Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
JCI Insight ; 7(17)2022 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-35917173

RESUMEN

The sarcoplasmic reticulum (SR) plays an important role in calcium homeostasis. SR calcium mishandling is described in pathological conditions, such as myopathies. Here, we investigated whether the nuclear receptor subfamily 1 group D member (NR1D1, also called REV-ERBα) regulates skeletal muscle SR calcium homeostasis. Our data demonstrate that NR1D1 deficiency in mice impaired sarco/endoplasmic reticulum calcium ATPase-dependent (SERCA-dependent) SR calcium uptake. NR1D1 acts on calcium homeostasis by repressing the SERCA inhibitor myoregulin through direct binding to its promoter. Restoration of myoregulin counteracted the effects of NR1D1 overexpression on SR calcium content. Interestingly, myoblasts from patients with Duchenne muscular dystrophy displayed lower NR1D1 expression, whereas pharmacological NR1D1 activation ameliorated SR calcium homeostasis and improved muscle structure and function in dystrophic mdx/Utr+/- mice. Our findings demonstrate that NR1D1 regulates muscle SR calcium homeostasis, pointing to its therapeutic potential for mitigating myopathy.


Asunto(s)
Calcio , Músculo Esquelético , Animales , Calcio/metabolismo , Homeostasis , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/metabolismo , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/genética , Retículo Sarcoplasmático/metabolismo
2.
Sci Rep ; 10(1): 21095, 2020 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-33273527

RESUMEN

Retinoic acid receptor-related orphan receptor-alpha (RORα) is a transcription factor from the nuclear receptor family expressed by immune cells and involved in the development of obesity, insulin resistance (IR) and non-alcoholic steatohepatitis (NASH). It was recently reported that mice deficient for RORα in macrophages develop more severe NASH upon high fat diet (HFD) feeding due to altered Kupffer cell function. To better understand the role of RORα in obesity and IR, we independently generated a macrophage RORα-deficient mouse line. We report that RORα deletion in macrophages does not impact on HFD-induced obesity and IR. Surprisingly, we did not confirm an effect on NASH development upon HFD feeding nor in the more severe and obesity-independent choline-deficient, L-amino acid-defined diet model. Our results therefore show that RORα deletion in macrophages does not alter the development of obesity and IR and question its role in NASH.


Asunto(s)
Resistencia a la Insulina , Macrófagos/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Obesidad/metabolismo , Animales , Células Cultivadas , Dieta Alta en Grasa/efectos adversos , Eliminación de Gen , Macrófagos del Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/etiología , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Obesidad/etiología
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.
Acta Diabetol ; 57(7): 819-826, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32086613

RESUMEN

AIMS: Not all people with obesity become glucose intolerant, suggesting differential activation of cellular pathways. The unfolded protein response (UPR) may contribute to the development of insulin resistance in several organs, but its role in skeletal muscle remains debated. Therefore, we explored the UPR activation in muscle from non-diabetic glucose tolerant or intolerant patients with obesity and the impact of bariatric procedures. METHODS: Muscle biopsies from 22 normoglycemic (NG, blood glucose measured 120 min after an oral glucose tolerance test, G120 < 7.8 mM) and 22 glucose intolerant (GI, G120 between 7.8 and 11.1 mM) patients with obesity were used to measure UPR activation by RTqPCR and western blot. Then, UPR was studied in biopsies from 7 NG and 7 GI patients before and 1 year after bariatric surgery. RESULTS: Binding immunoglobulin protein (BIP) protein was ~ 40% higher in the GI compared to NG subjects. Contrastingly, expression of the UPR-related genes BIP, activating transcription factor 6 (ATF6) and unspliced X-box binding protein 1 (XBP1u) were significantly lower and C/EBP homologous protein (CHOP) tended to decrease (p = 0.08) in GI individuals. While BIP protein positively correlated with fasting blood glucose (r = 0.38, p = 0.01), ATF6 and CHOP were associated with G120 (r = - 0.38 and r = - 0.41, p < 0.05) and the Matsuda index (r = 0.37 and r = 0.38, p < 0.05). Bariatric surgery improved metabolic parameters, associated with higher CHOP expression in GI patients, while ATF6 tended to increase (p = 0.08). CONCLUSIONS: CHOP and ATF6 expression decreased in non-diabetic GI patients with obesity and was modified by bariatric surgery. These genes may contribute to glucose homeostasis in human skeletal muscle.


Asunto(s)
Cirugía Bariátrica , Intolerancia a la Glucosa/cirugía , Músculo Esquelético/metabolismo , Obesidad Mórbida/cirugía , Respuesta de Proteína Desplegada , Factor de Transcripción Activador 6/genética , Factor de Transcripción Activador 6/metabolismo , Adulto , Biopsia , Glucemia/metabolismo , Estudios de Casos y Controles , Estudios de Cohortes , Estrés del Retículo Endoplásmico/genética , Estrés del Retículo Endoplásmico/fisiología , Femenino , Regulación de la Expresión Génica , Intolerancia a la Glucosa/complicaciones , Intolerancia a la Glucosa/metabolismo , Intolerancia a la Glucosa/patología , Prueba de Tolerancia a la Glucosa , Humanos , Resistencia a la Insulina/fisiología , Masculino , Músculo Esquelético/patología , Obesidad Mórbida/complicaciones , Obesidad Mórbida/metabolismo , Obesidad Mórbida/patología , Factor de Transcripción CHOP/genética , Factor de Transcripción CHOP/metabolismo
5.
Cell Rep ; 29(6): 1410-1418.e6, 2019 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-31693883

RESUMEN

Browning induction or transplantation of brown adipose tissue (BAT) or brown/beige adipocytes derived from progenitor or induced pluripotent stem cells (iPSCs) can represent a powerful strategy to treat metabolic diseases. However, our poor understanding of the mechanisms that govern the differentiation and activation of brown adipocytes limits the development of such therapy. Various genetic factors controlling the differentiation of brown adipocytes have been identified, although most studies have been performed using in vitro cultured pre-adipocytes. We investigate here the differentiation of brown adipocytes from adipose progenitors in the mouse embryo. We demonstrate that the formation of multiple lipid droplets (LDs) is initiated within clusters of glycogen, which is degraded through glycophagy to provide the metabolic substrates essential for de novo lipogenesis and LD formation. Therefore, this study uncovers the role of glycogen in the generation of LDs.


Asunto(s)
Adipocitos Marrones/metabolismo , Adipogénesis/genética , Tejido Adiposo Pardo/metabolismo , Embrión de Mamíferos/metabolismo , Glucógeno/metabolismo , Gotas Lipídicas/metabolismo , Adipocitos Marrones/ultraestructura , Tejido Adiposo Pardo/embriología , Tejido Adiposo Pardo/ultraestructura , Animales , Autofagia/efectos de los fármacos , Autofagia/genética , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Células Cultivadas , Proteínas de Unión a Ácidos Grasos/genética , Proteínas de Unión a Ácidos Grasos/metabolismo , Glucógeno/ultraestructura , Humanos , Gotas Lipídicas/ultraestructura , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , PPAR gamma/genética , PPAR gamma/metabolismo , ARN Interferente Pequeño , Transcriptoma
6.
JCI Insight ; 3(9)2018 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-29720572

RESUMEN

Metabolic stresses such as dietary energy restriction or physical activity exert beneficial metabolic effects. In the liver, endospanin-1 and endospanin-2 cooperatively modulate calorie restriction-mediated (CR-mediated) liver adaptations by controlling growth hormone sensitivity. Since we found CR to induce endospanin protein expression in skeletal muscle, we investigated their role in this tissue. In vivo and in vitro endospanin-2 triggers ERK phosphorylation in skeletal muscle through an autophagy-dependent pathway. Furthermore, endospanin-2, but not endospanin-1, overexpression decreases muscle mitochondrial ROS production, induces fast-to-slow fiber-type switch, increases skeletal muscle glycogen content, and improves glucose homeostasis, ultimately promoting running endurance capacity. In line, endospanin-2-/- mice display higher lipid peroxidation levels, increased mitochondrial ROS production under mitochondrial stress, decreased ERK phosphorylation, and reduced endurance capacity. In conclusion, our results identify endospanin-2 as a potentially novel player in skeletal muscle metabolism, plasticity, and function.


Asunto(s)
Metabolismo Energético , Proteínas de la Membrana/fisiología , Músculo Esquelético/metabolismo , Resistencia Física/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/fisiología , Animales , Autofagia , Restricción Calórica , Plasticidad de la Célula/genética , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular , Sistema de Señalización de MAP Quinasas , Masculino , Proteínas de la Membrana/genética , Ratones , Mitocondrias/metabolismo , Fibras Musculares de Contracción Rápida/fisiología , Fibras Musculares de Contracción Lenta/fisiología , Músculo Esquelético/citología , Músculo Esquelético/fisiología , Estrés Oxidativo , Fenotipo , Fosforilación , Esfuerzo Físico , ARN Mensajero/metabolismo
7.
Gastroenterology ; 154(5): 1449-1464.e20, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29277561

RESUMEN

BACKGROUND & AIMS: The innate immune system responds not only to bacterial signals, but also to non-infectious danger-associated molecular patterns that activate the NLRP3 inflammasome complex after tissue injury. Immune functions vary over the course of the day, but it is not clear whether these changes affect the activity of the NLRP3 inflammasome. We investigated whether the core clock component nuclear receptor subfamily 1 group D member 1 (NR1D1, also called Rev-erbα) regulates expression, activity of the NLRP3 inflammasome, and its signaling pathway. METHODS: We collected naïve peritoneal macrophages and plasma, at multiple times of day, from Nr1d1-/- mice and their Nr1d1+/+ littermates (controls) and analyzed expression NLRP3, interleukin 1ß (IL1B, in plasma), and IL18 (in plasma). We also collected bone marrow-derived primary macrophages from these mice. Levels of NR1D1 were knocked down with small hairpin RNAs in human primary macrophages. Bone marrow-derived primary macrophages from mice and human primary macrophages were incubated with lipopolysaccharide (LPS) to induce expression of NLRP3, IL1B, and IL18; cells were incubated with LPS and adenosine triphosphate to activate the NLRP3 complex. We analyzed caspase 1 activity and cytokine secretion. NR1D1 was activated in primary mouse and human macrophages by incubation with SR9009; some of the cells were also incubated with an NLRP3 inhibitor or inhibitors of caspase 1. Nr1d1-/- mice and control mice were given intraperitoneal injections of LPS to induce peritoneal inflammation; plasma samples were isolated and levels of cytokines were measured. Nr1d1-/- mice, control mice, and control mice given injections of SR9009 were given LPS and D-galactosamine to induce fulminant hepatitis and MCC950 to specifically inhibit NLRP3; plasma was collected to measure cytokines and a marker of liver failure (alanine aminotransferase); liver tissues were collected and analyzed by quantitative polymerase chain reaction, immunohistochemistry, and flow cytometry. RESULTS: In peritoneal macrophages, expression of NLRP3 and activation of its complex varied with time of day (circadian rhythm)-this regulation required NR1D1. Primary macrophages from Nr1d1-/- mice and human macrophages with knockdown of NR1D1 had altered expression patterns of NLRP3, compared to macrophages that expressed NR1D1, and altered patterns of IL1B and 1L18 production. Mice with disruption of Nr1d1 developed more-severe acute peritoneal inflammation and fulminant hepatitis than control mice. Incubation of macrophage with the NR1D1 activator SR9009 reduced expression of NLRP3 and secretion of cytokines. Mice given SR9009 developed less-severe liver failure and had longer survival times than mice given saline (control). CONCLUSIONS: In studies of Nr1d1-/- mice and human macrophages with pharmacologic activation of NR1D1, we found NR1D1 to regulate the timing of NLRP3 expression and production of inflammatory cytokines by macrophages. Activation of NR1D1 reduced the severity of peritoneal inflammation and fulminant hepatitis in mice.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Ritmo Circadiano , Inflamasomas/metabolismo , Fallo Hepático Agudo/prevención & control , Hígado/metabolismo , Macrófagos Peritoneales/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/metabolismo , Animales , Caspasa 1/metabolismo , Células Cultivadas , Enfermedad Hepática Inducida por Sustancias y Drogas/inmunología , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Galactosamina , Predisposición Genética a la Enfermedad , Inflamasomas/genética , Inflamasomas/inmunología , Lipopolisacáridos , Hígado/efectos de los fármacos , Hígado/inmunología , Hígado/patología , Fallo Hepático Agudo/inmunología , Fallo Hepático Agudo/metabolismo , Fallo Hepático Agudo/patología , Activación de Macrófagos , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/patología , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/agonistas , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/deficiencia , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/genética , Peritonitis/inmunología , Peritonitis/metabolismo , Peritonitis/prevención & control , Fenotipo , Pirrolidinas/farmacología , Interferencia de ARN , Índice de Severidad de la Enfermedad , Transducción de Señal , Tiofenos/farmacología , Factores de Tiempo , Transfección
8.
Sci Rep ; 7(1): 14383, 2017 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-29085009

RESUMEN

The nuclear receptor Rev-erb-α modulates hepatic lipid and glucose metabolism, adipogenesis and thermogenesis. We have previously demonstrated that Rev-erb-α is also an important regulator of skeletal muscle mitochondrial biogenesis and function, and autophagy. As such, Rev-erb-α over-expression in skeletal muscle or its pharmacological activation improved mitochondrial respiration and enhanced exercise capacity. Here, in gain- and loss-of function studies, we show that Rev-erb-α also controls muscle mass. Rev-erb-α-deficiency in skeletal muscle leads to increased expression of the atrophy-related genes (atrogenes), associated with reduced muscle mass and decreased fiber size. By contrast, in vivo and in vitro Rev-erb-α over-expression results in reduced atrogenes expression and increased fiber size. Finally, Rev-erb-α pharmacological activation blocks dexamethasone-induced upregulation of atrogenes and muscle atrophy. This study identifies Rev-erb-α as a promising pharmacological target to preserve muscle mass.


Asunto(s)
Atrofia Muscular/genética , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/fisiología , Adipogénesis , Animales , Autofagia , Diferenciación Celular , Hígado/metabolismo , Ratones , Ratones Noqueados , Músculo Esquelético/metabolismo , Atrofia Muscular/metabolismo , Enfermedades Musculares/metabolismo , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/metabolismo , Proteínas Represoras/genética , Activación Transcripcional
9.
Diab Vasc Dis Res ; 14(6): 516-524, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28868898

RESUMEN

The genomic CDKN2A/B locus, encoding p16INK4a among others, is linked to an increased risk for cardiovascular disease and type 2 diabetes. Obesity is a risk factor for both cardiovascular disease and type 2 diabetes. p16INK4a is a cell cycle regulator and tumour suppressor. Whether it plays a role in adipose tissue formation is unknown. p16INK4a knock-down in 3T3/L1 preadipocytes or p16INK4a deficiency in mouse embryonic fibroblasts enhanced adipogenesis, suggesting a role for p16INK4a in adipose tissue formation. p16INK4a-deficient mice developed more epicardial adipose tissue in response to the adipogenic peroxisome proliferator activated receptor gamma agonist rosiglitazone. Additionally, adipose tissue around the aorta from p16INK4a-deficient mice displayed enhanced rosiglitazone-induced gene expression of adipogenic markers and stem cell antigen, a marker of bone marrow-derived precursor cells. Mice transplanted with p16INK4a-deficient bone marrow had more epicardial adipose tissue compared to controls when fed a high-fat diet. In humans, p16INK4a gene expression was enriched in epicardial adipose tissue compared to other adipose tissue depots. Moreover, epicardial adipose tissue from obese humans displayed increased expression of stem cell antigen compared to lean controls, supporting a bone marrow origin of epicardial adipose tissue. These results show that p16INK4a modulates epicardial adipose tissue development, providing a potential mechanistic link between the genetic association of the CDKN2A/B locus and cardiovascular disease risk.


Asunto(s)
Adipocitos/metabolismo , Adipogénesis , Tejido Adiposo/metabolismo , Médula Ósea/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/metabolismo , Obesidad/metabolismo , Células Madre/metabolismo , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Adipocitos/patología , Adipogénesis/efectos de los fármacos , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/patología , Adiposidad , Adulto , Anciano , Animales , Trasplante de Médula Ósea , Estudios de Casos y Controles , Inhibidor p16 de la Quinasa Dependiente de Ciclina/deficiencia , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Modelos Animales de Enfermedad , Femenino , Genotipo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Obesidad/genética , Obesidad/patología , Obesidad/fisiopatología , PPAR gamma/agonistas , PPAR gamma/metabolismo , Fenotipo , Interferencia de ARN , Receptores de LDL/genética , Receptores de LDL/metabolismo , Rosiglitazona , Transducción de Señal , Células Madre/efectos de los fármacos , Tiazolidinedionas/farmacología , Transfección
10.
Nat Med ; 19(8): 1039-46, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23852339

RESUMEN

The nuclear receptor Rev-erb-α modulates hepatic lipid and glucose metabolism, adipogenesis and the inflammatory response in macrophages. We show here that Rev-erb-α is highly expressed in oxidative skeletal muscle and that its deficiency in muscle leads to reduced mitochondrial content and oxidative function, as well as upregulation of autophagy. These cellular effects resulted in both impaired mitochondrial biogenesis and increased clearance of this organelle, leading to compromised exercise capacity. On a molecular level, Rev-erb-α deficiency resulted in deactivation of the Lkb1-Ampk-Sirt1-Ppargc-1α signaling pathway. These effects were recapitulated in isolated fibers and in muscle cells after knockdown of the gene encoding Rev-erb-α, Nr1d1. In complementary experiments, Rev-erb-α overexpression in vitro increased the number of mitochondria and improved respiratory capacity, whereas muscle overexpression or pharmacological activation of Rev-erb-α in vivo increased exercise capacity. This study identifies Rev-erb-α as a pharmacological target that improves muscle oxidative function by modulating gene networks controlling mitochondrial number and function.


Asunto(s)
Autofagia , Recambio Mitocondrial , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/metabolismo , Animales , Respiración de la Célula , Ratones , Mitocondrias Musculares/metabolismo , Mitocondrias Musculares/ultraestructura , Actividad Motora , Músculo Esquelético/ultraestructura , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/deficiencia , Oxidación-Reducción , Condicionamiento Físico Animal , Transducción de Señal , Factores de Tiempo
11.
Arterioscler Thromb Vasc Biol ; 32(3): 677-85, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22207732

RESUMEN

OBJECTIVE: 11ß-Hydroxysteroid dehydrogenase type 1 (11ß-HSD1) catalyzes the intracellular reduction of inactive cortisone to active cortisol, the natural ligand activating the glucocorticoid receptor (GR). Peroxisome proliferator- activated receptor-γ (PPARγ) is a nuclear receptor controlling inflammation, lipid metabolism, and the macrophage polarization state. In this study, we investigated the impact of macrophage polarization on the expression and activity of 11ß-HSD1 and the role of PPARγ therein. METHODS AND RESULTS: 11ß-HSD1 gene expression is higher in proinflammatory M1 and anti-inflammatory M2 macrophages than in resting macrophages, whereas its activity is highest in M2 macrophages. Interestingly, PPARγ activation induces 11ß-HSD1 enzyme activity in M2 macrophages but not in resting macrophages or M1 macrophages. Consequently, human M2 macrophages displayed enhanced responsiveness to the 11ß-HSD1 substrate cortisone, an effect amplified by PPARγ induction of 11ß-HSD1 activity, as illustrated by an increased expression of GR target genes. CONCLUSION: Our data identify a positive cross-talk between PPARγ and GR in human M2 macrophages via the induction of 11ß-HSD1 expression and activity.


Asunto(s)
Inflamación/enzimología , Macrófagos/efectos de los fármacos , PPAR gamma/agonistas , Tiazolidinedionas/farmacología , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/biosíntesis , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/genética , Células Cultivadas , Cortisona/metabolismo , Inducción Enzimática , Genes Reporteros , Humanos , Hidrocortisona/metabolismo , Inflamación/genética , Inflamación/inmunología , Interleucina-4/metabolismo , Macrófagos/enzimología , Macrófagos/inmunología , PPAR gamma/genética , PPAR gamma/metabolismo , Interferencia de ARN , Receptores de Glucocorticoides/metabolismo , Rosiglitazona , Factores de Tiempo , Transfección
12.
PLoS One ; 6(7): e22545, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21818335

RESUMEN

Retinoic acid-related Orphan Receptor alpha (RORα; NR1F1) is a widely distributed nuclear receptor involved in several (patho)physiological functions including lipid metabolism, inflammation, angiogenesis, and circadian rhythm. To better understand the role of this nuclear receptor in liver, we aimed at displaying genes controlled by RORα in liver cells by generating HepG2 human hepatoma cells stably over-expressing RORα. Genes whose expression was altered in these cells versus control cells were displayed using micro-arrays followed by qRT-PCR analysis. Expression of these genes was also altered in cells in which RORα was transiently over-expressed after adenoviral infection. A number of the genes found were involved in known pathways controlled by RORα, for instance LPA, NR1D2 and ADIPOQ in lipid metabolism, ADIPOQ and PLG in inflammation, PLG in fibrinolysis and NR1D2 and NR1D1 in circadian rhythm. This study also revealed that genes such as G6PC, involved in glucose homeostasis, and AGRP, involved in the control of body weight, are also controlled by RORα. Lastly, SPARC, involved in cell growth and adhesion, and associated with liver carcinogenesis, was up-regulated by RORα. SPARC was found to be a new putative RORα target gene since it possesses, in its promoter, a functional RORE as evidenced by EMSAs and transfection experiments. Most of the other genes that we found regulated by RORα also contained putative ROREs in their regulatory regions. Chromatin immunoprecipitation (ChIP) confirmed that the ROREs present in the SPARC, PLG, G6PC, NR1D2 and AGRP genes were occupied by RORα in HepG2 cells. Therefore these genes must now be considered as direct RORα targets. Our results open new routes on the roles of RORα in glucose metabolism and carcinogenesis within cells of hepatic origin.


Asunto(s)
Carcinoma Hepatocelular/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Hepáticas/genética , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Adenoviridae/metabolismo , Secuencia de Bases , Inmunoprecipitación de Cromatina , Células Hep G2 , Humanos , Datos de Secuencia Molecular , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Osteonectina/genética , Unión Proteica , Elementos de Respuesta/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética , Regulación hacia Arriba/genética
13.
Gastroenterology ; 140(5): 1564-74, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21300064

RESUMEN

BACKGROUND & AIMS: Glucagon-like peptide (GLP)-1, an intestinal incretin produced by L cells through proglucagon processing, is secreted after nutrient ingestion and acts on endocrine pancreas beta cells to enhance insulin secretion. Peroxisome proliferator-activated receptor (PPAR) ß/δ is a nuclear receptor that improves glucose homeostasis and pancreas islet function in diabetic animal models. Here, we investigated whether PPARß/δ activation regulates L cell GLP-1 production. METHODS: Proglucagon regulation and GLP-1 release were evaluated in murine GLUTag and human NCI-H716 L cells and in vivo using wild-type, PPARß/δ-null, and ob/ob C57Bl/6 mice treated with the PPARß/δ synthetic agonists GW501516 or GW0742. RESULTS: PPARß/δ activation increased proglucagon expression and enhanced glucose- and bile acid-induced GLP-1 release by intestinal L cells in vitro and ex vivo in human jejunum. In vivo treatment with GW0742 increased proglucagon messenger RNA levels in the small intestine in wild-type but not in PPARß/δ-deficient mice. Treatment of wild-type and ob/ob mice with GW501516 enhanced the increase in plasma GLP-1 level after an oral glucose load and improved glucose tolerance. Concomitantly, proglucagon and GLP-1 receptor messenger RNA levels increased in the small intestine and pancreas, respectively. Finally, PPARß/δ agonists activate the proglucagon gene transcription by interfering with the ß-catenin/TCF-4 pathway. CONCLUSIONS: Our data show that PPARß/δ activation potentiates GLP-1 production by the small intestine. Pharmacologic targeting of PPARß/δ is a promising approach in the treatment of patients with type 2 diabetes mellitus, especially in combination with dipeptidyl peptidase IV inhibitors.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Células Enteroendocrinas/metabolismo , Regulación de la Expresión Génica , Péptido 1 Similar al Glucagón/biosíntesis , PPAR-beta/metabolismo , ARN Mensajero/genética , Animales , Glucemia/metabolismo , Western Blotting , Células Cultivadas , Diabetes Mellitus Experimental/patología , Modelos Animales de Enfermedad , Células Enteroendocrinas/patología , Péptido 1 Similar al Glucagón/genética , Humanos , Masculino , Ratones , Reacción en Cadena de la Polimerasa , ARN Mensajero/biosíntesis , Ratas
14.
J Biol Chem ; 285(47): 36759-67, 2010 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-20851881

RESUMEN

The bile acid receptor farnesoid X receptor (FXR) is expressed in adipose tissue, but its function remains poorly defined. Peroxisome proliferator-activated receptor-γ (PPARγ) is a master regulator of adipocyte differentiation and function. The aim of this study was to analyze the role of FXR in adipocyte function and to assess whether it modulates PPARγ action. Therefore, we tested the responsiveness of FXR-deficient mice (FXR(-/-)) and cells to the PPARγ activator rosiglitazone. Our results show that genetically obese FXR(-/-)/ob/ob mice displayed a resistance to rosiglitazone treatment. In vitro, rosiglitazone treatment did not induce normal adipocyte differentiation and lipid droplet formation in FXR(-/-) mouse embryonic fibroblasts (MEFs) and preadipocytes. Moreover, FXR(-/-) MEFs displayed both an increased lipolysis and a decreased de novo lipogenesis, resulting in reduced intracellular triglyceride content, even upon PPARγ activation. Retroviral-mediated FXR re-expression in FXR(-/-) MEFs restored the induction of adipogenic marker genes during rosiglitazone-forced adipocyte differentiation. The expression of Wnt/ß-catenin pathway and target genes was increased in FXR(-/-) adipose tissue and MEFs. Moreover, the expression of several endogenous inhibitors of this pathway was decreased early during the adipocyte differentiation of FXR(-/-) MEFs. These findings demonstrate that FXR regulates adipocyte differentiation and function by regulating two counteracting pathways of adipocyte differentiation, the PPARγ and Wnt/ß-catenin pathways.


Asunto(s)
Adipocitos/citología , Diferenciación Celular , PPAR gamma/metabolismo , Receptores Citoplasmáticos y Nucleares/fisiología , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Animales , Biomarcadores/metabolismo , Western Blotting , Células Cultivadas , Resistencia a Medicamentos , Embrión de Mamíferos/citología , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/metabolismo , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Proteína-1 Reguladora de Fusión , Perfilación de la Expresión Génica , Humanos , Hipoglucemiantes/farmacología , Riñón/citología , Riñón/efectos de los fármacos , Riñón/metabolismo , Lipólisis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Análisis de Secuencia por Matrices de Oligonucleótidos , PPAR gamma/genética , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rosiglitazona , Transducción de Señal , Tiazolidinedionas/farmacología , Proteínas Wnt/genética , beta Catenina/genética
15.
FEBS J ; 277(16): 3308-20, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20608974

RESUMEN

Obesity is a low-grade chronic inflammatory disease associated with an increased number of macrophages (adipose tissue macrophages) in adipose tissue. Within the adipose tissue, adipose tissue macrophages are the major source of visfatin/pre-B-cell colony-enhancing factor/nicotinamide phosphoribosyl transferase. The nuclear receptor peroxisome proliferator-activated receptor gamma (PPARgamma) exerts anti-inflammatory effects in macrophages by inhibiting cytokine production and enhancing alternative differentiation. In this study, we investigated whether PPARgamma modulates visfatin expression in murine (bone marrow-derived macrophage) and human (primary human resting macrophage, classical macrophage, alternative macrophage or adipose tissue macrophage) macrophage models and pre-adipocyte-derived adipocytes. We show that synthetic PPARgamma ligands increase visfatin gene expression in a PPARgamma-dependent manner in primary human resting macrophages and in adipose tissue macrophages, but not in adipocytes. The threefold increase of visfatin mRNA was paralleled by an increase of protein expression (30%) and secretion (30%). Electrophoretic mobility shift assay experiments and transient transfection assays indicated that PPARgamma induces visfatin promoter activity in human macrophages by binding to a DR1-PPARgamma response element. Finally, we show that PPARgamma ligands increase NAD(+) production in primary human macrophages and that this regulation is dampened in the presence of visfatin small interfering RNA or by the visfatin-specific inhibitor FK866. Taken together, our results suggest that PPARgamma regulates the expression of visfatin in macrophages, leading to increased levels of NAD(+).


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Leucocitos Mononucleares/enzimología , Nicotinamida Fosforribosiltransferasa/metabolismo , PPAR gamma/metabolismo , Animales , Secuencia de Bases , Western Blotting , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Ensayo de Inmunoadsorción Enzimática , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , NAD/química , Nicotinamida Fosforribosiltransferasa/genética , PPAR gamma/agonistas , ARN Mensajero/metabolismo
16.
FEBS Lett ; 583(12): 2031-6, 2009 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-19450581

RESUMEN

Here we show that gene expression of the nuclear receptor RORalpha is induced during adipogenesis, with RORalpha4 being the most abundantly expressed isoform in human and murine adipose tissue. Over-expression of RORalpha4 in 3T3-L1 cells impairs adipogenesis as shown by the decreased expression of adipogenic markers and lipid accumulation, accompanied by decreased free fatty acid and glucose uptake. By contrast, mouse embryonic fibroblasts from staggerer mice, which carry a mutation in the RORalpha gene, differentiate more efficiently into mature adipocytes compared to wild-type cells, a phenotype which is reversed by ectopic RORalpha4 restoration.


Asunto(s)
Adipocitos/citología , Adipocitos/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Transactivadores/metabolismo , Células 3T3-L1 , Adipogénesis/genética , Adipogénesis/fisiología , Adulto , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Ácidos Grasos no Esterificados/metabolismo , Expresión Génica , Glucosa/metabolismo , Humanos , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes Neurológicos , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Citoplasmáticos y Nucleares/deficiencia , Receptores Citoplasmáticos y Nucleares/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transactivadores/deficiencia , Transactivadores/genética
17.
Gastroenterology ; 135(2): 689-98, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18565334

RESUMEN

BACKGROUND & AIMS: Conversion into bile acids represents an important route to remove excess cholesterol from the body. Rev-erbalpha is a nuclear receptor that participates as one of the clock genes in the control of circadian rhythmicity and plays a regulatory role in lipid metabolism and adipogenesis. Here, we investigate a potential role for Rev-erbalpha in the control of bile acid metabolism via the regulation of the neutral bile acid synthesis pathway. METHODS: Bile acid synthesis and CYP7A1 gene expression were studied in vitro and in vivo in mice deficient for or over expressing Rev-erbalpha. RESULTS: Rev-erbalpha-deficient mice display a lower synthesis rate and an impaired excretion of bile acids into the bile and feces. Expression of CYP7A1, the rate-limiting enzyme of the neutral pathway, is decreased in livers of Rev-erbalpha-deficient mice, whereas adenovirus-mediated hepatic Rev-erbalpha overexpression induces its expression. Moreover, bile acid feeding resulted in a more pronounced suppression of hepatic CYP7A1 expression in Rev-erbalpha-deficient mice. Hepatic expression of E4BP4 and the orphan nuclear receptor small heterodimer partner (SHP), both negative regulators of CYP7A1 expression, is increased in Rev-erbalpha-deficient mice. Promoter analysis and chromatin immunoprecipitation experiments demonstrated that SHP and E4BP4 are direct Rev-erbalpha target genes. Finally, the circadian rhythms of liver CYP7A1, SHP, and E4BP4 messenger RNA levels were perturbed in Rev-erbalpha-deficient mice. CONCLUSIONS: These data identify a role for Rev-erbalpha in the regulatory loop of bile acid synthesis, likely acting by regulating both hepatic SHP and E4BP4 expression.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Proteínas de Unión al ADN/metabolismo , Hígado/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Bilis/metabolismo , Línea Celular Tumoral , Colesterol 7-alfa-Hidroxilasa/genética , Colesterol 7-alfa-Hidroxilasa/metabolismo , Ritmo Circadiano , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Heces/química , Regulación Enzimológica de la Expresión Génica , Humanos , Hígado/enzimología , Ratones , Ratones Noqueados , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Receptores Citoplasmáticos y Nucleares/deficiencia , Receptores Citoplasmáticos y Nucleares/genética , Factores de Tiempo , Transfección
18.
Mol Endocrinol ; 22(8): 1797-811, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18511497

RESUMEN

A role of the nuclear receptor Rev-erbalpha in the regulation of transcription pathways involving other nuclear receptors is emerging. Indeed, Rev-erbalpha is a negative regulator of transcription by binding to overlapping response elements shared with various nuclear receptors, including the peroxisome proliferator-activated receptors and the retinoid-related orphan receptor alpha (RORalpha). Here, we show that Rev-erbalpha is expressed in primary human macrophages and that its expression is induced by synthetic ligands for the liver X receptors (LXRs), which control cholesterol homeostasis, inflammation, and the immune response in macrophages. LXRalpha binds to a specific response element in the human Rev-erbalpha promoter, thus inducing Rev-erbalpha transcriptional expression. Interestingly, Rev-erbalpha does not influence basal or LXR-regulated cholesterol homeostasis. However, Rev-erbalpha overexpression represses the induction of toll-like receptor (TLR)-4 by LXR agonists, whereas Rev-erbalpha silencing by short interfering RNA results in enhanced TLR-4 expression upon LXR activation. Electrophoretic mobility shift, chromatin immunoprecipitation, and transient transfection experiments demonstrate that Rev-erbalpha represses human TLR-4 promoter activity by binding as a monomer to a RevRE site overlapping with the LXR response element site in the TLR-4 promoter. These data identify Rev-erbalpha as a new LXR target gene, inhibiting LXR-induction of TLR-4 in a negative transcriptional feedback loop, but not cholesterol homeostasis gene expression.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Retroalimentación Fisiológica , Macrófagos/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Secuencia de Bases , Células Cultivadas , Colesterol/metabolismo , Dimerización , Retroalimentación Fisiológica/efectos de los fármacos , Humanos , Lipopolisacáridos/farmacología , Receptores X del Hígado , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Modelos Genéticos , Datos de Secuencia Molecular , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares , Receptores Nucleares Huérfanos , Unión Proteica/efectos de los fármacos , Proteínas Represoras/metabolismo , Elementos de Respuesta/genética , Receptor alfa X Retinoide/metabolismo , Receptor Toll-Like 4/genética , Activación Transcripcional/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
19.
Arterioscler Thromb Vasc Biol ; 28(5): 932-9, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18292390

RESUMEN

OBJECTIVE: Macrophages play a central role in the immune response against infectious organisms. Once activated, macrophages secrete proinflammatory cytokines and chemokines. Interleukin (IL)-8 and related CXC chemokines play a role in the recruitment and activation of phagocytes acting through CXCR1 and CXCR2 receptors. The nuclear receptor peroxisome proliferator-activated receptor (PPAR) gamma exerts antiinflammatory properties in macrophages, by inhibiting cytokine and CC chemokine production. In this study, we investigated whether PPAR-gamma also plays a role in the regulation of the CXC chemokine pathway. METHODS AND RESULTS: Synthetic PPAR-gamma ligands increase CXCR2 but not CXCR1 gene expression in a PPAR-gamma-dependent manner in primary human macrophages in vitro and in atherosclerotic plaques in vivo. The increase of CXCR2 mRNA was paralleled by an increase in membrane protein expression. EMSA, ChIP, and transient transfection assays indicate that PPAR-gamma activates the CXCR2 promoter by binding to a PPAR response element (PPRE). Finally, human macrophages acquire responsiveness to the CXCR2 ligands (IL-8 and Grobeta), as measured by superoxide anion production, after induction of CXCR2 expression by PPAR-gamma ligands. CONCLUSIONS: Our results provide a novel mechanism via which PPAR-gamma can enhance the immune response in human macrophages.


Asunto(s)
Macrófagos/metabolismo , PPAR gamma/metabolismo , Receptores de Interleucina-8B/metabolismo , Animales , Aterosclerosis/metabolismo , Aterosclerosis/patología , Células COS , Quimiocina CXCL2/farmacología , Chlorocebus aethiops , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Interleucina-8/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/patología , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores de Interleucina-8B/genética , Transducción de Señal , Superóxidos/metabolismo
20.
Biochem J ; 407(3): 461-9, 2007 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-17655523

RESUMEN

FXR (farnesoid X receptor), a nuclear receptor activated by BAs (bile acids), is a key factor in the regulation of BA, lipid and carbohydrate metabolism. The recent development of synthetic FXR agonists and knockout mouse models has accelerated the discovery of FXR target genes. In the present study, we identify human fetuin-B as a novel FXR target gene. Treatment with FXR agonists increased fetuin-B expression in human primary hepatocytes and in the human hepatoma HepG2 cell line. In contrast, fetuin-B expression was not responsive to FXR agonist treatment in murine primary hepatocytes. Fetuin-B induction by FXR agonist was abolished upon FXR knockdown by siRNA (small interfering RNA). In addition to the previously described P1 promoter, we show that the human fetuin-B gene is also transcribed from an alternative promoter, termed P2. Transcription via the P2 promoter was induced by FXR agonist treatment, whereas P1 promoter activity was not sensitive to FXR agonist treatment. Two putative FXR-response elements [IR-1 (inverted repeat-1)] were identified in the region -1.6 kb upstream of the predicted P2 transcriptional start site. Both motifs bound FXR-RXR (retinoid X receptor) complexes in vitro and were activated by FXR in transient transfection reporter assays. Mutations in the IR-1 sites abolished FXR-RXR binding and activation. Taken together, these results identify human fetuin-B as a new FXR target gene in human hepatocytes.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Hepatocitos/metabolismo , Receptores Citoplasmáticos y Nucleares/fisiología , Factores de Transcripción/fisiología , Regulación hacia Arriba/fisiología , alfa-Fetoproteínas/biosíntesis , alfa-Fetoproteínas/genética , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular Tumoral , Proteínas de Unión al ADN/agonistas , Fetuína-B , Perfilación de la Expresión Génica , Humanos , Ratones , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Receptores Citoplasmáticos y Nucleares/agonistas , Factores de Transcripción/agonistas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA