Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Hepatology ; 77(4): 1287-1302, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35735979

RESUMEN

BACKGROUND: NAFLD affects nearly 25% of the global population. Cardiovascular disease (CVD) is the most common cause of death among patients with NAFLD, in line with highly prevalent dyslipidemia in this population. Increased plasma triglyceride (TG)-rich lipoprotein (TRL) concentrations, an important risk factor for CVD, are closely linked with hepatic TG content. Therefore, it is of great interest to identify regulatory mechanisms of hepatic TRL production and remnant uptake in the setting of hepatic steatosis. APPROACH AND RESULTS: To identify liver-regulated pathways linking intrahepatic and plasma TG metabolism, we performed transcriptomic analysis of liver biopsies from two independent cohorts of obese patients. Hepatic encoding apolipoprotein F ( APOF ) expression showed the fourth-strongest negatively correlation with hepatic steatosis and the strongest negative correlation with plasma TG levels. The effects of adenoviral-mediated human ApoF (hApoF) overexpression on plasma and hepatic TG were assessed in C57BL6/J mice. Surprisingly, hApoF overexpression increased both hepatic very low density lipoprotein (VLDL)-TG secretion and hepatic lipoprotein remnant clearance, associated a ~25% reduction in plasma TG levels. Conversely, reducing endogenous ApoF expression reduced VLDL secretion in vivo , and reduced hepatocyte VLDL uptake by ~15% in vitro . Transcriptomic analysis of APOF -overexpressing mouse livers revealed a gene signature related to enhanced ApoB-lipoprotein clearance, including increased expression of Ldlr and Lrp1 , among others. CONCLUSION: These data reveal a previously undescribed role for ApoF in the control of plasma and hepatic lipoprotein metabolism by favoring VLDL-TG secretion and hepatic lipoprotein remnant particle clearance.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Ratones , Animales , Humanos , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Lipoproteínas/metabolismo , Apolipoproteínas/metabolismo , Apolipoproteínas/farmacología , Triglicéridos/metabolismo , Hígado/metabolismo , Lipoproteínas VLDL/metabolismo
2.
J Hepatol ; 79(4): 898-909, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37230231

RESUMEN

BACKGROUND & AIMS: Roux-en-Y gastric bypass (RYGB), the most effective surgical procedure for weight loss, decreases obesity and ameliorates comorbidities, such as non-alcoholic fatty liver (NAFLD) and cardiovascular (CVD) diseases. Cholesterol is a major CVD risk factor and modulator of NAFLD development, and the liver tightly controls its metabolism. How RYGB surgery modulates systemic and hepatic cholesterol metabolism is still unclear. METHODS: We studied the hepatic transcriptome of 26 patients with obesity but not diabetes before and 1 year after undergoing RYGB. In parallel, we measured quantitative changes in plasma cholesterol metabolites and bile acids (BAs). RESULTS: RYGB surgery improved systemic cholesterol metabolism and increased plasma total and primary BA levels. Transcriptomic analysis revealed specific alterations in the liver after RYGB, with the downregulation of a module of genes implicated in inflammation and the upregulation of three modules, one associated with BA metabolism. A dedicated analysis of hepatic genes related to cholesterol homeostasis pointed towards increased biliary cholesterol elimination after RYGB, associated with enhancement of the alternate, but not the classical, BA synthesis pathway. In parallel, alterations in the expression of genes involved in cholesterol uptake and intracellular trafficking indicate improved hepatic free cholesterol handling. Finally, RYGB decreased plasma markers of cholesterol synthesis, which correlated with an improvement in liver disease status after surgery. CONCLUSIONS: Our results identify specific regulatory effects of RYGB on inflammation and cholesterol metabolism. RYGB alters the hepatic transcriptome signature, likely improving liver cholesterol homeostasis. These gene regulatory effects are reflected by systemic post-surgery changes of cholesterol-related metabolites, corroborating the beneficial effects of RYGB on both hepatic and systemic cholesterol homeostasis. IMPACT AND IMPLICATIONS: Roux-en-Y gastric bypass (RYGB) is a widely used bariatric surgery procedure with proven efficacy in body weight management, combatting cardiovascular disease (CVD) and non-alcoholic fatty liver disease (NAFLD). RYGB exerts many beneficial metabolic effects, by lowering plasma cholesterol and improving atherogenic dyslipidemia. Using a cohort of patients undergoing RYGB, studied before and 1 year after surgery, we analyzed how RYGB modulates hepatic and systemic cholesterol and bile acid metabolism. The results of our study provide important insights on the regulation of cholesterol homeostasis after RYGB and open avenues that could guide future monitoring and treatment strategies targeting CVD and NAFLD in obesity.


Asunto(s)
Derivación Gástrica , Enfermedad del Hígado Graso no Alcohólico , Obesidad Mórbida , Humanos , Derivación Gástrica/métodos , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/cirugía , Transcriptoma , Obesidad/complicaciones , Colesterol , Homeostasis , Inflamación/complicaciones , Obesidad Mórbida/complicaciones
3.
Mol Syst Biol ; 16(5): e9156, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32407006

RESUMEN

Liver injury triggers adaptive remodeling of the hepatic transcriptome for repair/regeneration. We demonstrate that this involves particularly profound transcriptomic alterations where acute induction of genes involved in handling of endoplasmic reticulum stress (ERS) is accompanied by partial hepatic dedifferentiation. Importantly, widespread hepatic gene downregulation could not simply be ascribed to cofactor squelching secondary to ERS gene induction, but rather involves a combination of active repressive mechanisms. ERS acts through inhibition of the liver-identity (LIVER-ID) transcription factor (TF) network, initiated by rapid LIVER-ID TF protein loss. In addition, induction of the transcriptional repressor NFIL3 further contributes to LIVER-ID gene repression. Alteration to the liver TF repertoire translates into compromised activity of regulatory regions characterized by the densest co-recruitment of LIVER-ID TFs and decommissioning of BRD4 super-enhancers driving hepatic identity. While transient repression of the hepatic molecular identity is an intrinsic part of liver repair, sustained disequilibrium between the ERS and LIVER-ID transcriptional programs is linked to liver dysfunction as shown using mouse models of acute liver injury and livers from deceased human septic patients.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Estrés del Retículo Endoplásmico/genética , Regulación de la Expresión Génica/genética , Hepatopatías/metabolismo , Transcriptoma/genética , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Células Cultivadas , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Secuenciación de Inmunoprecipitación de Cromatina , Regulación hacia Abajo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Hepatopatías/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Tapsigargina/toxicidad , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Regulación hacia Arriba
4.
J Hepatol ; 70(5): 963-973, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30677458

RESUMEN

BACKGROUND & AIMS: Although the role of inflammation to combat infection is known, the contribution of metabolic changes in response to sepsis is poorly understood. Sepsis induces the release of lipid mediators, many of which activate nuclear receptors such as the peroxisome proliferator-activated receptor (PPAR)α, which controls both lipid metabolism and inflammation. We aimed to elucidate the previously unknown role of hepatic PPARα in the response to sepsis. METHODS: Sepsis was induced by intraperitoneal injection of Escherichia coli in different models of cell-specific Ppara-deficiency and their controls. The systemic and hepatic metabolic response was analyzed using biochemical, transcriptomic and functional assays. PPARα expression was analyzed in livers from elective surgery and critically ill patients and correlated with hepatic gene expression and blood parameters. RESULTS: Both whole body and non-hematopoietic Ppara-deficiency in mice decreased survival upon bacterial infection. Livers of septic Ppara-deficient mice displayed an impaired metabolic shift from glucose to lipid utilization resulting in more severe hypoglycemia, impaired induction of hyperketonemia and increased steatosis due to lower expression of genes involved in fatty acid catabolism and ketogenesis. Hepatocyte-specific deletion of PPARα impaired the metabolic response to sepsis and was sufficient to decrease survival upon bacterial infection. Hepatic PPARA expression was lower in critically ill patients and correlated positively with expression of lipid metabolism genes, but not with systemic inflammatory markers. CONCLUSION: During sepsis, Ppara-deficiency in hepatocytes is deleterious as it impairs the adaptive metabolic shift from glucose to FA utilization. Metabolic control by PPARα in hepatocytes plays a key role in the host defense against infection. LAY SUMMARY: As the main cause of death in critically ill patients, sepsis remains a major health issue lacking efficacious therapies. While current clinical literature suggests an important role for inflammation, metabolic aspects of sepsis have mostly been overlooked. Here, we show that mice with an impaired metabolic response, due to deficiency of the nuclear receptor PPARα in the liver, exhibit enhanced mortality upon bacterial infection despite a similar inflammatory response, suggesting that metabolic interventions may be a viable strategy for improving sepsis outcomes.


Asunto(s)
Adaptación Fisiológica , Hígado/metabolismo , PPAR alfa/fisiología , Sepsis/metabolismo , Animales , Infecciones Bacterianas/metabolismo , Ácidos Grasos/metabolismo , Glucosa/metabolismo , Humanos , Inflamación/etiología , Ratones , Ratones Endogámicos C57BL
5.
Hepatology ; 60(5): 1593-606, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24995693

RESUMEN

UNLABELLED: Nonalcoholic fatty liver disease (NAFLD) is increasingly prevalent and strongly associated with central obesity, dyslipidemia, and insulin resistance. According to the multiple-hit model of NAFLD pathogenesis, lipid accumulation drives nonalcoholic steatohepatitis (NASH) initiation by triggering oxidative stress, lipotoxicity, and subsequent activation of hepatic inflammatory responses that may progress, in predisposed individuals, to fibrosis and cirrhosis. While there is an unmet therapeutical need for NASH and fibrosis, recent preclinical studies showed that peroxisome proliferator-activated receptor (PPAR)-α agonism can efficiently oppose these symptoms. To dissect the relative contribution of antisteatotic versus anti-inflammatory PPAR-α activities in counteracting dietary-induced liver fibrosis, we used a PPAR-α mutant lacking its DNA-binding-dependent activity on fatty acid metabolism. Liver-specific expression of wild-type or a DNA-binding-deficient PPAR-α in acute and chronic models of inflammation were used to study PPAR-α's anti-inflammatory versus metabolic activities in NASH and fibrosis. Pharmacologically activated PPAR-α inhibited hepatic inflammatory responses and the transition from steatosis toward NASH and fibrosis through a direct, anti-inflammatory mechanism independent of its lipid handling properties. CONCLUSION: The transrepression activity of PPAR-α on chronic liver inflammation is sufficient to prevent progression of NASH to liver fibrosis. Dissociated PPAR-α agonists, selectively modulating PPAR-α transrepression activity, could thus be an option to prevent NASH and fibrosis progression.


Asunto(s)
Hígado Graso/complicaciones , Cirrosis Hepática/etiología , PPAR alfa/metabolismo , Animales , Expresión Génica , Lipopolisacáridos , Cirrosis Hepática/metabolismo , Cirrosis Hepática/prevención & control , Ratones Endogámicos C57BL , Mutación , PPAR alfa/agonistas , PPAR alfa/genética , Transducción de Señal
6.
Arterioscler Thromb Vasc Biol ; 31(7): 1573-9, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21474829

RESUMEN

OBJECTIVE: Peroxisome proliferator-activated receptor-α (PPARα) is a ligand-activated transcription factor that controls lipid metabolism and inflammation. PPARα is activated by fibrates, hypolipidemic drugs used in the treatment of dyslipidemia. Previous studies assessing the influence of PPARα agonists on atherosclerosis in mice yielded conflicting results, and the implication of PPARα therein has not been assessed. The human apolipoprotein E2 knock-in (apoE2-KI) mouse is a model of mixed dyslipidemia, atherosclerosis, and nonalcoholic steatohepatitis (NASH). The aim of this study was to analyze, using homo- and heterozygous PPARα-deficient mice, the consequences of quantitative variations of PPARα gene levels and their response to the synthetic PPARα agonist fenofibrate on NASH and atherosclerosis in apoE2-KI mice. METHODS AND RESULTS: Wild-type (+/+), heterozygous (+/-), and homozygous (-/-) PPARα-deficient mice in the apoE2-KI background were generated and subjected to a Western diet supplemented with fenofibrate or not supplemented. Western diet-fed PPARα-/- apoE2-KI mice displayed an aggravation of liver steatosis and inflammation compared with PPARα+/+ and PPARα+/- apoE2-KI mice, indicating a role of PPARα in liver protection. Moreover, PPARα expression was required for the fenofibrate-induced protection against NASH. Interestingly, fenofibrate treatment induced a similar response on hepatic lipid metabolism in PPARα+/+ and PPARα+/- apoE2-KI mice, whereas, for a maximal antiinflammatory response, both alleles of the PPARα gene were required. Surprisingly, atherosclerosis development was not significantly different among PPARα+/+, PPARα+/-, and PPARα-/- apoE2-KI mice. However, PPARα gene level determined both the antiatherosclerotic and vascular antiinflammatory responses to fenofibrate in a dose-dependent manner. CONCLUSIONS: These results demonstrate a necessary but quantitatively different role of PPARα in the modulation of liver metabolism, inflammation, and atherogenesis.


Asunto(s)
Aorta/metabolismo , Apolipoproteína E2/metabolismo , Aterosclerosis/metabolismo , Inflamación/metabolismo , Metabolismo de los Lípidos , Hígado/metabolismo , PPAR alfa/metabolismo , Análisis de Varianza , Animales , Antiinflamatorios/farmacología , Aorta/efectos de los fármacos , Aorta/patología , Apolipoproteína E2/genética , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/genética , Aterosclerosis/patología , Modelos Animales de Enfermedad , Hígado Graso/tratamiento farmacológico , Hígado Graso/genética , Hígado Graso/metabolismo , Hígado Graso/patología , Femenino , Fenofibrato/farmacología , Regulación de la Expresión Génica , Técnicas de Sustitución del Gen , Heterocigoto , Homocigoto , Humanos , Hipolipemiantes/farmacología , Inflamación/tratamiento farmacológico , Inflamación/genética , Inflamación/patología , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/genética , Lípidos/sangre , Hígado/efectos de los fármacos , Hígado/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Enfermedad del Hígado Graso no Alcohólico , PPAR alfa/agonistas , PPAR alfa/genética
7.
Nat Metab ; 4(8): 1071-1083, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35995995

RESUMEN

Dual agonists activating the peroxisome proliferator-activated receptors alpha and gamma (PPARɑ/ɣ) have beneficial effects on glucose and lipid metabolism in patients with type 2 diabetes, but their development was discontinued due to potential adverse effects. Here we report the design and preclinical evaluation of a molecule that covalently links the PPARɑ/ɣ dual-agonist tesaglitazar to a GLP-1 receptor agonist (GLP-1RA) to allow for GLP-1R-dependent cellular delivery of tesaglitazar. GLP-1RA/tesaglitazar does not differ from the pharmacokinetically matched GLP-1RA in GLP-1R signalling, but shows GLP-1R-dependent PPARɣ-retinoic acid receptor heterodimerization and enhanced improvements of body weight, food intake and glucose metabolism relative to the GLP-1RA or tesaglitazar alone in obese male mice. The conjugate fails to affect body weight and glucose metabolism in GLP-1R knockout mice and shows preserved effects in obese mice at subthreshold doses for the GLP-1RA and tesaglitazar. Liquid chromatography-mass spectrometry-based proteomics identified PPAR regulated proteins in the hypothalamus that are acutely upregulated by GLP-1RA/tesaglitazar. Our data show that GLP-1RA/tesaglitazar improves glucose control with superior efficacy to the GLP-1RA or tesaglitazar alone and suggest that this conjugate might hold therapeutic value to acutely treat hyperglycaemia and insulin resistance.


Asunto(s)
Diabetes Mellitus Tipo 2 , PPAR alfa , Alcanosulfonatos , Animales , Peso Corporal , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Péptido 1 Similar al Glucagón/uso terapéutico , Receptor del Péptido 1 Similar al Glucagón , Glucosa , Masculino , Ratones , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , PPAR alfa/agonistas , PPAR alfa/uso terapéutico , Fenilpropionatos
8.
Nat Commun ; 13(1): 5324, 2022 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-36088459

RESUMEN

Tissue injury triggers activation of mesenchymal lineage cells into wound-repairing myofibroblasts, whose unrestrained activity leads to fibrosis. Although this process is largely controlled at the transcriptional level, whether the main transcription factors involved have all been identified has remained elusive. Here, we report multi-omics analyses unraveling Basonuclin 2 (BNC2) as a myofibroblast identity transcription factor. Using liver fibrosis as a model for in-depth investigations, we first show that BNC2 expression is induced in both mouse and human fibrotic livers from different etiologies and decreases upon human liver fibrosis regression. Importantly, we found that BNC2 transcriptional induction is a specific feature of myofibroblastic activation in fibrotic tissues. Mechanistically, BNC2 expression and activities allow to integrate pro-fibrotic stimuli, including TGFß and Hippo/YAP1 signaling, towards induction of matrisome genes such as those encoding type I collagen. As a consequence, Bnc2 deficiency blunts collagen deposition in livers of mice fed a fibrogenic diet. Additionally, our work establishes BNC2 as potentially druggable since we identified the thalidomide derivative CC-885 as a BNC2 inhibitor. Altogether, we propose that BNC2 is a transcription factor involved in canonical pathways driving myofibroblastic activation in fibrosis.


Asunto(s)
Cirrosis Hepática , Miofibroblastos , Animales , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Genómica , Humanos , Cirrosis Hepática/genética , Cirrosis Hepática/metabolismo , Ratones , Miofibroblastos/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
9.
Arterioscler Thromb Vasc Biol ; 30(5): 894-9, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20393155

RESUMEN

Several decades ago, fibrates were approved for the treatment of dyslipidemia, whereas thiazolidinediones were screened in animal models to improve glucose homeostasis and were subsequently developed for the treatment of type 2 diabetes mellitus. Relatively recently, these drugs were found to act via peroxisome proliferator-activated receptors, nuclear receptors that control lipid metabolism and glucose homeostasis. In this historical perspective, we discuss the history of discovery of the peroxisome proliferator-activated receptors, from the clinical development of their agonists to the subsequent discovery of these receptors and their mechanisms of action, to finally evoke possibilities of targeted pharmacology for future development of selective peroxisome proliferator-activated receptor modulators.


Asunto(s)
Ácido Clofíbrico/historia , Diabetes Mellitus Tipo 2/historia , Dislipidemias/historia , Hipoglucemiantes/historia , Hipolipemiantes/historia , Receptores Activados del Proliferador del Peroxisoma/historia , Tiazolidinedionas/historia , Animales , Glucemia/efectos de los fármacos , Ácido Clofíbrico/uso terapéutico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Descubrimiento de Drogas/historia , Dislipidemias/tratamiento farmacológico , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Hipoglucemiantes/uso terapéutico , Hipolipemiantes/uso terapéutico , Metabolismo de los Lípidos/efectos de los fármacos , Receptores Activados del Proliferador del Peroxisoma/agonistas , Tiazolidinedionas/uso terapéutico , Resultado del Tratamiento
10.
JCI Insight ; 6(16)2021 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-34228639

RESUMEN

Among genetic susceptibility loci associated with late-onset Alzheimer disease (LOAD), genetic polymorphisms identified in genes encoding lipid carriers led to the hypothesis that a disruption of lipid metabolism could promote disease progression. We previously reported that amyloid precursor protein (APP) involved in Alzheimer disease (AD) physiopathology impairs lipid synthesis needed for cortical networks' activity and that activation of peroxisome proliferator-activated receptor α (PPARα), a metabolic regulator involved in lipid metabolism, improves synaptic plasticity in an AD mouse model. These observations led us to investigate a possible correlation between PPARα function and full-length APP expression. Here, we report that PPARα expression and activation were inversely related to APP expression both in LOAD brains and in early-onset AD cases with a duplication of the APP gene, but not in control human brains. Moreover, human APP expression decreased PPARA expression and its related target genes in transgenic mice and in cultured cortical cells, while opposite results were observed in APP-silenced cortical networks. In cultured neurons, APP-mediated decrease or increase in synaptic activity was corrected by a PPARα-specific agonist and antagonist, respectively. APP-mediated control of synaptic activity was abolished following PPARα deficiency, indicating a key function of PPARα in this process.


Asunto(s)
Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/metabolismo , Corteza Cerebral/patología , PPAR alfa/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Animales , Estudios de Casos y Controles , Línea Celular , Corteza Cerebral/citología , Modelos Animales de Enfermedad , Femenino , Duplicación de Gen , Regulación de la Expresión Génica , Humanos , Lipogénesis/genética , Masculino , Ratones Transgénicos , Neuronas , PPAR alfa/agonistas , PPAR alfa/antagonistas & inhibidores , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo
11.
Arterioscler Thromb Vasc Biol ; 29(10): 1488-95, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19592467

RESUMEN

OBJECTIVE: Bexarotene (Targretin) is a clinically used antitumoral agent which exerts its action through binding to and activation of the retinoid-X-receptor (RXR). The most frequent side-effect of bexarotene administration is an increase in plasma triglycerides, an independent risk factor of cardiovascular disease. The molecular mechanism behind this hypertriglyceridemia remains poorly understood. METHODS AND RESULTS: Using wild-type and LXR alpha/beta-deficient mice, we show here that bexarotene induces hypertriglyceridemia and activates hepatic LXR-target genes of lipogenesis in an LXR-dependent manner, hence exerting a permissive effect on RXR/LXR heterodimers. Interestingly, RNA analysis and Chromatin Immunoprecipitation assays performed in the liver reveal that the in vivo permissive effect of bexarotene on the RXR/LXR heterodimer is restricted to lipogenic genes without modulation of genes controlling cholesterol homeostasis. CONCLUSIONS: These findings demonstrate that the hypertriglyceridemic action of bexarotene occurs via the RXR/LXR heterodimer and show that RXR heterodimers can act with a selective permissivity on target genes of specific metabolic pathways in the liver.


Asunto(s)
Colesterol/metabolismo , Proteínas de Unión al ADN/fisiología , Hígado/metabolismo , Receptores Citoplasmáticos y Nucleares/fisiología , Receptores X Retinoide/fisiología , Tetrahidronaftalenos/farmacología , Triglicéridos/metabolismo , Animales , Bexaroteno , Proteínas de Unión al ADN/química , Dimerización , Femenino , Homeostasis , Lipogénesis , Receptores X del Hígado , Ratones , Ratones Endogámicos C57BL , Receptores Nucleares Huérfanos , Receptores Citoplasmáticos y Nucleares/química , Receptores X Retinoide/química
12.
Life Sci Alliance ; 2(2)2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30894406

RESUMEN

Mechanisms driving cognitive improvements following nuclear receptor activation are poorly understood. The peroxisome proliferator-activated nuclear receptor alpha (PPARα) forms heterodimers with the nuclear retinoid X receptor (RXR). We report that PPARα mediates the improvement of hippocampal synaptic plasticity upon RXR activation in a transgenic mouse model with cognitive deficits. This improvement results from an increase in GluA1 subunit expression of the alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor, eliciting an AMPA response at the excitatory synapses. Associated with a two times higher PPARα expression in males than in females, we show that male, but not female, PPARα null mutants display impaired hippocampal long-term potentiation. Moreover, PPARα knockdown in the hippocampus of cognition-impaired mice compromises the beneficial effects of RXR activation on synaptic plasticity only in males. Furthermore, selective PPARα activation with pemafibrate improves synaptic plasticity in male cognition-impaired mice, but not in females. We conclude that striking sex differences in hippocampal synaptic plasticity are observed in mice, related to differences in PPARα expression levels.


Asunto(s)
Dosificación de Gen/genética , Potenciación a Largo Plazo/genética , Plasticidad Neuronal/genética , PPAR alfa/genética , PPAR alfa/metabolismo , Animales , Benzoxazoles/farmacología , Butiratos/farmacología , Células Cultivadas , Disfunción Cognitiva/genética , Disfunción Cognitiva/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Hipocampo/citología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Ratones , Ratones Transgénicos , PPAR alfa/agonistas , Ratas , Ratas Wistar , Receptores AMPA/metabolismo , Receptores X Retinoide/metabolismo , Factores Sexuales , Transducción de Señal/efectos de los fármacos
13.
Diabetes ; 55(6): 1605-13, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16731822

RESUMEN

Peroxisome proliferator-activated receptor (PPAR) alpha is a transcription factor controlling lipid and glucose homeostasis. PPARalpha-deficient (-/-) mice are protected from high-fat diet-induced insulin resistance. However, the impact of PPARalpha in the pathophysiological setting of obesity-related insulin resistance is unknown. Therefore, PPARalpha(-/-) mice in an obese (ob/ob) background were generated. PPARalpha deficiency did not influence the growth curves of the obese mice but surprisingly resulted in a severe, age-dependent hyperglycemia. PPARalpha deficiency did not aggravate peripheral insulin resistance. By contrast, PPARalpha(-/-) ob/ob mice developed pancreatic beta-cell dysfunction characterized by reduced mean islet area and decreased insulin secretion in response to glucose in vitro and in vivo. In primary human pancreatic islets, PPARalpha agonist treatment prevented fatty acid-induced impairment of glucose-stimulated insulin secretion, apoptosis, and triglyceride accumulation. These results indicate that PPARalpha improves the adaptative response of the pancreatic beta-cell to pathological conditions. PPARalpha could thus represent a promising target in the prevention of type 2 diabetes.


Asunto(s)
Resistencia a la Insulina/fisiología , Islotes Pancreáticos/metabolismo , PPAR alfa/fisiología , Páncreas/metabolismo , Adulto , Factores de Edad , Animales , Apoptosis/efectos de los fármacos , Peso Corporal , Relación Dosis-Respuesta a Droga , Expresión Génica , Glucosa/farmacología , Humanos , Hiperglucemia/genética , Hiperglucemia/metabolismo , Hiperglucemia/fisiopatología , Técnicas In Vitro , Insulina/metabolismo , Resistencia a la Insulina/genética , Secreción de Insulina , Islotes Pancreáticos/citología , Islotes Pancreáticos/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Ratones Obesos , Persona de Mediana Edad , PPAR alfa/genética , Palmitatos/farmacología , Páncreas/citología , Páncreas/efectos de los fármacos , Triglicéridos/metabolismo
14.
Arterioscler Thromb Vasc Biol ; 26(12): 2731-7, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17008586

RESUMEN

OBJECTIVE: The activity of the antitumoral agent bexarotene (Targretin, Bexarotene) depends on its binding to the nuclear retinoid-X receptor (RXR) and subsequent transcriptional regulation of target genes. Through RXR activation, bexarotene may modulate numerous metabolic pathways involved in atherosclerosis. Here, we investigated the effect of bexarotene on atherosclerosis progression in a dyslipidemic murine model, the human apolipoprotein E2 knockin mouse, that develops essentially macrophage-laden lesions. METHODS AND RESULTS: Atherosclerotic lesions together with different metabolic pathways involved in atherosclerosis were investigated in mice treated or not with bexarotene. Bexarotene protects from atherosclerosis development in mice, at least in part by improving the circulating cholesterol distribution profile likely via a marked decrease of dietary cholesterol absorption caused by modulation of intestinal expression of genes recently identified as major players in this process, Niemann-Pick-C1-Like1 (NPC1L1) and CD13. This atheroprotection appears despite a strong hypertriglyceridemia. Moreover, bexarotene treatment only modestly modulates inflammatory gene expression in the vascular wall, but markedly enhanced the capacity of macrophages to efflux cellular lipids. CONCLUSIONS: These data provide evidence of a favorable pharmacological effect of bexarotene on atherosclerosis despite the induction of hypertriglyceridemia, likely via a beneficial action on intestinal absorption and macrophage efflux.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Aterosclerosis/etiología , Colesterol/metabolismo , Dislipidemias/complicaciones , Homeostasis/efectos de los fármacos , Receptores X Retinoide/agonistas , Tetrahidronaftalenos/farmacología , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1 , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Apolipoproteína E2/genética , Apolipoproteína E2/metabolismo , Aterosclerosis/metabolismo , Bexaroteno , Antígenos CD13/genética , Antígenos CD13/metabolismo , Modelos Animales de Enfermedad , Dislipidemias/tratamiento farmacológico , Dislipidemias/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Homeostasis/fisiología , Absorción Intestinal/efectos de los fármacos , Lipoproteínas/genética , Lipoproteínas/metabolismo , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Tetrahidronaftalenos/uso terapéutico , Triglicéridos/sangre
15.
JCI Insight ; 2(13)2017 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-28679947

RESUMEN

Nonalcoholic fatty liver disease prevalence is soaring with the obesity pandemic, but the pathogenic mechanisms leading to the progression toward active nonalcoholic steatohepatitis (NASH) and fibrosis, major causes of liver-related death, are poorly defined. To identify key components during the progression toward NASH and fibrosis, we investigated the liver transcriptome in a human cohort of NASH patients. The transition from histologically proven fatty liver to NASH and fibrosis was characterized by gene expression patterns that successively reflected altered functions in metabolism, inflammation, and epithelial-mesenchymal transition. A meta-analysis combining our and public human transcriptomic datasets with murine models of NASH and fibrosis defined a molecular signature characterizing NASH and fibrosis and evidencing abnormal inflammation and extracellular matrix (ECM) homeostasis. Dermatopontin expression was found increased in fibrosis, and reversal of fibrosis after gastric bypass correlated with decreased dermatopontin expression. Functional studies in mice identified an active role for dermatopontin in collagen deposition and fibrosis. PPARα activation lowered dermatopontin expression through a transrepressive mechanism affecting the Klf6/TGFß1 pathway. Liver fibrotic histological damages are thus characterized by the deregulated expression of a restricted set of inflammation- and ECM-related genes. Among them, dermatopontin may be a valuable target to reverse the hepatic fibrotic process.

16.
Mol Endocrinol ; 29(8): 1134-43, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26087172

RESUMEN

Acute liver failure (ALF) is a severe and rapid liver injury, often occurring without any preexisting liver disease, which may precipitate multiorgan failure and death. ALF is often associated with impaired ß-oxidation and increased oxidative stress (OS), characterized by elevated levels of hepatic reactive oxygen species (ROS) and lipid peroxidation (LPO) products. Peroxisome proliferator-activated receptor (PPAR)α has been shown to confer hepatoprotection in acute and chronic liver injury, at least in part, related to its ability to control peroxisomal and mitochondrial ß-oxidation. To study the pathophysiological role of PPARα in hepatic response to high OS, we induced a pronounced LPO by treating wild-type and Pparα-deficient mice with high doses of fish oil (FO), containing n-3 polyunsaturated fatty acids. FO feeding of Pparα-deficient mice, in contrast to control sunflower oil, surprisingly induced coma and death due to ALF as indicated by elevated serum alanine aminotransferase, aspartate aminotransferase, ammonia, and a liver-specific increase of ROS and LPO-derived malondialdehyde. Reconstitution of PPARα specifically in the liver using adeno-associated serotype 8 virus-PPARα in Pparα-deficient mice restored ß-oxidation and ketogenesis and protected mice from FO-induced lipotoxicity and death. Interestingly, administration of the ketone body ß-hydroxybutyrate prevented FO-induced ALF in Pparα-deficient mice, and normalized liver ROS and malondialdehyde levels. Therefore, PPARα protects the liver from FO-induced OS through its regulatory actions on ketone body levels. ß-Hydroxybutyrate treatment could thus be an option to prevent LPO-induced liver damage.


Asunto(s)
Hígado Graso/metabolismo , Cuerpos Cetónicos/química , Fallo Hepático Agudo/metabolismo , PPAR alfa/deficiencia , PPAR alfa/metabolismo , Ácido 3-Hidroxibutírico/química , Animales , Ácidos Grasos/metabolismo , Hígado Graso/prevención & control , Femenino , Peroxidación de Lípido , Hígado/metabolismo , Fallo Hepático Agudo/prevención & control , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Oxidación-Reducción , Estrés Oxidativo , Peroxisomas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Triglicéridos/metabolismo
17.
Mol Cell Biol ; 32(4): 852-67, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22158963

RESUMEN

The liver X receptors (LXRs) are nuclear receptors that form permissive heterodimers with retinoid X receptor (RXR) and are important regulators of lipid metabolism in the liver. We have recently shown that RXR agonist-induced hypertriglyceridemia and hepatic steatosis in mice are dependent on LXRs and correlate with an LXR-dependent hepatic induction of lipogenic genes. To further investigate the roles of RXR and LXR in the regulation of hepatic gene expression, we have mapped the ligand-regulated genome-wide binding of these factors in mouse liver. We find that the RXR agonist bexarotene primarily increases the genomic binding of RXR, whereas the LXR agonist T0901317 greatly increases both LXR and RXR binding. Functional annotation of putative direct LXR target genes revealed a significant association with classical LXR-regulated pathways as well as peroxisome proliferator-activated receptor (PPAR) signaling pathways, and subsequent chromatin immunoprecipitation-sequencing (ChIP-seq) mapping of PPARα binding demonstrated binding of PPARα to 71 to 88% of the identified LXR-RXR binding sites. The combination of sequence analysis of shared binding regions and sequential ChIP on selected sites indicate that LXR-RXR and PPARα-RXR bind to degenerate response elements in a mutually exclusive manner. Together, our findings suggest extensive and unexpected cross talk between hepatic LXR and PPARα at the level of binding to shared genomic sites.


Asunto(s)
Hígado/metabolismo , Receptores Nucleares Huérfanos/genética , PPAR alfa/genética , Receptores X Retinoide/genética , Animales , Secuencia de Bases , Sitios de Unión/genética , Cartilla de ADN/genética , Femenino , Perfilación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Receptores X del Hígado , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Nucleares Huérfanos/deficiencia , Receptores Nucleares Huérfanos/metabolismo , PPAR alfa/agonistas , PPAR alfa/metabolismo , Receptor Cross-Talk , Receptores X Retinoide/metabolismo
18.
Atherosclerosis ; 218(1): 69-76, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21529810

RESUMEN

BACKGROUND: Atherosclerosis and non-alcoholic fatty liver disease (NAFLD) are complex pathologies characterized by lipid accumulation, chronic inflammation and extensive tissue remodelling. Microparticles (MPs), small membrane vesicles produced by activated and apoptotic cells, might not only be biomarkers, but also functional actors in these pathologies. The apoE2-KI mouse is a model of atherosclerosis and NAFLD. Activation of the nuclear receptor PPARα decreases atherosclerosis and components of non-alcoholic steatohepatitis (NASH) in the apoE2-KI mouse. OBJECTIVES: (1) To determine whether MPs are present in atherosclerotic lesions, liver and plasma during atherosclerosis and NASH progression in apoE2-KI mice, and (2) to study whether PPARα activation modulates MP concentrations. METHODS: ApoE2-KI mice were fed a Western diet to induce atherosclerosis and NASH. MPs were isolated from atherosclerotic lesions, liver and blood and quantified by flow cytometry. RESULTS: An increase of MPs was observed in the atherosclerotic lesions and in the liver of apoE2-KI mice upon Western diet feeding. PPARα activation with fenofibrate decreased MP levels in the atherosclerotic lesions in a PPARα-dependent manner, but did not influence MP concentrations in the liver. CONCLUSION: Here we report that MPs are present in atherosclerotic lesions and in the liver of apoE2-KI mice. Their concentration increased during atherosclerosis and NASH development. PPARα activation differentially modulates MP levels in a tissue-specific manner.


Asunto(s)
Aterosclerosis/metabolismo , Hígado Graso/metabolismo , Hígado/metabolismo , PPAR alfa/metabolismo , Animales , Biomarcadores/metabolismo , Micropartículas Derivadas de Células/metabolismo , Modelos Animales de Enfermedad , Femenino , Fenofibrato/química , Citometría de Flujo/métodos , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Enfermedad del Hígado Graso no Alcohólico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA