA increases fission in adipocytes by reversing the inhibitory Ser637 phosphorylation of the fission protein Drp1, leading to more mitochondrial fragmentation. Adipose tissue expression of the human homolog of Drp1, DNM1L, is positively correlated with obesity and insulin resistance. Thus, chronic activation of RalA plays a key role in repressing energy expenditure in obese adipose tissue by shifting the balance of mitochondrial dynamics toward excessive fission, contributing to weight gain and metabolic dysfunction.


Asunto(s)
Resistencia a la Insulina , Proteínas de Unión al GTP ral , Animales , Humanos , Masculino , Ratones , Adipocitos Blancos/metabolismo , Tejido Adiposo/metabolismo , Obesidad/etiología , Obesidad/metabolismo , Aumento de Peso , Proteínas de Unión al GTP ral/metabolismo
2.
Cell Metab ; 35(6): 1009-1021.e9, 2023 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-37084733

RESUMEN

Insulin inhibits gluconeogenesis and stimulates glucose conversion to glycogen and lipids. How these activities are coordinated to prevent hypoglycemia and hepatosteatosis is unclear. Fructose-1,6-bisphosphatase (FBP1) is rate controlling for gluconeogenesis. However, inborn human FBP1 deficiency does not cause hypoglycemia unless accompanied by fasting or starvation, which also trigger paradoxical hepatomegaly, hepatosteatosis, and hyperlipidemia. Hepatocyte FBP1-ablated mice exhibit identical fasting-conditional pathologies along with AKT hyperactivation, whose inhibition reversed hepatomegaly, hepatosteatosis, and hyperlipidemia but not hypoglycemia. Surprisingly, fasting-mediated AKT hyperactivation is insulin dependent. Independently of its catalytic activity, FBP1 prevents insulin hyperresponsiveness by forming a stable complex with AKT, PP2A-C, and aldolase B (ALDOB), which specifically accelerates AKT dephosphorylation. Enhanced by fasting and weakened by elevated insulin, FBP1:PP2A-C:ALDOB:AKT complex formation, which is disrupted by human FBP1 deficiency mutations or a C-terminal FBP1 truncation, prevents insulin-triggered liver pathologies and maintains lipid and glucose homeostasis. Conversely, an FBP1-derived complex disrupting peptide reverses diet-induced insulin resistance.


Asunto(s)
Fructosa , Hipoglucemia , Humanos , Ratones , Animales , Fructosa-Bifosfatasa/genética , Proteínas Proto-Oncogénicas c-akt , Insulina , Hepatomegalia/complicaciones , Hipoglucemia/etiología , Glucosa
3.
JCI Insight ; 7(17)2022 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-35917178

RESUMEN

Cardiovascular diseases, especially atherosclerosis and its complications, are a leading cause of death. Inhibition of the noncanonical IκB kinases TANK-binding kinase 1 and IKKε with amlexanox restores insulin sensitivity and glucose homeostasis in diabetic mice and human patients. Here we report that amlexanox improves diet-induced hypertriglyceridemia and hypercholesterolemia in Western diet-fed (WD-fed) Ldlr-/- mice and protects against atherogenesis. Amlexanox ameliorated dyslipidemia, inflammation, and vascular dysfunction through synergistic actions that involve upregulation of bile acid synthesis to increase cholesterol excretion. Transcriptomic profiling demonstrated an elevated expression of key bile acid synthesis genes. Furthermore, we found that amlexanox attenuated monocytosis, eosinophilia, and vascular dysfunction during WD-induced atherosclerosis. These findings demonstrate the potential of amlexanox as a therapy for hypercholesterolemia and atherosclerosis.


Asunto(s)
Aterosclerosis , Diabetes Mellitus Experimental , Hipercolesterolemia , Aminopiridinas , Animales , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/metabolismo , Aterosclerosis/prevención & control , Ácidos y Sales Biliares , Humanos , Hipercolesterolemia/tratamiento farmacológico , Quinasa I-kappa B/metabolismo , Ratones , Proteínas Serina-Treonina Quinasas
4.
Cell Rep ; 40(1): 111008, 2022 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-35793637

RESUMEN

Compositional oscillations of the gut microbiome are essential for normal peripheral circadian rhythms, both of which are disrupted in diet-induced obesity (DIO). Although time-restricted feeding (TRF) maintains circadian synchrony and protects against DIO, its impact on the dynamics of the cecal gut microbiome is modest. Thus, other regions of the gut, particularly the ileum, the nexus for incretin and bile acid signaling, may play an important role in entraining peripheral circadian rhythms. We demonstrate the effect of diet and feeding rhythms on the ileal microbiome composition and transcriptome in mice. The dynamic rhythms of ileal microbiome composition and transcriptome are dampened in DIO. TRF partially restores diurnal rhythms of the ileal microbiome and transcriptome, increases GLP-1 release, and alters the ileal bile acid pool and farnesoid X receptor (FXR) signaling, which could explain how TRF exerts its metabolic benefits. Finally, we provide a web resource for exploration of ileal microbiome and transcriptome circadian data.


Asunto(s)
Microbiota , Transcriptoma , Animales , Ácidos y Sales Biliares , Dieta , Conducta Alimentaria , Íleon/metabolismo , Ratones , Obesidad/metabolismo , Transcriptoma/genética
5.
J Clin Invest ; 132(2)2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-34847077

RESUMEN

The dysregulation of energy homeostasis in obesity involves multihormone resistance. Although leptin and insulin resistance have been well characterized, catecholamine resistance remains largely unexplored. Murine ß3-adrenergic receptor expression in adipocytes is orders of magnitude higher compared with that of other isoforms. While resistant to classical desensitization pathways, its mRNA (Adrb3) and protein expression are dramatically downregulated after ligand exposure (homologous desensitization). ß3-Adrenergic receptor downregulation also occurs after high-fat diet feeding, concurrent with catecholamine resistance and elevated inflammation. This downregulation is recapitulated in vitro by TNF-α treatment (heterologous desensitization). Both homologous and heterologous desensitization of Adrb3 were triggered by induction of the pseudokinase TRIB1 downstream of the EPAC/RAP2A/PI-PLC pathway. TRIB1 in turn degraded the primary transcriptional activator of Adrb3, CEBPα. EPAC/RAP inhibition enhanced catecholamine-stimulated lipolysis and energy expenditure in obese mice. Moreover, adipose tissue expression of genes in this pathway correlated with body weight extremes in a cohort of genetically diverse mice and with BMI in 2 independent cohorts of humans. These data implicate a signaling axis that may explain reduced hormone-stimulated lipolysis in obesity and resistance to therapeutic interventions with ß3-adrenergic receptor agonists.


Asunto(s)
Adipocitos/metabolismo , Catecolaminas/farmacología , Regulación hacia Abajo/efectos de los fármacos , Resistencia a Medicamentos/efectos de los fármacos , Obesidad/metabolismo , Receptores Adrenérgicos beta 3/metabolismo , Células 3T3-L1 , Animales , Regulación hacia Abajo/genética , Resistencia a Medicamentos/genética , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/genética , Lipólisis/efectos de los fármacos , Lipólisis/genética , Masculino , Ratones , Obesidad/tratamiento farmacológico , Obesidad/genética , Receptores Adrenérgicos beta 3/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
6.
JCI Insight ; 7(1)2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-34813504

RESUMEN

While current thinking posits that insulin signaling to glucose transporter 4 (GLUT4) exocytic translocation and glucose uptake in skeletal muscle and adipocytes is controlled by phosphorylation-based signaling, many proteins in this pathway are acetylated on lysine residues. However, the importance of acetylation and lysine acetyltransferases to insulin-stimulated glucose uptake is incompletely defined. Here, we demonstrate that combined loss of the acetyltransferases E1A binding protein p300 (p300) and cAMP response element binding protein binding protein (CBP) in mouse skeletal muscle caused a complete loss of insulin-stimulated glucose uptake. Similarly, brief (i.e., 1 hour) pharmacological inhibition of p300/CBP acetyltransferase activity recapitulated this phenotype in human and rodent myotubes, 3T3-L1 adipocytes, and mouse muscle. Mechanistically, these effects were due to p300/CBP-mediated regulation of GLUT4 exocytic translocation and occurred downstream of Akt signaling. Taken together, we highlight a fundamental role for acetylation and p300/CBP in the direct regulation of insulin-stimulated glucose transport in skeletal muscle and adipocytes.


Asunto(s)
Adipocitos , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteína p300 Asociada a E1A/metabolismo , Glucosa/metabolismo , Músculo Esquelético , Adipocitos/citología , Adipocitos/metabolismo , Animales , Femenino , Insulina/metabolismo , Masculino , Ratones , Músculo Esquelético/citología , Músculo Esquelético/metabolismo
7.
Exp Mol Med ; 53(11): 1697-1705, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34848839

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver disease and is strongly associated with obesity-related ectopic fat accumulation in the liver. Hepatic lipid accumulation encompasses a histological spectrum ranging from simple steatosis to nonalcoholic steatohepatitis (NASH), which can progress to cirrhosis and hepatocellular carcinoma. Given that dysregulated hepatic lipid metabolism may be an onset factor in NAFLD, understanding how hepatic lipid metabolism is modulated in healthy subjects and which steps are dysregulated in NAFLD subjects is crucial to identify effective therapeutic targets. Additionally, hepatic inflammation is involved in chronic hepatocyte damage during NAFLD progression. As a key immune signaling hub that mediates NF-κB activation, the IκB kinase (IKK) complex, including IKKα, IKKß, and IKKγ (NEMO), has been studied as a crucial regulator of the hepatic inflammatory response and hepatocyte survival. Notably, TANK-binding kinase 1 (TBK1), an IKK-related kinase, has recently been revealed as a potential link between hepatic inflammation and energy metabolism. Here, we review (1) the biochemical steps of hepatic lipid metabolism; (2) dysregulated lipid metabolism in obesity and NAFLD; and (3) the roles of IKKs and TBK1 in obesity and NAFLD.


Asunto(s)
Quinasa I-kappa B/metabolismo , Metabolismo de los Lípidos , Hígado/metabolismo , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Biomarcadores , Citocinas/metabolismo , Susceptibilidad a Enfermedades , Regulación de la Expresión Génica , Humanos , Mediadores de Inflamación , Hígado/patología , Enfermedad del Hígado Graso no Alcohólico/patología , Transducción de Señal
8.
Nature ; 599(7884): 296-301, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34707293

RESUMEN

Adipocytes increase energy expenditure in response to prolonged sympathetic activation via persistent expression of uncoupling protein 1 (UCP1)1,2. Here we report that the regulation of glycogen metabolism by catecholamines is critical for UCP1 expression. Chronic ß-adrenergic activation leads to increased glycogen accumulation in adipocytes expressing UCP1. Adipocyte-specific deletion of a scaffolding protein, protein targeting to glycogen (PTG), reduces glycogen levels in beige adipocytes, attenuating UCP1 expression and responsiveness to cold or ß-adrenergic receptor-stimulated weight loss in obese mice. Unexpectedly, we observed that glycogen synthesis and degradation are increased in response to catecholamines, and that glycogen turnover is required to produce reactive oxygen species leading to the activation of p38 MAPK, which drives UCP1 expression. Thus, glycogen has a key regulatory role in adipocytes, linking glucose metabolism to thermogenesis.


Asunto(s)
Adipocitos/metabolismo , Glucosa/metabolismo , Glucógeno/metabolismo , Homeostasis , Termogénesis , Adaptación Fisiológica , Adipocitos Beige/metabolismo , Animales , Frío , Metabolismo Energético , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Ratones , Ratones Noqueados , Proteína Desacopladora 1/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
9.
Sci Transl Med ; 13(604)2021 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-34321322

RESUMEN

Type 2 diabetes (T2D) is a metabolic disorder characterized by hyperglycemia, hyperinsulinemia, and insulin resistance (IR). During the early phase of T2D, insulin synthesis and secretion by pancreatic ß cells is enhanced, which can lead to proinsulin misfolding that aggravates endoplasmic reticulum (ER) protein homeostasis in ß cells. Moreover, increased circulating insulin may contribute to fatty liver disease. Medical interventions aimed at alleviating ER stress in ß cells while maintaining optimal insulin secretion are therefore an attractive therapeutic strategy for T2D. Previously, we demonstrated that germline Chop gene deletion preserved ß cells in high-fat diet (HFD)-fed mice and in leptin receptor-deficient db/db mice. In the current study, we further investigated whether targeting Chop/Ddit3 specifically in murine ß cells conferred therapeutic benefits. First, we showed that Chop deletion in ß cells alleviated ß cell ER stress and delayed glucose-stimulated insulin secretion (GSIS) in HFD-fed mice. Second, ß cell-specific Chop deletion prevented liver steatosis and hepatomegaly in aged HFD-fed mice without affecting basal glucose homeostasis. Third, we provide mechanistic evidence that Chop depletion reduces ER Ca2+ buffering capacity and modulates glucose-induced islet Ca2+ oscillations, leading to transcriptional changes of ER chaperone profile ("ER remodeling"). Last, we demonstrated that a GLP1-conjugated Chop antisense oligonucleotide strategy recapitulated the reduction in liver triglycerides and pancreatic insulin content. In summary, our results demonstrate that Chop depletion in ß cells provides a therapeutic strategy to alleviate dysregulated insulin secretion and consequent fatty liver disease in T2D.


Asunto(s)
Diabetes Mellitus Tipo 2 , Hígado Graso , Células Secretoras de Insulina , Animales , Diabetes Mellitus Tipo 2/metabolismo , Dieta Alta en Grasa/efectos adversos , Estrés del Retículo Endoplásmico , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Ratones , Ratones Endogámicos C57BL
10.
Cell Rep ; 35(13): 109331, 2021 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-34192547

RESUMEN

The contribution of adipose-derived FGF21 to energy homeostasis is unclear. Here we show that browning of inguinal white adipose tissue (iWAT) by ß-adrenergic agonists requires autocrine FGF21 signaling. Adipose-specific deletion of the FGF21 co-receptor ß-Klotho renders mice unresponsive to ß-adrenergic stimulation. In contrast, mice with liver-specific ablation of FGF21, which eliminates circulating FGF21, remain sensitive to ß-adrenergic browning of iWAT. Concordantly, transgenic overexpression of FGF21 in adipocytes promotes browning in a ß-Klotho-dependent manner without increasing circulating FGF21. Mechanistically, we show that ß-adrenergic stimulation of thermogenic gene expression requires FGF21 in adipocytes to promote phosphorylation of phospholipase C-γ and mobilization of intracellular calcium. Moreover, we find that the ß-adrenergic-dependent increase in circulating FGF21 occurs through an indirect mechanism in which fatty acids released by adipocyte lipolysis subsequently activate hepatic PPARα to increase FGF21 expression. These studies identify FGF21 as a cell-autonomous autocrine regulator of adipose tissue function.


Asunto(s)
Adipocitos/metabolismo , Comunicación Autocrina , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación de la Expresión Génica , Termogénesis/genética , Células 3T3-L1 , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Agonistas Adrenérgicos beta , Animales , Comunicación Autocrina/genética , Factores de Crecimiento de Fibroblastos/sangre , Factores de Crecimiento de Fibroblastos/genética , Lipólisis , Hígado/metabolismo , Ratones , Especificidad de Órganos , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Adrenérgicos beta 3/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo
11.
Nat Commun ; 12(1): 2876, 2021 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-34001883

RESUMEN

Activation of non-shivering thermogenesis is considered a promising approach to lower body weight in obesity. p62 deficiency in adipocytes reduces systemic energy expenditure but its role in sustaining mitochondrial function and thermogenesis remains unresolved. NBR1 shares a remarkable structural similarity with p62 and can interact with p62 through their respective PB1 domains. However, the physiological relevance of NBR1 in metabolism, as compared to that of p62, was not clear. Here we show that whole-body and adipocyte-specific ablation of NBR1 reverts the obesity phenotype induced by p62 deficiency by restoring global energy expenditure and thermogenesis in brown adipose tissue. Impaired adrenergic-induced browning of p62-deficient adipocytes is rescued by NBR1 inactivation, unveiling a negative role of NBR1 in thermogenesis under conditions of p62 loss. We demonstrate that upon p62 inactivation, NBR1 represses the activity of PPARγ, establishing an unexplored p62/NBR1-mediated paradigm in adipocyte thermogenesis that is critical for the control of obesity.


Asunto(s)
Adipocitos/metabolismo , Péptidos y Proteínas de Señalización Intracelular/deficiencia , PPAR gamma/metabolismo , Proteína Sequestosoma-1/deficiencia , Termogénesis , Tejido Adiposo Pardo/citología , Tejido Adiposo Pardo/metabolismo , Animales , Animales Recién Nacidos , Núcleo Celular/metabolismo , Células Cultivadas , Metabolismo Energético/genética , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , PPAR gamma/genética , Unión Proteica , Receptor alfa X Retinoide/genética , Receptor alfa X Retinoide/metabolismo , Proteína Sequestosoma-1/genética
13.
J Clin Invest ; 131(10)2021 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-33822771

RESUMEN

The protein kinases IKKε and TBK1 are activated in liver and fat in mouse models of obesity. We have previously demonstrated that treatment with the IKKε/TBK1 inhibitor amlexanox produces weight loss and relieves insulin resistance in obese animals and patients. While amlexanox treatment caused a transient reduction in food intake, long-term weight loss was attributable to increased energy expenditure via FGF21-dependent beiging of white adipose tissue (WAT). Amlexanox increased FGF21 synthesis and secretion in several tissues. Interestingly, although hepatic secretion determined circulating levels, it was dispensable for regulating energy expenditure. In contrast, adipocyte-secreted FGF21 may have acted as an autocrine factor that led to adipose tissue browning and weight loss in obese mice. Moreover, increased energy expenditure was an important determinant of improved insulin sensitivity by amlexanox. Conversely, the immediate reductions in fasting blood glucose observed with acute amlexanox treatment were mediated by the suppression of hepatic glucose production via activation of STAT3 by adipocyte-secreted IL-6. These findings demonstrate that amlexanox improved metabolic health via FGF21 action in adipocytes to increase energy expenditure via WAT beiging and that adipocyte-derived IL-6 has an endocrine role in decreasing gluconeogenesis via hepatic STAT3 activation, thereby producing a coordinated improvement in metabolic parameters.


Asunto(s)
Aminopiridinas/farmacología , Glucemia/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Gluconeogénesis/efectos de los fármacos , Quinasa I-kappa B/metabolismo , Hígado/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Glucemia/genética , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/genética , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/genética , Factores de Crecimiento de Fibroblastos/genética , Gluconeogénesis/genética , Quinasa I-kappa B/genética , Interleucina-6/genética , Interleucina-6/metabolismo , Ratones , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/genética , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo
14.
J Clin Invest ; 131(1)2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-33393497

RESUMEN

The molecular mechanisms of cellular insulin action have been the focus of much investigation since the discovery of the hormone 100 years ago. Insulin action is impaired in metabolic syndrome, a condition known as insulin resistance. The actions of the hormone are initiated by binding to its receptor on the surface of target cells. The receptor is an α2ß2 heterodimer that binds to insulin with high affinity, resulting in the activation of its tyrosine kinase activity. Once activated, the receptor can phosphorylate a number of intracellular substrates that initiate discrete signaling pathways. The tyrosine phosphorylation of some substrates activates phosphatidylinositol-3-kinase (PI3K), which produces polyphosphoinositides that interact with protein kinases, leading to activation of the kinase Akt. Phosphorylation of Shc leads to activation of the Ras/MAP kinase pathway. Phosphorylation of SH2B2 and of Cbl initiates activation of G proteins such as TC10. Activation of Akt and other protein kinases produces phosphorylation of a variety of substrates, including transcription factors, GTPase-activating proteins, and other kinases that control key metabolic events. Among the cellular processes controlled by insulin are vesicle trafficking, activities of metabolic enzymes, transcriptional factors, and degradation of insulin itself. Together these complex processes are coordinated to ensure glucose homeostasis.


Asunto(s)
Resistencia a la Insulina , Insulina/metabolismo , Sistema de Señalización de MAP Quinasas , Síndrome Metabólico/metabolismo , Receptor de Insulina/metabolismo , Animales , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Insulina/historia , Síndrome Metabólico/historia , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo
15.
Front Immunol ; 11: 592949, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33193441

RESUMEN

Adipocytes and adipose tissue play critical roles in the regulation of metabolic homeostasis. In obesity and obesity-associated metabolic diseases, immune cells infiltrate into adipose tissues. Interaction between adipocytes and immune cells re-shapes both metabolic and immune properties of adipose tissue and dramatically changes metabolic set points. Both the expression and activity of the non-canonical IKK family member TBK1 are induced in adipose tissues during diet-induced obesity. TBK1 plays important roles in the regulation of both metabolism and inflammation in adipose tissue and thus affects glucose and energy metabolism. Here we review the regulation and functions of TBK1 and the molecular mechanisms by which TBK1 regulates both metabolism and inflammation in adipose tissue. Finally, we discuss the potential of a TBK1/IKKε inhibitor as a new therapy for metabolic diseases.


Asunto(s)
Adipocitos/inmunología , Adipocitos/metabolismo , Metabolismo Energético , Inmunomodulación , Proteínas Serina-Treonina Quinasas/metabolismo , Tejido Adiposo/inmunología , Tejido Adiposo/metabolismo , Animales , Biomarcadores , Susceptibilidad a Enfermedades , Humanos , Enfermedades Metabólicas/etiología , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/patología , Obesidad/etiología , Obesidad/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal
16.
Cell Metab ; 32(6): 1012-1027.e7, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33152322

RESUMEN

Hepatic TANK (TRAF family member associated NFκB activator)-binding kinase 1 (TBK1) activity is increased during obesity, and administration of a TBK1 inhibitor reduces fatty liver. Surprisingly, liver-specific TBK1 knockout in mice produces fatty liver by reducing fatty acid oxidation. TBK1 functions as a scaffolding protein to localize acyl-CoA synthetase long-chain family member 1 (ACSL1) to mitochondria, which generates acyl-CoAs that are channeled for ß-oxidation. TBK1 is induced during fasting and maintained in the unphosphorylated, inactive state, enabling its high affinity binding to ACSL1 in mitochondria. In TBK1-deficient liver, ACSL1 is shifted to the endoplasmic reticulum to promote fatty acid re-esterification in lieu of oxidation in response to fasting, which accelerates hepatic lipid accumulation. The impaired fatty acid oxidation in TBK1-deficient hepatocytes is rescued by the expression of kinase-dead TBK1. Thus, TBK1 operates as a rheostat to direct the fate of fatty acids in hepatocytes, supporting oxidation when inactive during fasting and promoting re-esterification when activated during obesity.


Asunto(s)
Coenzima A Ligasas/metabolismo , Ácidos Grasos/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Obesidad/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxidación-Reducción
17.
J Biol Chem ; 295(34): 12279-12289, 2020 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-32651233

RESUMEN

Nonalcoholic fatty liver diseases (NAFLDs), especially nonalcoholic steatohepatitis (NASH), have become a major cause of liver transplant and liver-associated death. However, the pathogenesis of NASH is still unclear. Currently, there is no FDA-approved medication to treat this devastating disease. AMP-activated protein kinase (AMPK) senses energy status and regulates metabolic processes to maintain homeostasis. The activity of AMPK is regulated by the availability of nutrients, such as carbohydrates, lipids, and amino acids. AMPK activity is increased by nutrient deprivation and inhibited by overnutrition, inflammation, and hypersecretion of certain anabolic hormones, such as insulin, during obesity. The repression of hepatic AMPK activity permits the transition from simple steatosis to hepatocellular death; thus, activation might ameliorate multiple aspects of NASH. Here we review the pathogenesis of NAFLD and the impact of AMPK activity state on hepatic steatosis, inflammation, liver injury, and fibrosis during the transition of NAFL to NASH and liver failure.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Fallo Hepático/enzimología , Hígado/enzimología , Enfermedad del Hígado Graso no Alcohólico/enzimología , Hipernutrición/enzimología , Humanos , Hígado/patología , Fallo Hepático/patología , Enfermedad del Hígado Graso no Alcohólico/patología , Hipernutrición/patología
18.
Nat Metab ; 2(7): 620-634, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32694788

RESUMEN

Catecholamines stimulate the mobilization of stored triglycerides in adipocytes to provide fatty acids (FAs) for other tissues. However, a large proportion is taken back up and either oxidized or re-esterified. What controls the disposition of these FAs in adipocytes remains unknown. Here, we report that catecholamines redirect FAs for oxidation through the phosphorylation of signal transducer and activator of transcription 3 (STAT3). Adipocyte STAT3 is phosphorylated upon activation of ß-adrenergic receptors, and in turn suppresses FA re-esterification to promote FA oxidation. Adipocyte-specific Stat3 KO mice exhibit normal rates of lipolysis, but exhibit defective lipolysis-driven oxidative metabolism, resulting in reduced energy expenditure and increased adiposity when they are on a high-fat diet. This previously unappreciated, non-genomic role of STAT3 explains how sympathetic activation can increase both lipolysis and FA oxidation in adipocytes, revealing a new regulatory axis in metabolism.


Asunto(s)
Adipocitos Blancos/metabolismo , Catecolaminas/farmacología , Ácidos Grasos no Esterificados/metabolismo , Factor de Transcripción STAT3/metabolismo , Agonistas Adrenérgicos beta/farmacología , Animales , Dieta Alta en Grasa , Metabolismo Energético , Ésteres/metabolismo , Lipólisis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/metabolismo , Oxidación-Reducción , Fosforilación , Factor de Transcripción STAT3/genética
19.
Science ; 367(6478): 652-660, 2020 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-32029622

RESUMEN

Liver cell death has an essential role in nonalcoholic steatohepatitis (NASH). The activity of the energy sensor adenosine monophosphate (AMP)-activated protein kinase (AMPK) is repressed in NASH. Liver-specific AMPK knockout aggravated liver damage in mouse NASH models. AMPK phosphorylated proapoptotic caspase-6 protein to inhibit its activation, keeping hepatocyte apoptosis in check. Suppression of AMPK activity relieved this inhibition, rendering caspase-6 activated in human and mouse NASH. AMPK activation or caspase-6 inhibition, even after the onset of NASH, improved liver damage and fibrosis. Once phosphorylation was decreased, caspase-6 was activated by caspase-3 or -7. Active caspase-6 cleaved Bid to induce cytochrome c release, generating a feedforward loop that leads to hepatocyte death. Thus, the AMPK-caspase-6 axis regulates liver damage in NASH, implicating AMPK and caspase-6 as therapeutic targets.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Caspasa 6/metabolismo , Hígado/enzimología , Hígado/patología , Enfermedad del Hígado Graso no Alcohólico/enzimología , Enfermedad del Hígado Graso no Alcohólico/patología , Proteínas Quinasas Activadas por AMP/genética , Animales , Apoptosis/genética , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Inhibidores de Caspasas/farmacología , Inhibidores de Caspasas/uso terapéutico , Activación Enzimática , Hepatocitos/enzimología , Hepatocitos/patología , Humanos , Ratones , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Fosforilación
20.
Am J Hum Genet ; 106(2): 246-255, 2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-32004447

RESUMEN

Ral (Ras-like) GTPases play an important role in the control of cell migration and have been implicated in Ras-mediated tumorigenicity. Recently, variants in RALA were also described as a cause of intellectual disability and developmental delay, indicating the relevance of this pathway to neuropediatric diseases. Here, we report the identification of bi-allelic variants in RALGAPA1 (encoding Ral GTPase activating protein catalytic alpha subunit 1) in four unrelated individuals with profound neurodevelopmental disability, muscular hypotonia, feeding abnormalities, recurrent fever episodes, and infantile spasms . Dysplasia of corpus callosum with focal thinning of the posterior part and characteristic facial features appeared to be unifying findings. RalGAPA1 was absent in the fibroblasts derived from two affected individuals suggesting a loss-of-function effect of the RALGAPA1 variants. Consequently, RalA activity was increased in these cell lines, which is in keeping with the idea that RalGAPA1 deficiency causes a constitutive activation of RalA. Additionally, levels of RalGAPB, a scaffolding subunit of the RalGAP complex, were dramatically reduced, indicating a dysfunctional RalGAP complex. Moreover, RalGAPA1 deficiency clearly increased cell-surface levels of lipid raft components in detached fibroblasts, which might indicate that anchorage-dependence of cell growth signaling is disturbed. Our findings indicate that the dysregulation of the RalA pathway has an important impact on neuronal function and brain development. In light of the partially overlapping phenotype between RALA- and RALGAPA1-associated diseases, it appears likely that dysregulation of the RalA signaling pathway leads to a distinct group of genetic syndromes that we suggest could be named RALopathies.


Asunto(s)
Trastornos de Alimentación y de la Ingestión de Alimentos/etiología , Proteínas Activadoras de GTPasa/genética , Hipotonía Muscular/etiología , Mutación , Proteínas del Tejido Nervioso/genética , Trastornos del Neurodesarrollo/etiología , Espasmos Infantiles/etiología , Alelos , Movimiento Celular , Proliferación Celular , Preescolar , Familia , Trastornos de Alimentación y de la Ingestión de Alimentos/patología , Femenino , Humanos , Lactante , Masculino , Hipotonía Muscular/patología , Trastornos del Neurodesarrollo/patología , Fenotipo , Espasmos Infantiles/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA