Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
EMBO Rep ; 24(10): e55981, 2023 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-37560809

RESUMEN

Accumulation of excess nutrients hampers proper liver function and is linked to nonalcoholic fatty liver disease (NAFLD) in obesity. However, the signals responsible for an impaired adaptation of hepatocytes to obesogenic dietary cues remain still largely unknown. Post-translational modification by the small ubiquitin-like modifier (SUMO) allows for a dynamic regulation of numerous processes including transcriptional reprogramming. We demonstrate that specific SUMOylation of transcription factor Prox1 represents a nutrient-sensitive determinant of hepatic fasting metabolism. Prox1 is highly SUMOylated on lysine 556 in the liver of ad libitum and refed mice, while this modification is abolished upon fasting. In the context of diet-induced obesity, Prox1 SUMOylation becomes less sensitive to fasting cues. The hepatocyte-selective knock-in of a SUMOylation-deficient Prox1 mutant into mice fed a high-fat/high-fructose diet leads to a reduction of systemic cholesterol levels, associated with the induction of liver bile acid detoxifying pathways during fasting. The generation of tools to maintain the nutrient-sensitive SUMO-switch on Prox1 may thus contribute to the development of "fasting-based" approaches for the preservation of metabolic health.

2.
FASEB J ; 35 Suppl 12021 05.
Artículo en Inglés | MEDLINE | ID: mdl-34318951

RESUMEN

Withdrawal: Valeria Lopez Salazar, Rhoda Anane Karikari, Lun Li, Rabih El-Merahbi, Maria Troullinaki, Moya Wu, Tobias Wiedemann, Alina Walth, Manuel Gil Lozano, Maria Rohm, Stephan Herzig, Anastasia Georgiadi. Adipocyte Deletion of ADAM17 Leads to Insulin Resistance in Association with Age and HFD in Mice (2021). The FASEB Journal. 35:s1. doi: 10.1096/fasebj.2021.35.S1.00447. The above abstract, published online on May 14, 2021 in Wiley Online Library (wileyonlinelibrary.com), has been withdrawn by agreement between the authors, FASEB, and Wiley Periodicals Inc. The withdrawal is due to a request made by the authors prior to publication. The Publisher apologizes that this abstract was published in error.

3.
Proc Natl Acad Sci U S A ; 114(20): 5265-5270, 2017 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-28461471

RESUMEN

Aging is accompanied by major changes in adipose tissue distribution and function. In particular, with time, thermogenic-competent beige adipocytes progressively gain a white adipocyte morphology. However, the mechanisms controlling the age-related transition of beige adipocytes to white adipocytes remain unclear. Lysine-specific demethylase 1 (Lsd1) is an epigenetic eraser enzyme positively regulating differentiation and function of adipocytes. Here we show that Lsd1 levels decrease in aging inguinal white adipose tissue concomitantly with beige fat cell decline. Accordingly, adipocyte-specific increase of Lsd1 expression is sufficient to rescue the age-related transition of beige adipocytes to white adipocytes in vivo, whereas loss of Lsd1 precipitates it. Lsd1 maintains beige adipocytes by controlling the expression of peroxisome proliferator-activated receptor α (Ppara), and treatment with a Ppara agonist is sufficient to rescue the loss of beige adipocytes caused by Lsd1 ablation. In summary, our data provide insights into the mechanism controlling the age-related beige-to-white adipocyte transition and identify Lsd1 as a regulator of beige fat cell maintenance.


Asunto(s)
Tejido Adiposo Beige/efectos de los fármacos , Tejido Adiposo Beige/metabolismo , Histona Demetilasas/metabolismo , Adipocitos/metabolismo , Adipocitos Beige , Adipocitos Blancos , Tejido Adiposo Blanco/metabolismo , Factores de Edad , Envejecimiento/metabolismo , Envejecimiento/fisiología , Animales , Diferenciación Celular , Ratones , Ratones Transgénicos , Obesidad/metabolismo , PPAR alfa/metabolismo , Termogénesis
4.
J Biol Chem ; 289(28): 19279-93, 2014 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-24876382

RESUMEN

Peroxisome proliferator-activated receptors (PPARs) play major roles in the regulation of hepatic lipid metabolism through the control of numerous genes involved in processes such as lipid uptake and fatty acid oxidation. Here we identify hypoxia-inducible lipid droplet-associated (Hilpda/Hig2) as a novel PPAR target gene and demonstrate its involvement in hepatic lipid metabolism. Microarray analysis revealed that Hilpda is one of the most highly induced genes by the PPARα agonist Wy14643 in mouse precision cut liver slices. Induction of Hilpda mRNA by Wy14643 was confirmed in mouse and human hepatocytes. Oral dosing with Wy14643 similarly induced Hilpda mRNA levels in livers of wild-type mice but not Ppara(-/-) mice. Transactivation studies and chromatin immunoprecipitation showed that Hilpda is a direct PPARα target gene via a conserved PPAR response element located 1200 base pairs upstream of the transcription start site. Hepatic overexpression of HILPDA in mice via adeno-associated virus led to a 4-fold increase in liver triglyceride storage, without any changes in key genes involved in de novo lipogenesis, ß-oxidation, or lipolysis. Moreover, intracellular lipase activity was not affected by HILPDA overexpression. Strikingly, HILPDA overexpression significantly impaired hepatic triglyceride secretion. Taken together, our data uncover HILPDA as a novel PPAR target that raises hepatic triglyceride storage via regulation of triglyceride secretion.


Asunto(s)
Lipogénesis/fisiología , Hígado/metabolismo , Proteínas de Neoplasias/metabolismo , PPAR alfa/metabolismo , Triglicéridos/metabolismo , Animales , Anticolesterolemiantes/farmacología , Línea Celular , Humanos , Lipogénesis/efectos de los fármacos , Hígado/citología , Ratones , Ratones Noqueados , Proteínas de Neoplasias/genética , PPAR alfa/genética , Pirimidinas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Elementos de Respuesta/fisiología , Triglicéridos/genética
5.
Arterioscler Thromb Vasc Biol ; 33(7): 1529-37, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23640487

RESUMEN

OBJECTIVE: Macrophage foam cells play a crucial role in several pathologies including multiple sclerosis, glomerulosclerosis, and atherosclerosis. Angiopoietin-like protein 4 (Angptl4) was previously shown to inhibit chyle-induced foam cell formation in mesenteric lymph nodes. Here we characterized the regulation of Angptl4 expression in macrophages and examined the impact of Angptl4 on atherosclerosis development. APPROACH AND RESULTS: Macrophage activation elicited by pathogen-recognition receptor agonists decreased Angptl4 expression, whereas lipid loading by intralipid and oxidized low-density lipoprotein increased Angptl4 expression. Consistent with an antilipotoxic role of Angptl4, recombinant Angptl4 significantly decreased uptake of oxidized low-density lipoprotein by macrophages, via lipolysis-dependent and -independent mechanisms. Angptl4 protein was detectable in human atherosclerotic lesions and localized to macrophages. Transgenic overexpression of Angptl4 in atherosclerosis-prone apolipoprotein E*3-Leiden mice did not significantly alter plasma cholesterol and triglyceride levels. Nevertheless, Angptl4 overexpression reduced lesion area by 34% (P<0.05). In addition, Angptl4 overexpression decreased macrophage content (-41%; P<0.05) and numbers of monocytes adhering to the endothelium wall (-37%; P<0.01). Finally, plasma Angptl4 was independently and negatively associated with carotid artery sclerosis measured by 3-T MRI in subjects with metabolic syndrome and low-grade systemic inflammation. CONCLUSIONS: Angptl4 suppresses foam cell formation to reduce atherosclerosis development. Stimulation of Angptl4 in macrophages by oxidized low-density lipoprotein may protect against lipid overload.


Asunto(s)
Angiopoyetinas/sangre , Angiopoyetinas/metabolismo , Enfermedades de la Aorta/prevención & control , Aterosclerosis/prevención & control , Estenosis Carotídea/prevención & control , Macrófagos/metabolismo , Proteína 4 Similar a la Angiopoyetina , Angiopoyetinas/genética , Animales , Aorta/metabolismo , Aorta/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Apolipoproteína E3/genética , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Arterias Carótidas/patología , Estenosis Carotídea/sangre , Estenosis Carotídea/patología , Línea Celular , Quimiotaxis , Colesterol/sangre , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Femenino , Células Espumosas/metabolismo , Humanos , Ligandos , Lipoproteínas LDL/metabolismo , Activación de Macrófagos , Macrófagos/efectos de los fármacos , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Factores de Tiempo , Receptor Toll-Like 3/agonistas , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 4/agonistas , Receptor Toll-Like 4/metabolismo , Triglicéridos/sangre , Regulación hacia Arriba
6.
Nat Commun ; 15(1): 1391, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38360943

RESUMEN

In obesity, sustained adipose tissue (AT) inflammation constitutes a cellular memory that limits the effectiveness of weight loss interventions. Yet, the impact of fasting regimens on the regulation of AT immune infiltration is still elusive. Here we show that intermittent fasting (IF) exacerbates the lipid-associated macrophage (LAM) inflammatory phenotype of visceral AT in obese mice. Importantly, this increase in LAM abundance is strongly p53 dependent and partly mediated by p53-driven adipocyte apoptosis. Adipocyte-specific deletion of p53 prevents LAM accumulation during IF, increases the catabolic state of adipocytes, and enhances systemic metabolic flexibility and insulin sensitivity. Finally, in cohorts of obese/diabetic patients, we describe a p53 polymorphism that links to efficacy of a fasting-mimicking diet and that the expression of p53 and TREM2 in AT negatively correlates with maintaining weight loss after bariatric surgery. Overall, our results demonstrate that p53 signalling in adipocytes dictates LAM accumulation in AT under IF and modulates fasting effectiveness in mice and humans.


Asunto(s)
Resistencia a la Insulina , Ayuno Intermitente , Proteína p53 Supresora de Tumor , Animales , Humanos , Ratones , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Inflamación/metabolismo , Resistencia a la Insulina/genética , Obesidad/genética , Obesidad/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Pérdida de Peso
7.
J Biol Chem ; 287(3): 1847-60, 2012 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-22117073

RESUMEN

Obesity and insulin resistance are associated with chronic, low grade inflammation. Moreover, regulation of energy metabolism and immunity are highly integrated. We hypothesized that energy-sensitive coactivator peroxisome proliferator-activated receptor γ coactivator 1-α (PGC-1α) and AMP-activated protein kinase (AMPK) may modulate inflammatory gene expression in liver. Microarray analysis revealed that PGC-1α up-regulated expression of several cytokines and cytokine receptors, including interleukin 15 receptor α (IL15Rα) and, even more importantly, anti-inflammatory interleukin 1 receptor antagonist (IL1Rn). Overexpression of PGC-1α and induction of PGC-1α by fasting, physical exercise, glucagon, or cAMP was associated with increased IL1Rn mRNA and protein expression in hepatocytes. Knockdown of PGC-1α by siRNA down-regulated cAMP-induced expression of IL1Rn in mouse hepatocytes. Furthermore, knockdown of peroxisome proliferator-activated receptor α (PPARα) attenuated IL1Rn induction by PGC-1α. Overexpression of PGC-1α, at least partially through IL1Rn, suppressed interleukin 1ß-induced expression of acute phase proteins, C-reactive protein, and haptoglobin. Fasting and exercise also induced IL15Rα expression, whereas glucagon and cAMP resulted in reduction in IL15Rα mRNA levels. Finally, AMPK activator metformin and adenoviral overexpression of AMPK up-regulated IL1Rn and down-regulated IL15Rα in primary hepatocytes. We conclude that PGC-1α and AMPK alter inflammatory gene expression in liver and thus integrate energy homeostasis and inflammation. Induction of IL1Rn by PGC-1α and AMPK may be involved in the beneficial effects of exercise and caloric restriction and putative anti-inflammatory effects of metformin.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Metabolismo Energético , Mediadores de Inflamación/metabolismo , Proteína Antagonista del Receptor de Interleucina 1/biosíntesis , Hígado/metabolismo , Proteínas de Unión al ARN/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Animales , Proteína C-Reactiva/biosíntesis , Proteína C-Reactiva/genética , Restricción Calórica , Células Cultivadas , Activadores de Enzimas/farmacología , Ayuno/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Técnicas de Silenciamiento del Gen , Haptoglobinas/biosíntesis , Haptoglobinas/genética , Hepatocitos/metabolismo , Hepatocitos/patología , Hipoglucemiantes/farmacología , Resistencia a la Insulina/genética , Proteína Antagonista del Receptor de Interleucina 1/genética , Hígado/patología , Masculino , Metformina/farmacología , Ratones , Ratones Endogámicos DBA , Obesidad/genética , Obesidad/metabolismo , Obesidad/terapia , PPAR alfa/genética , PPAR alfa/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Condicionamiento Físico Animal , Proteínas de Unión al ARN/genética , Ratas , Receptores de Interleucina-15/biosíntesis , Receptores de Interleucina-15/genética , Transactivadores/genética , Factores de Transcripción/genética
8.
IUBMB Life ; 65(10): 845-50, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24078392

RESUMEN

Exercise is known to prevent and treat metabolic diseases such as diabetes. However, the underlying mechanisms are not fully understood, and there is currently much focus on detailing such pathways. Traditionally, much emphasis has been placed on skeletal muscle; however, recently, nonmuscle organs such as adipose tissue have been highlighted in mediating protective actions after training. Moreover, novel paracrine- and endocrine-signaling molecules have been shown to trigger important responses in nonmuscle organs after exercise. This is exciting because, when administered exogenously, such signals have obvious therapeutic potential. In this review, the authors have described some general and historical aspects of training and disease protection. The authors have also highlighted some of the current knowledge on how exercise impacts nonmuscle organs.


Asunto(s)
Ejercicio Físico , Músculo Esquelético/metabolismo , Tejido Adiposo/metabolismo , Encéfalo/metabolismo , Encéfalo/fisiología , Humanos , Hígado/metabolismo , Hígado/fisiología , Músculo Esquelético/fisiología , Miocardio/metabolismo
9.
Physiol Genomics ; 44(6): 352-61, 2012 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-22274564

RESUMEN

Fatty acids comprise the primary energy source for the heart and are mainly taken up via hydrolysis of circulating triglyceride-rich lipoproteins. While most of the fatty acids entering the cardiomyocyte are oxidized, a small portion is involved in altering gene transcription to modulate cardiometabolic functions. So far, no in vivo model has been developed enabling study of the transcriptional effects of specific fatty acids in the intact heart. In the present study, mice were given a single oral dose of synthetic triglycerides composed of one single fatty acid. Hearts were collected 6 h thereafter and used for whole genome gene expression profiling. Experiments were conducted in wild-type and peroxisome proliferator-activated receptor (PPAR)α-/- mice to allow exploration of the specific contribution of PPARα. It was found that: 1) C18:3 had the most pronounced effect on cardiac gene expression. 2) The largest similarity in gene regulation was observed between C18:2 and C18:3. Large similarity was also observed between PPARα agonist Wy14643 and C22:6. 3) Many genes were regulated by one particular treatment only. Genes regulated by one particular treatment showed large functional divergence. 4) The majority of genes responding to fatty acid treatment were regulated in a PPARα-dependent manner, emphasizing the importance of PPARα in mediating transcriptional regulation by fatty acids in the heart. 5) Several genes were robustly regulated by all or many of the fatty acids studied, mostly representing well-described targets of PPARs (e.g., Acot1, Angptl4, Ucp3) but also including Zbtb16/PLZF, a transcription factor crucial for natural killer T cell function. 6) Deletion and activation of PPARα had a major effect on expression of numerous genes involved in metabolism and immunity. Our analysis demonstrates the marked impact of dietary fatty acids on gene regulation in the heart via PPARα.


Asunto(s)
Ácidos Grasos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Miocardio/metabolismo , Administración Oral , Animales , Ácidos Docosahexaenoicos/farmacología , Ácidos Grasos/administración & dosificación , Perfilación de la Expresión Génica , Ácido Linoleico/farmacología , Ratones , Ratones Noqueados , Análisis por Micromatrices , Ácido Oléico/farmacología , PPAR alfa/genética , Pirimidinas/farmacología , Ácido alfa-Linolénico/farmacología
10.
Circ Res ; 106(11): 1712-21, 2010 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-20378851

RESUMEN

RATIONALE: Although dietary fatty acids are a major fuel for the heart, little is known about the direct effects of dietary fatty acids on gene regulation in the intact heart. OBJECTIVE: To study the effect of dietary fatty acids on cardiac gene expression and explore the functional consequences. METHODS AND RESULTS: Oral administration of synthetic triglycerides composed of one single fatty acid altered cardiac expression of numerous genes, many of which are involved in the oxidative stress response. The gene most significantly and consistently upregulated by dietary fatty acids encoded Angiopoietin-like protein (Angptl)4, a circulating inhibitor of lipoprotein lipase expressed by cardiomyocytes. Induction of Angptl4 by the fatty acid linolenic acid was specifically abolished in peroxisome proliferator-activated receptor (PPAR)beta/delta(-/-) and not PPARalpha(-/-) mice and was blunted on siRNA-mediated PPARbeta/delta knockdown in cultured cardiomyocytes. Consistent with these data, linolenic acid stimulated binding of PPARbeta/delta but not PPARalpha to the Angptl4 gene. Upregulation of Angptl4 resulted in decreased cardiac uptake of plasma triglyceride-derived fatty acids and decreased fatty acid-induced oxidative stress and lipid peroxidation. In contrast, Angptl4 deletion led to enhanced oxidative stress in the heart, both after an acute oral fat load and after prolonged high fat feeding. CONCLUSIONS: Stimulation of cardiac Angptl4 gene expression by dietary fatty acids and via PPARbeta/delta is part of a feedback mechanism aimed at protecting the heart against lipid overload and consequently fatty acid-induced oxidative stress.


Asunto(s)
Angiopoyetinas/metabolismo , Cardiomiopatías/prevención & control , Grasas de la Dieta/metabolismo , Ácidos Grasos Insaturados/metabolismo , Miocardio/metabolismo , Estrés Oxidativo , PPAR delta/metabolismo , PPAR-beta/metabolismo , Proteína 4 Similar a la Angiopoyetina , Angiopoyetinas/deficiencia , Angiopoyetinas/genética , Animales , Animales Recién Nacidos , Cardiomiopatías/inducido químicamente , Cardiomiopatías/genética , Cardiomiopatías/metabolismo , Células Cultivadas , Citoprotección , Grasas de la Dieta/administración & dosificación , Grasas de la Dieta/sangre , Grasas de la Dieta/toxicidad , Ácidos Grasos Insaturados/administración & dosificación , Ácidos Grasos Insaturados/sangre , Ácidos Grasos Insaturados/toxicidad , Retroalimentación Fisiológica , Ácido Linoleico/metabolismo , Peroxidación de Lípido , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ácido Oléico/metabolismo , Estrés Oxidativo/genética , PPAR delta/deficiencia , PPAR delta/genética , PPAR-beta/deficiencia , PPAR-beta/genética , Interferencia de ARN , Factores de Tiempo , Regulación hacia Arriba , Ácido alfa-Linolénico/metabolismo
11.
Mol Metab ; 56: 101412, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34890852

RESUMEN

OBJECTIVE: Multiple genome-wide association studies (GWAS) have identified SNPs in the 8q24 locus near TRIB1 that are significantly associated with plasma lipids and other markers of cardiometabolic health, and prior studies have revealed the roles of hepatic and myeloid Trib1 in plasma lipid regulation and atherosclerosis. The same 8q24 SNPs are additionally associated with plasma adiponectin levels in humans, implicating TRIB1 in adipocyte biology. Here, we hypothesize that TRIB1 in adipose tissue regulates plasma adiponectin, lipids, and metabolic health. METHODS: We investigate the metabolic phenotype of adipocyte-specific Trib1 knockout mice (Trib1_ASKO) fed on chow and high-fat diet (HFD). Through secretomics of adipose tissue explants and RNA-seq of adipocytes and livers from these mice, we further investigate the mechanism of TRIB1 in adipose tissue. RESULTS: Trib1_ASKO mice have an improved metabolic phenotype with increased plasma adiponectin levels, improved glucose tolerance, and decreased plasma lipids. Trib1_ASKO adipocytes have increased adiponectin production and secretion independent of the known TRIB1 function of regulating proteasomal degradation. RNA-seq analysis of adipocytes and livers from Trib1_ASKO mice indicates that alterations in adipocyte function underlie the observed plasma lipid changes. Adipose tissue explant secretomics further reveals that Trib1_ASKO adipose tissue has decreased ANGPTL4 production, and we demonstrate an accompanying increase in the lipoprotein lipase (LPL) activity that likely underlies the triglyceride phenotype. CONCLUSIONS: This study shows that adipocyte Trib1 regulates multiple aspects of metabolic health, confirming previously observed genetic associations in humans and shedding light on the further mechanisms by which TRIB1 regulates plasma lipids and metabolic health.


Asunto(s)
Adiponectina , Estudio de Asociación del Genoma Completo , Adipocitos/metabolismo , Adiponectina/genética , Adiponectina/metabolismo , Animales , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Triglicéridos/metabolismo
12.
Life Sci Alliance ; 4(4)2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33536239

RESUMEN

Members of the lipocalin protein family serve as biomarkers for kidney disease and acute phase inflammatory reactions, and are under preclinical development for the diagnosis and therapy of allergies. However, none of the lipocalin family members has made the step into clinical development, mostly due to their complex biological activity and the lack of in-depth mechanistic knowledge. Here, we show that the hepatokine lipocalin 13 (LCN13) triggers glucose-dependent insulin secretion and cell proliferation of primary mouse islets. However, inhibition of endogenous LCN13 expression in lean mice did not alter glucose and lipid homeostasis. Enhanced hepatic secretion of LCN13 in either diet-induced or genetic obesity led to no discernible impact on systemic glucose and lipid metabolism, neither in preventive nor therapeutic setting. Of note, loss or forced LCN13 hepatic secretion did not trigger any compensatory regulation of related lipocalin family members. Together, these data are in stark contrast to the suggested gluco-regulatory and therapeutic role of LCN13 in obesity, and imply complex regulatory steps in LCN13 biology at the organismic level mitigating its principal insulinotropic effects.


Asunto(s)
Metabolismo Energético , Secreción de Insulina , Lipocalinas/genética , Lipocalinas/metabolismo , Animales , Biomarcadores , Técnica del Anticuerpo Fluorescente , Expresión Génica , Técnicas de Silenciamiento del Gen , Glucosa/metabolismo , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Metabolismo de los Lípidos , Lipocalinas/sangre , Hígado/metabolismo , Masculino , Ratones , Obesidad/etiología , Obesidad/metabolismo
13.
Nat Commun ; 12(1): 2999, 2021 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-34016966

RESUMEN

The proper functional interaction between different tissues represents a key component in systemic metabolic control. Indeed, disruption of endocrine inter-tissue communication is a hallmark of severe metabolic dysfunction in obesity and diabetes. Here, we show that the FNDC4-GPR116, liver-white adipose tissue endocrine axis controls glucose homeostasis. We found that the liver primarily controlled the circulating levels of soluble FNDC4 (sFNDC4) and lowering of the hepatokine FNDC4 led to prediabetes in mice. Further, we identified the orphan adhesion GPCR GPR116 as a receptor of sFNDC4 in the white adipose tissue. Upon direct and high affinity binding of sFNDC4 to GPR116, sFNDC4 promoted insulin signaling and insulin-mediated glucose uptake in white adipocytes. Indeed, supplementation with FcsFNDC4 in prediabetic mice improved glucose tolerance and inflammatory markers in a white-adipocyte selective and GPR116-dependent manner. Of note, the sFNDC4-GPR116, liver-adipose tissue axis was dampened in (pre) diabetic human patients. Thus our findings will now allow for harnessing this endocrine circuit for alternative therapeutic strategies in obesity-related pre-diabetes.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Proteínas de la Membrana/metabolismo , Estado Prediabético/metabolismo , Proteínas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Células 3T3-L1 , Adipocitos/metabolismo , Tejido Adiposo Blanco/citología , Adolescente , Adulto , Anciano , Animales , Células CHO , Estudios de Cohortes , Cricetulus , Estudios Transversales , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/prevención & control , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Femenino , Técnicas de Silenciamiento del Gen , Glucosa/metabolismo , Células HEK293 , Células Hep G2 , Humanos , Insulina/metabolismo , Resistencia a la Insulina , Islotes Pancreáticos/metabolismo , Hígado/metabolismo , Masculino , Proteínas de la Membrana/administración & dosificación , Proteínas de la Membrana/sangre , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Persona de Mediana Edad , Células 3T3 NIH , Estado Prediabético/sangre , Estado Prediabético/tratamiento farmacológico , Estado Prediabético/etiología , Cultivo Primario de Células , Proteínas/análisis , Receptores Acoplados a Proteínas G/sangre , Receptores Acoplados a Proteínas G/genética , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/aislamiento & purificación , Adulto Joven
14.
Cell Rep ; 30(6): 1811-1822.e6, 2020 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-32049012

RESUMEN

Obesity leads to a state of chronic, low-grade inflammation that features the accumulation of lipid-laden macrophages in adipose tissue. Here, we determined the role of macrophage lipid-droplet accumulation in the development of obesity-induced adipose-tissue inflammation, using mice with myeloid-specific deficiency of the lipid-inducible HILPDA protein. HILPDA deficiency markedly reduced intracellular lipid levels and accumulation of fluorescently labeled fatty acids. Decreased lipid storage in HILPDA-deficient macrophages can be rescued by inhibition of adipose triglyceride lipase (ATGL) and is associated with increased oxidative metabolism. In diet-induced obese mice, HILPDA deficiency does not alter inflammatory and metabolic parameters, despite markedly reducing lipid accumulation in macrophages. Overall, we find that HILPDA is a lipid-inducible, physiological inhibitor of ATGL-mediated lipolysis in macrophages and uncouples lipid storage in adipose tissue macrophages from inflammation and metabolic dysregulation. Our data question the contribution of lipid droplet accumulation in adipose tissue macrophages in obesity-induced inflammation and metabolic dysregulation.


Asunto(s)
Tejido Adiposo/fisiopatología , Ácidos Grasos/metabolismo , Inflamación/metabolismo , Gotas Lipídicas/metabolismo , Metabolismo de los Lípidos/fisiología , Macrófagos/metabolismo , Proteínas de Neoplasias/metabolismo , Animales , Humanos , Ratones
15.
Mol Aspects Med ; 68: 90-100, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31283940

RESUMEN

For the past decade, brown adipose tissue (BAT) has been extensively studied as a potential therapy for obesity and metabolic diseases due to its thermogenic and glucose-consuming properties. It is now clear that the function of BAT goes beyond heat production, as it also plays an important endocrine role by secreting the so-called batokines to communicate with other metabolic tissues and regulate systemic energy homeostasis. However, despite numerous studies showing the benefits of BAT in rodents, it is still not clear whether recruitment of BAT can be utilized to treat human patients. Here, we review the advances on understanding the role of BAT in metabolism and its benefits on glucose and lipid homeostasis in both humans and rodents. Moreover, we discuss the latest methodological approaches to assess the contribution of BAT to human metabolism as well as the possibility to target BAT, pharmacologically or by lifestyle adaptations, to treat metabolic disorders.


Asunto(s)
Tejido Adiposo Beige/metabolismo , Tejido Adiposo Pardo/metabolismo , Hiperglucemia/patología , Animales , Metabolismo Energético , Glucosa/metabolismo , Humanos , Transducción de Señal
16.
Horm Mol Biol Clin Investig ; 31(2)2017 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-28850545

RESUMEN

The dramatically increasing world-wide prevalence of obesity is recognized as a risk factor for the development of various diseases. The growing research on the role of adipose tissue in controlling energy homeostasis and insulin sensitivity has revealed that the promotion of brown adipose tissue (BAT) activity and the browning of white adipose tissue (WAT) leads to multiple health benefits and prevents obesity and type 2 diabetes (T2D). Inducible thermogenic adipocytes do exist in adult humans and are linked with increased energy combustion and lower body fat mass. Thus brown adipocytes are currently placed at the center of attention for novel therapeutic strategies against metabolic diseases such as obesity and diabetes. Besides the classical, norepinephrine-mediated sympathetic recruitment and activation of thermogenic adipocytes, a number of novel circulating factors have been recently identified to have a positive or negative impact on thermogenic adipocyte formation and activity. In this review their mechanism of action and the plausible therapeutic applications will be summarized and discussed.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Comunicación Autocrina , Diabetes Mellitus Tipo 2/metabolismo , Sistema Endocrino/fisiología , Obesidad/metabolismo , Comunicación Paracrina , Adipoquinas/metabolismo , Tejido Adiposo Pardo/patología , Animales , Diabetes Mellitus Tipo 2/patología , Diabetes Mellitus Tipo 2/terapia , Metabolismo Energético , Hormonas/metabolismo , Humanos , Obesidad/patología , Obesidad/terapia , Termogénesis
17.
Nat Commun ; 7: 11314, 2016 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-27066907

RESUMEN

FNDC4 is a secreted factor sharing high homology with the exercise-associated myokine irisin (FNDC5). Here we report that Fndc4 is robustly upregulated in several mouse models of inflammation as well as in human inflammatory conditions. Specifically, FNDC4 levels are increased locally at inflamed sites of the intestine of inflammatory bowel disease patients. Interestingly, administration of recombinant FNDC4 in the mouse model of induced colitis markedly reduces disease severity compared with mice injected with a control protein. Conversely, mice lacking Fndc4 develop more severe colitis. Analysis of binding of FNDC4 to different immune cell types reveals strong and specific binding to macrophages and monocytes. FNDC4 treatment of bone marrow-derived macrophages in vitro results in reduced phagocytosis, increased cell survival and reduced proinflammatory chemokine expression. Hence, treatment with FNDC4 results in a state of dampened macrophage activity, while enhancing their survival. Thus, we have characterized FNDC4 as a factor with direct therapeutic potential in inflammatory bowel disease and possibly other inflammatory diseases.


Asunto(s)
Antiinflamatorios/metabolismo , Colitis/metabolismo , Macrófagos/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas/metabolismo , Secuencia de Aminoácidos , Animales , Células Cultivadas , Colitis/genética , Colitis/patología , Sulfato de Dextran , Progresión de la Enfermedad , Regulación de la Expresión Génica , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Masculino , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Fagocitosis/efectos de los fármacos , Proteínas/química , Proteínas/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología , Regulación hacia Arriba/efectos de los fármacos
18.
BMJ Open Diabetes Res Care ; 2(1): e000034, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25512873

RESUMEN

BACKGROUND: Angiopoietin-like protein 4 (ANGPTL4) inhibits lipoprotein lipase and associates with dyslipidemia. The expression of ANGPTL4 is regulated by free fatty acids (FFA) that activate lipid-sensing peroxisome proliferator-activated receptors (PPARs), but FFA can also activate pattern recognition receptors including Toll-like receptor 4 (TLR4) in macrophages. OBJECTIVE: To assess whether systemic low-grade inflammation is a determinant for plasma ANGPTL4 levels in patients with the metabolic syndrome (MetS) and type 2 diabetes mellitus (T2DM). DESIGN: We studied 335 male participants: healthy controls (Controls), patients with the MetS without inflammation (MetS-I) and with low-grade inflammation (MetS+I), and patients with T2DM. All patients without diabetes included in the present study were initially matched for waist circumference. In plasma, ANGPTL4, C reactive protein (CRP) and metabolic parameters were determined. Underlying mechanisms were examined using human macrophages in vitro. RESULTS: As compared with Controls, plasma ANGPTL4 levels were increased in patients with MetS-I, MetS+I, and T2DM. Furthermore, ANGPTL4 was increased in T2DM compared with MetS-I. In fact, plasma CRP correlated positively with plasma ANGPTL4. In vitro studies showed that TLR 3/4 activation largely increased the expression and release of ANGPTL4 by macrophages. CONCLUSIONS: Plasma ANGPTL4 levels in humans are predicted by CRP, a marker of inflammation, and ANGPTL4 expression by macrophages is increased by inflammatory stimuli.

19.
Cell Metab ; 19(1): 96-108, 2014 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-24411942

RESUMEN

The transcriptional coactivator peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC-1α) regulates metabolic genes in skeletal muscle and contributes to the response of muscle to exercise. Muscle PGC-1α transgenic expression and exercise both increase the expression of thermogenic genes within white adipose. How the PGC-1α-mediated response to exercise in muscle conveys signals to other tissues remains incompletely defined. We employed a metabolomic approach to examine metabolites secreted from myocytes with forced expression of PGC-1α, and identified ß-aminoisobutyric acid (BAIBA) as a small molecule myokine. BAIBA increases the expression of brown adipocyte-specific genes in white adipocytes and ß-oxidation in hepatocytes both in vitro and in vivo through a PPARα-mediated mechanism, induces a brown adipose-like phenotype in human pluripotent stem cells, and improves glucose homeostasis in mice. In humans, plasma BAIBA concentrations are increased with exercise and inversely associated with metabolic risk factors. BAIBA may thus contribute to exercise-induced protection from metabolic diseases.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Ácidos Aminoisobutíricos/farmacología , Enfermedades Cardiovasculares/metabolismo , Hígado/metabolismo , Enfermedades Metabólicas/metabolismo , Adipocitos Marrones/efectos de los fármacos , Adipocitos Marrones/metabolismo , Adipocitos Marrones/patología , Adipocitos Blancos/efectos de los fármacos , Adipocitos Blancos/metabolismo , Adipocitos Blancos/patología , Tejido Adiposo Pardo/citología , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Blanco/citología , Tejido Adiposo Blanco/efectos de los fármacos , Ácidos Aminoisobutíricos/sangre , Animales , Enfermedades Cardiovasculares/patología , Diferenciación Celular/efectos de los fármacos , Ejercicio Físico , Regulación de la Expresión Génica/efectos de los fármacos , Prueba de Tolerancia a la Glucosa , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Hígado/efectos de los fármacos , Enfermedades Metabólicas/patología , Ratones , Especificidad de Órganos/efectos de los fármacos , Especificidad de Órganos/genética , Oxidación-Reducción/efectos de los fármacos , PPAR alfa/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Fenotipo , Condicionamiento Físico Animal , Factores de Riesgo , Factores de Transcripción/metabolismo , Transcripción Genética/efectos de los fármacos , Aumento de Peso/efectos de los fármacos
20.
Adv Nutr ; 3(2): 127-34, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22516720

RESUMEN

Consumption of specific dietary fatty acids has been shown to influence risk and progression of several chronic diseases, such as cardiovascular disease, obesity, cancer, and arthritis. In recent years, insights into the mechanisms underlying the biological effects of fatty acids have improved considerably and have provided the foundation for the emerging concept of fatty acid sensing, which can be interpreted as the property of fatty acids to influence biological processes by serving as signaling molecules. An important mechanism of fatty acid sensing is via stimulation or inhibition of DNA transcription. Here, we focus on fatty acid sensing via regulation of gene transcription and address the role of peroxisome proliferator-activated receptors, sterol regulatory element binding protein 1, Toll-like receptor 4, G protein-coupled receptors, and other putative mediators.


Asunto(s)
Ácidos Grasos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Factor Nuclear 4 del Hepatocito/genética , Humanos , Factor 2 Relacionado con NF-E2/genética , Receptores Activados del Proliferador del Peroxisoma/genética , Receptores Acoplados a Proteínas G/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Receptor Toll-Like 4/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA