Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cell Metab ; 36(6): 1371-1393.e7, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38718791

RESUMEN

The role and molecular mechanisms of intermittent fasting (IF) in non-alcoholic steatohepatitis (NASH) and its transition to hepatocellular carcinoma (HCC) are unknown. Here, we identified that an IF 5:2 regimen prevents NASH development as well as ameliorates established NASH and fibrosis without affecting total calorie intake. Furthermore, the IF 5:2 regimen blunted NASH-HCC transition when applied therapeutically. The timing, length, and number of fasting cycles as well as the type of NASH diet were critical parameters determining the benefits of fasting. Combined proteome, transcriptome, and metabolome analyses identified that peroxisome-proliferator-activated receptor alpha (PPARα) and glucocorticoid-signaling-induced PCK1 act co-operatively as hepatic executors of the fasting response. In line with this, PPARα targets and PCK1 were reduced in human NASH. Notably, only fasting initiated during the active phase of mice robustly induced glucocorticoid signaling and free-fatty-acid-induced PPARα signaling. However, hepatocyte-specific glucocorticoid receptor deletion only partially abrogated the hepatic fasting response. In contrast, the combined knockdown of Ppara and Pck1 in vivo abolished the beneficial outcomes of fasting against inflammation and fibrosis. Moreover, overexpression of Pck1 alone or together with Ppara in vivo lowered hepatic triglycerides and steatosis. Our data support the notion that the IF 5:2 regimen is a promising intervention against NASH and subsequent liver cancer.


Asunto(s)
Carcinoma Hepatocelular , Ayuno , Neoplasias Hepáticas , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico , PPAR alfa , Fosfoenolpiruvato Carboxiquinasa (GTP) , PPAR alfa/metabolismo , Animales , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Humanos , Ratones , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/metabolismo , Masculino , Fosfoenolpiruvato Carboxiquinasa (GTP)/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Transducción de Señal , Ayuno Intermitente
3.
Proc Natl Acad Sci U S A ; 118(17)2021 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-33875597

RESUMEN

G protein-coupled receptor 182 (GPR182) has been shown to be expressed in endothelial cells; however, its ligand and physiological role has remained elusive. We found GPR182 to be expressed in microvascular and lymphatic endothelial cells of most organs and to bind with nanomolar affinity the chemokines CXCL10, CXCL12, and CXCL13. In contrast to conventional chemokine receptors, binding of chemokines to GPR182 did not induce typical downstream signaling processes, including Gq- and Gi-mediated signaling or ß-arrestin recruitment. GPR182 showed relatively high constitutive activity in regard to ß-arrestin recruitment and rapidly internalized in a ligand-independent manner. In constitutive GPR182-deficient mice, as well as after induced endothelium-specific loss of GPR182, we found significant increases in the plasma levels of CXCL10, CXCL12, and CXCL13. Global and induced endothelium-specific GPR182-deficient mice showed a significant decrease in hematopoietic stem cells in the bone marrow as well as increased colony-forming units of hematopoietic progenitors in the blood and the spleen. Our data show that GPR182 is a new atypical chemokine receptor for CXCL10, CXCL12, and CXCL13, which is involved in the regulation of hematopoietic stem cell homeostasis.


Asunto(s)
Receptores Acoplados a Proteínas G/metabolismo , Animales , Quimiocina CXCL10 , Quimiocina CXCL12 , Quimiocina CXCL13 , Quimiocinas/metabolismo , Células Endoteliales/metabolismo , Femenino , Células HEK293 , Células Madre Hematopoyéticas/metabolismo , Homeostasis , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Quimiocina/metabolismo , Receptores Acoplados a Proteínas G/genética , Transducción de Señal/fisiología , beta-Arrestinas/metabolismo
4.
Nat Med ; 25(4): 641-655, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30936549

RESUMEN

Non-alcoholic fatty liver disease ranges from steatosis to non-alcoholic steatohepatitis (NASH), potentially progressing to cirrhosis and hepatocellular carcinoma (HCC). Here, we show that platelet number, platelet activation and platelet aggregation are increased in NASH but not in steatosis or insulin resistance. Antiplatelet therapy (APT; aspirin/clopidogrel, ticagrelor) but not nonsteroidal anti-inflammatory drug (NSAID) treatment with sulindac prevented NASH and subsequent HCC development. Intravital microscopy showed that liver colonization by platelets depended primarily on Kupffer cells at early and late stages of NASH, involving hyaluronan-CD44 binding. APT reduced intrahepatic platelet accumulation and the frequency of platelet-immune cell interaction, thereby limiting hepatic immune cell trafficking. Consequently, intrahepatic cytokine and chemokine release, macrovesicular steatosis and liver damage were attenuated. Platelet cargo, platelet adhesion and platelet activation but not platelet aggregation were identified as pivotal for NASH and subsequent hepatocarcinogenesis. In particular, platelet-derived GPIbα proved critical for development of NASH and subsequent HCC, independent of its reported cognate ligands vWF, P-selectin or Mac-1, offering a potential target against NASH.


Asunto(s)
Plaquetas/metabolismo , Neoplasias Hepáticas/sangre , Neoplasias Hepáticas/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/sangre , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Animales , Plaquetas/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Citocinas/metabolismo , Gránulos Citoplasmáticos/efectos de los fármacos , Gránulos Citoplasmáticos/metabolismo , Endotelio/efectos de los fármacos , Endotelio/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/patología , Humanos , Receptores de Hialuranos/metabolismo , Ácido Hialurónico/metabolismo , Macrófagos del Hígado/efectos de los fármacos , Macrófagos del Hígado/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Ratones Transgénicos , Agregación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Recuento de Plaquetas
5.
EMBO Mol Med ; 10(8)2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29973382

RESUMEN

Most antidiabetic drugs treat disease symptoms rather than adipose tissue dysfunction as a key pathogenic cause in the metabolic syndrome and type 2 diabetes. Pharmacological targeting of adipose tissue through the nuclear receptor PPARg, as exemplified by glitazone treatments, mediates efficacious insulin sensitization. However, a better understanding of the context-specific PPARg responses is required for the development of novel approaches with reduced side effects. Here, we identified the transcriptional cofactor Cited4 as a target and mediator of rosiglitazone in human and murine adipocyte progenitor cells, where it promoted specific sets of the rosiglitazone-dependent transcriptional program. In mice, Cited4 was required for the proper induction of thermogenic expression by Rosi specifically in subcutaneous fat. This phenotype had high penetrance in females only and was not evident in beta-adrenergically stimulated browning. Intriguingly, this specific defect was associated with reduced capacity for systemic thermogenesis and compromised insulin sensitization upon therapeutic rosiglitazone treatment in female but not male mice. Our findings on Cited4 function reveal novel unexpected aspects of the pharmacological targeting of PPARg.


Asunto(s)
Adipocitos/efectos de los fármacos , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hipoglucemiantes/uso terapéutico , Rosiglitazona/uso terapéutico , Factores de Transcripción/metabolismo , Adipocitos/metabolismo , Animales , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Humanos , Masculino , Ratones , Terapia Molecular Dirigida , PPAR gamma/metabolismo , Factores Sexuales , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Termogénesis , Factores de Transcripción/biosíntesis , Transcripción Genética/efectos de los fármacos , Proteína Desacopladora 1/biosíntesis
6.
Circulation ; 137(24): 2592-2608, 2018 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-29353241

RESUMEN

BACKGROUND: Nutrients are transported through endothelial cells before being metabolized in muscle cells. However, little is known about the regulation of endothelial transport processes. Notch signaling is a critical regulator of metabolism and angiogenesis during development. Here, we studied how genetic and pharmacological manipulation of endothelial Notch signaling in adult mice affects endothelial fatty acid transport, cardiac angiogenesis, and heart function. METHODS: Endothelial-specific Notch inhibition was achieved by conditional genetic inactivation of Rbp-jκ in adult mice to analyze fatty acid metabolism and heart function. Wild-type mice were treated with neutralizing antibodies against the Notch ligand Delta-like 4. Fatty acid transport was studied in cultured endothelial cells and transgenic mice. RESULTS: Treatment of wild-type mice with Delta-like 4 neutralizing antibodies for 8 weeks impaired fractional shortening and ejection fraction in the majority of mice. Inhibition of Notch signaling specifically in the endothelium of adult mice by genetic ablation of Rbp-jκ caused heart hypertrophy and failure. Impaired heart function was preceded by alterations in fatty acid metabolism and an increase in cardiac blood vessel density. Endothelial Notch signaling controlled the expression of endothelial lipase, Angptl4, CD36, and Fabp4, which are all needed for fatty acid transport across the vessel wall. In endothelial-specific Rbp-jκ-mutant mice, lipase activity and transendothelial transport of long-chain fatty acids to muscle cells were impaired. In turn, lipids accumulated in the plasma and liver. The attenuated supply of cardiomyocytes with long-chain fatty acids was accompanied by higher glucose uptake, increased concentration of glycolysis intermediates, and mTOR-S6K signaling. Treatment with the mTOR inhibitor rapamycin or displacing glucose as cardiac substrate by feeding a ketogenic diet prolonged the survival of endothelial-specific Rbp-jκ-deficient mice. CONCLUSIONS: This study identifies Notch signaling as a novel regulator of fatty acid transport across the endothelium and as an essential repressor of angiogenesis in the adult heart. The data imply that the endothelium controls cardiomyocyte metabolism and function.


Asunto(s)
Endotelio Vascular/metabolismo , Ácidos Grasos/metabolismo , Miocardio/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Remodelación Vascular , Proteínas Adaptadoras Transductoras de Señales , Angiopoyetinas/genética , Angiopoyetinas/metabolismo , Animales , Antígenos CD36/genética , Antígenos CD36/metabolismo , Proteínas de Unión al Calcio , Endotelio Vascular/citología , Proteínas de Unión a Ácidos Grasos/genética , Proteínas de Unión a Ácidos Grasos/metabolismo , Ácidos Grasos/genética , Glucosa/genética , Glucosa/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Miocitos Cardíacos/metabolismo , Neovascularización Fisiológica , Receptores Notch/genética , Proteínas Quinasas S6 Ribosómicas/genética , Proteínas Quinasas S6 Ribosómicas/metabolismo , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
7.
Cell Rep ; 20(6): 1422-1434, 2017 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-28793265

RESUMEN

Increased pro-inflammatory signaling is a hallmark of metabolic dysfunction in obesity and diabetes. Although both inflammatory and energy substrate handling processes represent critical layers of metabolic control, their molecular integration sites remain largely unknown. Here, we identify the heterodimerization interface between the α and ß subunits of transcription factor GA-binding protein (GAbp) as a negative target of tumor necrosis factor alpha (TNF-α) signaling. TNF-α prevented GAbpα and ß complex formation via reactive oxygen species (ROS), leading to the non-energy-dependent transcriptional inactivation of AMP-activated kinase (AMPK) ß1, which was identified as a direct hepatic GAbp target. Impairment of AMPKß1, in turn, elevated downstream cellular cholesterol biosynthesis, and hepatocyte-specific ablation of GAbpα induced systemic hypercholesterolemia and early macro-vascular lesion formation in mice. As GAbpα and AMPKß1 levels were also found to correlate in obese human patients, the ROS-GAbp-AMPK pathway may represent a key component of a hepato-vascular axis in diabetic long-term complications.


Asunto(s)
Aterosclerosis/metabolismo , Factor de Transcripción de la Proteína de Unión a GA/metabolismo , Hepatocitos/metabolismo , Hipercolesterolemia/metabolismo , Proteínas Quinasas/metabolismo , Transducción de Señal , Quinasas de la Proteína-Quinasa Activada por el AMP , Animales , Aterosclerosis/etiología , Aterosclerosis/patología , Línea Celular , Células Cultivadas , Colesterol/metabolismo , Factor de Transcripción de la Proteína de Unión a GA/química , Hipercolesterolemia/complicaciones , Masculino , Ratones , Ratones Endogámicos C57BL , Multimerización de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
8.
Nat Commun ; 7: 13267, 2016 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-27827363

RESUMEN

Obesity-related insulin resistance represents the core component of the metabolic syndrome, promoting glucose intolerance, pancreatic beta cell failure and type 2 diabetes. Efficient and safe insulin sensitization and glucose control remain critical therapeutic aims to prevent diabetic late complications Here, we identify transforming growth factor beta-like stimulated clone (TSC) 22 D4 as a molecular determinant of insulin signalling and glucose handling. Hepatic TSC22D4 inhibition both prevents and reverses hyperglycaemia, glucose intolerance and insulin resistance in diabetes mouse models. TSC22D4 exerts its effects on systemic glucose homeostasis-at least in part-through the direct transcriptional regulation of the small secretory protein lipocalin 13 (LCN13). Human diabetic patients display elevated hepatic TSC22D4 expression, which correlates with decreased insulin sensitivity, hyperglycaemia and LCN13 serum levels. Our results establish TSC22D4 as a checkpoint in systemic glucose metabolism in both mice and humans, and propose TSC22D4 inhibition as an insulin sensitizing option in diabetes therapy.


Asunto(s)
Glucemia/metabolismo , Diabetes Mellitus Tipo 2/genética , Hiperglucemia/genética , Resistencia a la Insulina/genética , Factores de Transcripción/genética , Animales , Línea Celular , Diabetes Mellitus Tipo 2/sangre , Femenino , Regulación de la Expresión Génica , Humanos , Hiperglucemia/sangre , Lipocalinas/genética , Lipocalinas/metabolismo , Hígado/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Transcripción/metabolismo
9.
J Clin Invest ; 126(9): 3263-78, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27548521

RESUMEN

Dietary protein intake is linked to an increased incidence of type 2 diabetes (T2D). Although dietary protein dilution (DPD) can slow the progression of some aging-related disorders, whether this strategy affects the development and risk for obesity-associated metabolic disease such as T2D is unclear. Here, we determined that DPD in mice and humans increases serum markers of metabolic health. In lean mice, DPD promoted metabolic inefficiency by increasing carbohydrate and fat oxidation. In nutritional and polygenic murine models of obesity, DPD prevented and curtailed the development of impaired glucose homeostasis independently of obesity and food intake. DPD-mediated metabolic inefficiency and improvement of glucose homeostasis were independent of uncoupling protein 1 (UCP1), but required expression of liver-derived fibroblast growth factor 21 (FGF21) in both lean and obese mice. FGF21 expression and secretion as well as the associated metabolic remodeling induced by DPD also required induction of liver-integrated stress response-driven nuclear protein 1 (NUPR1). Insufficiency of select nonessential amino acids (NEAAs) was necessary and adequate for NUPR1 and subsequent FGF21 induction and secretion in hepatocytes in vitro and in vivo. Taken together, these data indicate that DPD promotes improved glucose homeostasis through an NEAA insufficiency-induced liver NUPR1/FGF21 axis.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Proteínas en la Dieta/administración & dosificación , Hígado/metabolismo , Tejido Adiposo/metabolismo , Adulto , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Metabolismo de los Hidratos de Carbono , Proteínas de Unión al ADN/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Glucosa/metabolismo , Hepatocitos/metabolismo , Homeostasis , Humanos , Metabolismo de los Lípidos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Neoplasias/metabolismo , Obesidad/metabolismo , Fenotipo , Proteína Desacopladora 1/metabolismo
10.
Nat Med ; 22(10): 1120-1130, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27571348

RESUMEN

Cachexia represents a fatal energy-wasting syndrome in a large number of patients with cancer that mostly results in a pathological loss of skeletal muscle and adipose tissue. Here we show that tumor cell exposure and tumor growth in mice triggered a futile energy-wasting cycle in cultured white adipocytes and white adipose tissue (WAT), respectively. Although uncoupling protein 1 (Ucp1)-dependent thermogenesis was dispensable for tumor-induced body wasting, WAT from cachectic mice and tumor-cell-supernatant-treated adipocytes were consistently characterized by the simultaneous induction of both lipolytic and lipogenic pathways. Paradoxically, this was accompanied by an inactivated AMP-activated protein kinase (Ampk), which is normally activated in peripheral tissues during states of low cellular energy. Ampk inactivation correlated with its degradation and with upregulation of the Ampk-interacting protein Cidea. Therefore, we developed an Ampk-stabilizing peptide, ACIP, which was able to ameliorate WAT wasting in vitro and in vivo by shielding the Cidea-targeted interaction surface on Ampk. Thus, our data establish the Ucp1-independent remodeling of adipocyte lipid homeostasis as a key event in tumor-induced WAT wasting, and we propose the ACIP-dependent preservation of Ampk integrity in the WAT as a concept in future therapies for cachexia.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Adipocitos Blancos/efectos de los fármacos , Tejido Adiposo Blanco/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/efectos de los fármacos , Caquexia/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Neoplasias/metabolismo , Fragmentos de Péptidos/farmacología , Proteínas Quinasas Activadas por AMP/farmacología , Adipocitos Blancos/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Caquexia/etiología , Células Cultivadas , Técnicas In Vitro , Lipogénesis/efectos de los fármacos , Lipólisis/efectos de los fármacos , Ratones , Neoplasias/complicaciones , Termogénesis/efectos de los fármacos , Proteína Desacopladora 1/efectos de los fármacos , Proteína Desacopladora 1/metabolismo
11.
EMBO Mol Med ; 8(6): 654-69, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27137487

RESUMEN

Recent studies have demonstrated that repeated short-term nutrient withdrawal (i.e. fasting) has pleiotropic actions to promote organismal health and longevity. Despite this, the molecular physiological mechanisms by which fasting is protective against metabolic disease are largely unknown. Here, we show that, metabolic control, particularly systemic and liver lipid metabolism, is aberrantly regulated in the fasted state in mouse models of metabolic dysfunction. Liver transcript assays between lean/healthy and obese/diabetic mice in fasted and fed states uncovered "growth arrest and DNA damage-inducible" GADD45ß as a dysregulated gene transcript during fasting in several models of metabolic dysfunction including ageing, obesity/pre-diabetes and type 2 diabetes, in both mice and humans. Using whole-body knockout mice as well as liver/hepatocyte-specific gain- and loss-of-function strategies, we revealed a role for liver GADD45ß in the coordination of liver fatty acid uptake, through cytoplasmic retention of FABP1, ultimately impacting obesity-driven hyperglycaemia. In summary, fasting stress-induced GADD45ß represents a liver-specific molecular event promoting adaptive metabolic function.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Ayuno , Ácidos Grasos/metabolismo , Hígado/metabolismo , Proteínas Nucleares/metabolismo , Animales , Proteínas de Unión a Ácidos Grasos/metabolismo , Ratones , Ratones Noqueados
12.
J Diabetes Res ; 2016: 2981639, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26788517

RESUMEN

Objective. Glyoxalase-1 is an enzyme detoxifying methylglyoxal (MG). MG is a potent precursor of advanced glycation endproducts which are regarded to be a key player in micro- and macrovascular damage. Yet, the role of Glo1 in atherosclerosis remains unclear. In this study, the effect of Glo1 on mouse metabolism and atherosclerosis is evaluated. Methods. Glo1 knockdown mice were fed a high fat or a standard diet for 10 weeks. Body weight and composition were investigated by Echo MRI. The PhenoMaster system was used to measure the energy expenditure. To evaluate the impact of Glo1 on atherosclerosis, Glo1(KD) mice were crossed with ApoE-knockout mice and fed a high fat diet for 14 weeks. Results. Glo1 activity was significantly reduced in heart, liver, and kidney lysates derived from Glo1(KD) mice. Yet, there was no increase in methylglyoxal-derived AGEs in all organs analyzed. The Glo1 knockdown did not affect body weight or body composition. Metabolic studies via indirect calorimetry did not show significant effects on energy expenditure. Glo1(KD) mice crossed to ApoE(-/-) mice did not show enhanced formation of atherosclerosis. Conclusion. A Glo1 knockdown does not have major short term effects on the energy expenditure or the formation of atherosclerotic plaques.


Asunto(s)
Enfermedades de la Aorta/enzimología , Aterosclerosis/enzimología , Metabolismo Energético , Lactoilglutatión Liasa/deficiencia , Animales , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/patología , Calorimetría Indirecta , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Metabolismo Energético/genética , Predisposición Genética a la Enfermedad , Riñón/enzimología , Lactoilglutatión Liasa/genética , Hígado/enzimología , Imagen por Resonancia Magnética , Masculino , Ratones Noqueados , Miocardio/enzimología , Fenotipo , Placa Aterosclerótica , Piruvaldehído/metabolismo
13.
Mol Metab ; 4(5): 406-17, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25973388

RESUMEN

OBJECTIVE: Type 2 diabetes arises from insulin resistance of peripheral tissues followed by dysfunction of ß-cells in the pancreas due to metabolic stress. Both depletion and supplementation of neutral amino acids have been discussed as strategies to improve insulin sensitivity. Here we characterise mice lacking the intestinal and renal neutral amino acid transporter B(0)AT1 (Slc6a19) as a model to study the consequences of selective depletion of neutral amino acids. METHODS: Metabolic tests, analysis of metabolite levels and signalling pathways were used to characterise mice lacking the intestinal and renal neutral amino acid transporter B(0)AT1 (Slc6a19). RESULTS: Reduced uptake of neutral amino acids in the intestine and loss of neutral amino acids in the urine causes an overload of amino acids in the lumen of the intestine and reduced systemic amino acid availability. As a result, higher levels of glucagon-like peptide 1 (GLP-1) are produced by the intestine after a meal, while the liver releases the starvation hormone fibroblast growth factor 21 (FGF21). The combination of these hormones generates a metabolic phenotype that is characterised by efficient removal of glucose, particularly by the heart, reduced adipose tissue mass, browning of subcutaneous white adipose tissue, enhanced production of ketone bodies and reduced hepatic glucose output. CONCLUSIONS: Reduced neutral amino acid availability improves glycaemic control. The epithelial neutral amino acid transporter B(0)AT1 could be a suitable target to treat type 2 diabetes.

14.
PLoS One ; 10(2): e0118534, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25714366

RESUMEN

Regulatory T (Treg) cells are critical determinants of both immune responses and metabolic control. Here we show that systemic ablation of Treg cells compromised the adaptation of whole-body energy expenditure to cold exposure, correlating with impairment in thermogenic marker gene expression and massive invasion of pro-inflammatory macrophages in brown adipose tissue (BAT). Indeed, BAT harbored a unique sub-set of Treg cells characterized by a unique gene signature. As these Treg cells respond to BAT activation upon cold exposure, this study defines a BAT-specific Treg sub-set with direct implications for the regulation of energy homeostasis in response to environmental stress.


Asunto(s)
Tejido Adiposo Pardo/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Pardo/patología , Animales , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Redes y Vías Metabólicas , Metaboloma , Metabolómica/métodos , Ratones , Fenotipo , Bazo/citología , Bazo/inmunología , Bazo/metabolismo , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Reguladores/metabolismo
15.
EMBO J ; 34(3): 344-60, 2015 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-25510864

RESUMEN

In mammals, glucocorticoids (GCs) and their intracellular receptor, the glucocorticoid receptor (GR), represent critical checkpoints in the endocrine control of energy homeostasis. Indeed, aberrant GC action is linked to severe metabolic stress conditions as seen in Cushing's syndrome, GC therapy and certain components of the Metabolic Syndrome, including obesity and insulin resistance. Here, we identify the hepatic induction of the mammalian conserved microRNA (miR)-379/410 genomic cluster as a key component of GC/GR-driven metabolic dysfunction. Particularly, miR-379 was up-regulated in mouse models of hyperglucocorticoidemia and obesity as well as human liver in a GC/GR-dependent manner. Hepatocyte-specific silencing of miR-379 substantially reduced circulating very-low-density lipoprotein (VLDL)-associated triglyceride (TG) levels in healthy mice and normalized aberrant lipid profiles in metabolically challenged animals, mediated through miR-379 effects on key receptors in hepatic TG re-uptake. As hepatic miR-379 levels were also correlated with GC and TG levels in human obese patients, the identification of a GC/GR-controlled miRNA cluster not only defines a novel layer of hormone-dependent metabolic control but also paves the way to alternative miRNA-based therapeutic approaches in metabolic dysfunction.


Asunto(s)
Glucocorticoides/metabolismo , Metabolismo de los Lípidos , Hígado/metabolismo , MicroARNs/metabolismo , Obesidad/metabolismo , Animales , Línea Celular , Femenino , Silenciador del Gen , Glucocorticoides/genética , Humanos , Lipoproteínas VLDL/genética , Lipoproteínas VLDL/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Obesos , MicroARNs/genética , Obesidad/genética , Triglicéridos/genética , Triglicéridos/metabolismo
16.
PLoS One ; 9(10): e110428, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25313899

RESUMEN

Presence of thermogenically active adipose tissue in adult humans has been inversely associated with obesity and type 2 diabetes. While it had been shown that insulin is crucial for the development of classical brown fat, its role in development and function of inducible brown-in-white (brite) adipose tissue is less clear. Here we show that insulin deficiency impaired differentiation of brite adipocytes. However, adrenergic stimulation almost fully induced the thermogenic program under these settings. Although brite differentiation of adipocytes as well as browning of white adipose tissue entailed substantially elevated glucose uptake by adipose tissue, the capacity of insulin to stimulate glucose uptake surprisingly was not higher in the brite state. Notably, in line with the insulin-independent stimulation of glucose uptake, our data revealed that brite recruitment results in induction of solute carrier family 2 (GLUT-1) expression in adipocytes and inguinal WAT. These results for the first time demonstrate that insulin signaling is neither essential for brite recruitment, nor is it improved in cells or tissues upon browning.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Glucosa/metabolismo , Insulina/metabolismo , Transducción de Señal , Adipocitos/citología , Adipocitos/metabolismo , Animales , Diferenciación Celular , Transportador 2 de Aminoácidos Excitadores/genética , Transportador 2 de Aminoácidos Excitadores/metabolismo , Expresión Génica , Masculino , Ratones , Células Madre/citología , Células Madre/metabolismo
17.
Mol Metab ; 3(2): 155-66, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24634828

RESUMEN

Disturbances in lipid homeostasis are hallmarks of severe metabolic disorders and their long-term complications, including obesity, diabetes, and atherosclerosis. Whereas elevation of triglyceride (TG)-rich very-low-density lipoproteins (VLDL) has been identified as a risk factor for cardiovascular complications, high-density lipoprotein (HDL)-associated cholesterol confers atheroprotection under obese and/or diabetic conditions. Here we show that hepatocyte-specific deficiency of transcription factor transforming growth factor ß 1-stimulated clone (TSC) 22 D1 led to a substantial reduction in HDL levels in both wild-type and obese mice, mediated through the transcriptional down-regulation of the HDL formation pathway in liver. Indeed, overexpression of TSC22D1 promoted high levels of HDL cholesterol in healthy animals, and hepatic expression of TSC22D1 was found to be aberrantly regulated in disease models of opposing energy availability. The hepatic TSC22D1 transcription factor complex may thus represent an attractive target in HDL raising strategies in obesity/diabetes-related dyslipidemia and atheroprotection.

18.
Cell Metab ; 17(4): 575-85, 2013 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-23499424

RESUMEN

Lipid mobilization (lipolysis) in white adipose tissue (WAT) critically controls lipid turnover and adiposity in humans. While the acute regulation of lipolysis has been studied in detail, the transcriptional determinants of WAT lipolytic activity remain still largely unexplored. Here we show that the genetic inactivation of transcriptional cofactor transducin beta-like-related 1(TBLR1) blunts the lipolytic response of white adipocytes through the impairment of cAMP-dependent signal transduction. Indeed, mice lacking TBLR1 in adipocytes are defective in fasting-induced lipid mobilization and, when placed on a high-fat-diet, show aggravated adiposity, glucose intolerance, and insulin resistance. TBLR1 levels are found to increase under lipolytic conditions in WAT of both human patients and mice, correlating with serum free fatty acids (FFAs). As a critical regulator of WAT cAMP signaling and lipid mobilization, proper activity of TBLR1 in adipocytes might thus represent a critical molecular checkpoint for the prevention of metabolic dysfunction in subjects with obesity-related disorders.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Movilización Lipídica/fisiología , Receptores Citoplasmáticos y Nucleares/metabolismo , Células 3T3-L1 , Animales , AMP Cíclico/metabolismo , Dieta Alta en Grasa , Ácidos Grasos no Esterificados/sangre , Humanos , Resistencia a la Insulina , Lipólisis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Obesidad/metabolismo , Obesidad/patología , Interferencia de ARN , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Receptores Adrenérgicos/genética , Receptores Adrenérgicos/metabolismo , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/genética , Transducción de Señal
19.
EMBO Mol Med ; 5(2): 294-308, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23307490

RESUMEN

In mammals, proper storage and distribution of lipids in and between tissues is essential for the maintenance of energy homeostasis. Here, we show that tumour growth triggers hepatic metabolic dysfunction as part of the cancer cachectic phenotype, particularly by reduced hepatic very-low-density-lipoprotein (VLDL) secretion and hypobetalipoproteinemia. As a molecular cachexia output pathway, hepatic levels of the transcription factor transforming growth factor beta 1-stimulated clone (TSC) 22 D4 were increased in cancer cachexia. Mimicking high cachectic levels of TSC22D4 in healthy livers led to the inhibition of hepatic VLDL release and lipogenic genes, and diminished systemic VLDL levels under both normal and high fat dietary conditions. Liver-specific ablation of TSC22D4 triggered hypertriglyceridemia through the induction of hepatic VLDL secretion. Furthermore, hepatic TSC22D4 expression levels were correlated with the degree of body weight loss and VLDL hypo-secretion in cancer cachexia, and TSC22D4 deficiency rescued tumour cell-induced metabolic dysfunction in hepatocytes. Therefore, hepatic TSC22D4 activity may represent a molecular rationale for peripheral energy deprivation in subjects with metabolic wasting diseases, including cancer cachexia.


Asunto(s)
Caquexia/metabolismo , Hígado/metabolismo , Neoplasias/complicaciones , Factores de Transcripción/metabolismo , Animales , Caquexia/etiología , Humanos , Lipoproteínas VLDL/metabolismo , Hepatopatías/etiología , Hepatopatías/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neoplasias/metabolismo , Factores de Transcripción/genética , Triglicéridos/metabolismo
20.
Am J Pathol ; 175(5): 1883-95, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19834063

RESUMEN

The role of vascular endothelial growth factor (VEGF) in renal fibrosis, tubular cyst formation, and glomerular diseases is incompletely understood. We studied a new conditional transgenic mouse system [Pax8-rtTA/(tetO)(7)VEGF], which allows increased tubular VEGF production in adult mice. The following pathology was observed. The interstitial changes consisted of a ubiquitous proliferation of peritubular capillaries and fibroblasts, followed by deposition of matrix leading to a unique kind of fibrosis, ie, healthy tubules amid a capillary-rich dense fibrotic tissue. In tubular segments with high expression of VEGF, cysts developed that were surrounded by a dense network of peritubular capillaries. The glomerular effects consisted of a proliferative enlargement of glomerular capillaries, followed by mesangial proliferation. This resulted in enlarged glomeruli with loss of the characteristic lobular structure. Capillaries became randomly embedded into mesangial nodules, losing their filtration surface. Serum VEGF levels were increased, whereas endogenous VEGF production by podocytes was down-regulated. Taken together, this study shows that systemic VEGF interferes with the intraglomerular cross-talk between podocytes and the endocapillary compartment. It suppresses VEGF secretion by podocytes but cannot compensate for the deficit. VEGF from podocytes induces a directional effect, attracting the capillaries to the lobular surface, a relevant mechanism to optimize filtration surface. Systemic VEGF lacks this effect, leading to severe deterioration in glomerular architecture, similar to that seen in diabetic nephropathy.


Asunto(s)
Quistes , Glomerulonefritis , Enfermedades Renales , Glomérulos Renales , Túbulos Renales , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Capilares/citología , Capilares/metabolismo , Capilares/patología , Quistes/metabolismo , Quistes/patología , Fibrosis/metabolismo , Fibrosis/patología , Glomerulonefritis/metabolismo , Glomerulonefritis/patología , Humanos , Hibridación in Situ , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Glomérulos Renales/citología , Glomérulos Renales/metabolismo , Glomérulos Renales/patología , Túbulos Renales/citología , Túbulos Renales/metabolismo , Túbulos Renales/patología , Ratones , Ratones Transgénicos , Podocitos/citología , Podocitos/metabolismo , Podocitos/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...