Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Circulation ; 149(3): 227-250, 2024 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-37961903

RESUMEN

BACKGROUND: Cardiac metabolic dysfunction is a hallmark of heart failure (HF). Estrogen-related receptors ERRα and ERRγ are essential regulators of cardiac metabolism. Therefore, activation of ERR could be a potential therapeutic intervention for HF. However, in vivo studies demonstrating the potential usefulness of ERR agonist for HF treatment are lacking, because compounds with pharmacokinetics appropriate for in vivo use have not been available. METHODS: Using a structure-based design approach, we designed and synthesized 2 structurally distinct pan-ERR agonists, SLU-PP-332 and SLU-PP-915. We investigated the effect of ERR agonist on cardiac function in a pressure overload-induced HF model in vivo. We conducted comprehensive functional, multi-omics (RNA sequencing and metabolomics studies), and genetic dependency studies both in vivo and in vitro to dissect the molecular mechanism, ERR isoform dependency, and target specificity. RESULTS: Both SLU-PP-332 and SLU-PP-915 significantly improved ejection fraction, ameliorated fibrosis, and increased survival associated with pressure overload-induced HF without affecting cardiac hypertrophy. A broad spectrum of metabolic genes was transcriptionally activated by ERR agonists, particularly genes involved in fatty acid metabolism and mitochondrial function. Metabolomics analysis showed substantial normalization of metabolic profiles in fatty acid/lipid and tricarboxylic acid/oxidative phosphorylation metabolites in the mouse heart with 6-week pressure overload. ERR agonists increase mitochondria oxidative capacity and fatty acid use in vitro and in vivo. Using both in vitro and in vivo genetic dependency experiments, we show that ERRγ is the main mediator of ERR agonism-induced transcriptional regulation and cardioprotection and definitively demonstrated target specificity. ERR agonism also led to downregulation of cell cycle and development pathways, which was partially mediated by E2F1 in cardiomyocytes. CONCLUSIONS: ERR agonists maintain oxidative metabolism, which confers cardiac protection against pressure overload-induced HF in vivo. Our results provide direct pharmacologic evidence supporting the further development of ERR agonists as novel HF therapeutics.


Asunto(s)
Insuficiencia Cardíaca , Ratones , Animales , Cardiomegalia/metabolismo , Mitocondrias/metabolismo , Miocitos Cardíacos/metabolismo , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Ácidos Grasos/metabolismo
2.
Development ; 150(10)2023 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-37232416

RESUMEN

Cilia are essential for the ontogeny and function of many tissues, including the kidney. Here, we report that transcription factor ERRγ ortholog estrogen related receptor gamma a (Esrrγa) is essential for renal cell fate choice and ciliogenesis in zebrafish. esrrγa deficiency altered proximodistal nephron patterning, decreased the multiciliated cell populace and disrupted ciliogenesis in the nephron, Kupffer's vesicle and otic vesicle. These phenotypes were consistent with interruptions in prostaglandin signaling, and we found that ciliogenesis was rescued by PGE2 or the cyclooxygenase enzyme Ptgs1. Genetic interaction revealed that peroxisome proliferator-activated receptor gamma, coactivator 1 alpha (Ppargc1a), which acts upstream of Ptgs1-mediated prostaglandin synthesis, has a synergistic relationship with Esrrγa in the ciliogenic pathway. These ciliopathic phenotypes were also observed in mice lacking renal epithelial cell (REC) ERRγ, where significantly shorter cilia formed on proximal and distal tubule cells. Decreased cilia length preceded cyst formation in REC-ERRγ knockout mice, suggesting that ciliary changes occur early during pathogenesis. These data position Esrrγa as a novel link between ciliogenesis and nephrogenesis through regulation of prostaglandin signaling and cooperation with Ppargc1a.


Asunto(s)
Proteínas de Pez Cebra , Pez Cebra , Animales , Ratones , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Nefronas/metabolismo , Riñón/metabolismo , Prostaglandinas/metabolismo , Cilios/metabolismo
3.
Cell Metab ; 33(2): 379-394.e8, 2021 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-33301705

RESUMEN

Kidney disease is poorly understood because of the organ's cellular diversity. We used single-cell RNA sequencing not only in resolving differences in injured kidney tissue cellular composition but also in cell-type-specific gene expression in mouse models of kidney disease. This analysis highlighted major changes in cellular diversity in kidney disease, which markedly impacted whole-kidney transcriptomics outputs. Cell-type-specific differential expression analysis identified proximal tubule (PT) cells as the key vulnerable cell type. Through unbiased cell trajectory analyses, we show that PT cell differentiation is altered in kidney disease. Metabolism (fatty acid oxidation and oxidative phosphorylation) in PT cells showed the strongest and most reproducible association with PT cell differentiation and disease. Coupling of cell differentiation and the metabolism was established by nuclear receptors (estrogen-related receptor alpha [ESRRA] and peroxisomal proliferation-activated receptor alpha [PPARA]) that directly control metabolic and PT-cell-specific gene expression in mice and patient samples while protecting from kidney disease in the mouse model.


Asunto(s)
Enfermedades Renales/metabolismo , Receptores de Estrógenos/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Enfermedades Renales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Estrógenos/deficiencia , Receptor Relacionado con Estrógeno ERRalfa
4.
JACC Basic Transl Sci ; 5(9): 949-960, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33015416

RESUMEN

The heart plays a central role in the circulatory system and provides essential oxygen, nutrients, and growth factors to the whole organism. The heart can synthesize and secrete endocrine signals to communicate with distant target organs. Studies of long-known and recently discovered heart-derived hormones highlight a shared theme and reveal a unified mechanism of heart-derived hormones in coordinating cardiac function and target organ biology. This paper reviews the biochemistry, signaling, function, regulation, and clinical significance of representative heart-derived hormones, with a focus on the cardiovascular system. This review also discusses important and exciting questions that will advance the field of cardiac endocrinology.

5.
Cell Metab ; 30(4): 784-799.e5, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31474566

RESUMEN

Fibrosis is the final common pathway leading to end-stage renal failure. By analyzing the kidneys of patients and animal models with fibrosis, we observed a significant mitochondrial defect, including the loss of the mitochondrial transcription factor A (TFAM) in kidney tubule cells. Here, we generated mice with tubule-specific deletion of TFAM (Ksp-Cre/Tfamflox/flox). While these mice developed severe mitochondrial loss and energetic deficit by 6 weeks of age, kidney fibrosis, immune cell infiltration, and progressive azotemia causing death were only observed around 12 weeks of age. In renal cells of TFAM KO (knockout) mice, aberrant packaging of the mitochondrial DNA (mtDNA) resulted in its cytosolic translocation, activation of the cytosolic cGAS-stimulator of interferon genes (STING) DNA sensing pathway, and thus cytokine expression and immune cell recruitment. Ablation of STING ameliorated kidney fibrosis in mouse models of chronic kidney disease, demonstrating how TFAM sequesters mtDNA to limit the inflammation leading to fibrosis.


Asunto(s)
ADN Mitocondrial/metabolismo , Túbulos Renales/metabolismo , Proteínas de la Membrana/metabolismo , Mitocondrias/metabolismo , Insuficiencia Renal Crónica/patología , Animales , Proteínas de Unión al ADN/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Fibrosis , Humanos , Inflamación/patología , Túbulos Renales/patología , Masculino , Ratones , Proteínas Mitocondriales/metabolismo , Células RAW 264.7 , Factores de Transcripción/metabolismo
6.
Mol Cell Biol ; 38(21)2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30104250

RESUMEN

Growth differentiation factor 15 (GDF15) is a secreted protein with pleotropic functions from the transforming growth factor ß (TGF-ß) family. GDF15 is synthesized as a precursor and undergoes proteolytic cleavage to generate mature GDF15. The strong appetite-suppressing effect of mature GDF15 makes it an attractive therapeutic agent/target for diseases such as obesity and cachexia. In addition, clinical studies indicate that circulating, mature GDF15 is an independent biomarker for heart failure. We recently found that GDF15 functions as a heart-derived hormone that inhibits liver growth hormone signaling and postnatal body growth in the pediatric period. However, little is known about the mechanism of GDF15 maturation, in particular the enzymes that mediate GDF15 precursor cleavage. We investigated which candidate proteases can cleave GDF15 precursor and generate mature GDF15 in cardiomyocytes in vitro and mouse hearts in vivo We discovered that three members of the proprotein convertase, subtilisin/kexin-type (PCSK) family, namely, PCSK3, PCSK5, and PCSK6, can efficiently cleave GDF15 precursor, therefore licensing its maturation both in vitro and in vivo Our studies suggest that PCSK3, -5, and -6 mediate a crucial step of GDF15 maturation through proteolytic cleavage of the precursor. These results also reveal new targets for therapeutic application of GDF15 in treating obesity and cachexia.


Asunto(s)
Factor 15 de Diferenciación de Crecimiento/metabolismo , Proproteína Convertasas/metabolismo , Animales , Línea Celular , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos Cardíacos/metabolismo , Proteolisis , Serina Endopeptidasas/metabolismo , Transducción de Señal/fisiología , Subtilisinas/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
7.
Proc Natl Acad Sci U S A ; 115(21): E4910-E4919, 2018 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-29735694

RESUMEN

Mitochondrial dysfunction is increasingly recognized as a critical determinant of both hereditary and acquired kidney diseases. However, it remains poorly understood how mitochondrial metabolism is regulated to support normal kidney function and how its dysregulation contributes to kidney disease. Here, we show that the nuclear receptor estrogen-related receptor gamma (ERRγ) and hepatocyte nuclear factor 1 beta (HNF1ß) link renal mitochondrial and reabsorptive functions through coordinated epigenomic programs. ERRγ directly regulates mitochondrial metabolism but cooperatively controls renal reabsorption via convergent binding with HNF1ß. Deletion of ERRγ in renal epithelial cells (RECs), in which it is highly and specifically expressed, results in severe renal energetic and reabsorptive dysfunction and progressive renal failure that recapitulates phenotypes of animals and patients with HNF1ß loss-of-function gene mutations. Moreover, ERRγ expression positively correlates with renal function and is decreased in patients with chronic kidney disease (CKD). REC-ERRγ KO mice share highly overlapping renal transcriptional signatures with human patients with CKD. Together these findings reveal a role for ERRγ in directing independent and HNF1ß-integrated programs for energy production and use essential for normal renal function and the prevention of kidney disease.


Asunto(s)
Quistes/prevención & control , Metabolismo Energético , Epigenómica , Regulación de la Expresión Génica , Factor Nuclear 1-beta del Hepatocito/genética , Receptores de Estrógenos/genética , Insuficiencia Renal Crónica/prevención & control , Animales , Quistes/metabolismo , Quistes/patología , Factor Nuclear 1-beta del Hepatocito/metabolismo , Factor Nuclear 1-beta del Hepatocito/fisiología , Humanos , Riñón/metabolismo , Riñón/patología , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Mitocondrias/patología , Regiones Promotoras Genéticas , Receptores de Estrógenos/metabolismo , Receptores de Estrógenos/fisiología , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/patología
8.
Nature ; 546(7659): 544-548, 2017 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-28614293

RESUMEN

Brown adipose tissue is a thermogenic organ that dissipates chemical energy as heat to protect animals against hypothermia and to counteract metabolic disease. However, the transcriptional mechanisms that determine the thermogenic capacity of brown adipose tissue before environmental cold are unknown. Here we show that histone deacetylase 3 (HDAC3) is required to activate brown adipose tissue enhancers to ensure thermogenic aptitude. Mice with brown adipose tissue-specific genetic ablation of HDAC3 become severely hypothermic and succumb to acute cold exposure. Uncoupling protein 1 (UCP1) is nearly absent in brown adipose tissue lacking HDAC3, and there is also marked downregulation of mitochondrial oxidative phosphorylation genes resulting in diminished mitochondrial respiration. Remarkably, although HDAC3 acts canonically as a transcriptional corepressor, it functions as a coactivator of oestrogen-related receptor α (ERRα) in brown adipose tissue. HDAC3 coactivation of ERRα is mediated by deacetylation of PGC-1α and is required for the transcription of Ucp1, Ppargc1a (encoding PGC-1α), and oxidative phosphorylation genes. Importantly, HDAC3 promotes the basal transcription of these genes independently of adrenergic stimulation. Thus, HDAC3 uniquely primes Ucp1 and the thermogenic transcriptional program to maintain a critical capacity for thermogenesis in brown adipose tissue that can be rapidly engaged upon exposure to dangerously cold temperature.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Regulación de la Expresión Génica , Histona Desacetilasas/metabolismo , Termogénesis , Animales , Respiración de la Célula , Frío , Elementos de Facilitación Genéticos/genética , Calor , Humanos , Masculino , Ratones , Mitocondrias/metabolismo , Fosforilación Oxidativa , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Receptores de Estrógenos/metabolismo , Termogénesis/genética , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo , Receptor Relacionado con Estrógeno ERRalfa
9.
Cell Metab ; 21(4): 628-36, 2015 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-25863252

RESUMEN

Neurons utilize mitochondrial oxidative phosphorylation (OxPhos) to generate energy essential for survival, function, and behavioral output. Unlike most cells that burn both fat and sugar, neurons only burn sugar. Despite its importance, how neurons meet the increased energy demands of complex behaviors such as learning and memory is poorly understood. Here we show that the estrogen-related receptor gamma (ERRγ) orchestrates the expression of a distinct neural gene network promoting mitochondrial oxidative metabolism that reflects the extraordinary neuronal dependence on glucose. ERRγ(-/-) neurons exhibit decreased metabolic capacity. Impairment of long-term potentiation (LTP) in ERRγ(-/-) hippocampal slices can be fully rescued by the mitochondrial OxPhos substrate pyruvate, functionally linking the ERRγ knockout metabolic phenotype and memory formation. Consistent with this notion, mice lacking neuronal ERRγ in cerebral cortex and hippocampus exhibit defects in spatial learning and memory. These findings implicate neuronal ERRγ in the metabolic adaptations required for memory formation.


Asunto(s)
Hipocampo/fisiología , Potenciación a Largo Plazo/fisiología , Mitocondrias/metabolismo , Neuronas/metabolismo , Receptores de Estrógenos/metabolismo , Análisis de Varianza , Animales , Inmunoprecipitación de Cromatina , Galactósidos , Técnicas de Inactivación de Genes , Glucólisis/fisiología , Hipocampo/metabolismo , Indoles , Memoria/fisiología , Ratones , Análisis por Micromatrices , Ácido Pirúvico , Reacción en Cadena en Tiempo Real de la Polimerasa , Aprendizaje Espacial/fisiología
10.
Mol Cell Biol ; 35(7): 1281-98, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25624346

RESUMEN

Almost all cellular functions are powered by a continuous energy supply derived from cellular metabolism. However, it is little understood how cellular energy production is coordinated with diverse energy-consuming cellular functions. Here, using the cardiac muscle system, we demonstrate that nuclear receptors estrogen-related receptor α (ERRα) and ERRγ are essential transcriptional coordinators of cardiac energy production and consumption. On the one hand, ERRα and ERRγ together are vital for intact cardiomyocyte metabolism by directly controlling expression of genes important for mitochondrial functions and dynamics. On the other hand, ERRα and ERRγ influence major cardiomyocyte energy consumption functions through direct transcriptional regulation of key contraction, calcium homeostasis, and conduction genes. Mice lacking both ERRα and cardiac ERRγ develop severe bradycardia, lethal cardiomyopathy, and heart failure featuring metabolic, contractile, and conduction dysfunctions. These results illustrate that the ERR transcriptional pathway is essential to couple cellular energy metabolism with energy consumption processes in order to maintain normal cardiac function.


Asunto(s)
Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Miocardio/metabolismo , Miocardio/patología , Receptores de Estrógenos/metabolismo , Animales , Cardiomiopatías/genética , Cardiomiopatías/fisiopatología , Células Cultivadas , Femenino , Eliminación de Gen , Regulación de la Expresión Génica , Corazón/fisiología , Corazón/fisiopatología , Canales Iónicos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias Cardíacas/metabolismo , Mitocondrias Cardíacas/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Receptores de Estrógenos/genética , Receptor Relacionado con Estrógeno ERRalfa
11.
J Lipid Res ; 54(3): 806-815, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23288947

RESUMEN

The formation of the atherosclerotic lesion is a complex process influenced by an array of inflammatory and lipid metabolism pathways. We previously demonstrated that NR4A nuclear receptors are highly induced in macrophages in response to inflammatory stimuli and modulate the expression of genes linked to inflammation in vitro. Here we used mouse genetic models to assess the impact of NR4A expression on atherosclerosis development and macrophage polarization. Transplantation of wild-type, Nur77⁻/⁻, or Nor1⁻/⁻ null hematopoetic precursors into LDL receptor (LDLR)⁻/⁻ recipient mice led to comparable development of atherosclerotic lesions after high-cholesterol diet. We also observed comparable induction of genes linked to M1 and M2 responses in wild-type and Nur77-null macrophages in response to lipopolysaccharides and interleukin (IL)-4, respectively. In contrast, activation of the nuclear receptor liver X receptor (LXR) strongly suppressed M1 responses, and ablation of signal transductor and activator of transcription 6 (STAT6) strongly suppressed M2 responses. Recent studies have suggested that alterations in levels of Ly6C(lo) monocytes may be a contributor to inflammation and atherosclerosis. In our study, loss of Nur77, but not Nor1, was associated with decreased abundance of Ly6C(lo) monocytes, but this change was not correlated with atherosclerotic lesion development. Collectively, our results suggest that alterations in the Ly6C(lo) monocyte population and bone marrow NR4A expression do not play dominant roles in macrophage polarization or the development of atherosclerosis in mice.


Asunto(s)
Aterosclerosis/metabolismo , Macrófagos/metabolismo , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Animales , Aterosclerosis/genética , Citometría de Flujo , Masculino , Ratones , Ratones Noqueados , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
12.
Hum Mol Genet ; 20(5): 917-26, 2011 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-21138943

RESUMEN

Congenital anomalies of the kidney and urinary tract (CAKUTs) are common disorders of human development affecting the renal parechyma, renal pelvis, ureter, bladder and urethra; they show evidence of shared genetic aetiology, although the molecular basis of this remains unknown in the majority of cases. Breakpoint mapping of a de novo, apparently balanced, reciprocal translocation associated with bilateral renal agenesis has implicated the gene encoding the nuclear steroid hormone receptor ESRRG as a candidate gene for CAKUT. Here we show that the Esrrg protein is detected throughout early ureteric ducts as cytoplasmic/sub-membranous staining; with nuclear localization seen in developing nephrons. In 14.5-16.5 dpc (days post-conception) mouse embryos, Esrrg localizes to the subset of ductal tissue within the kidney, liver and lung. The renal ductal expression becomes localized to renal papilla by 18.5 dpc. Perturbation of function was performed in embryonic mouse kidney culture using pooled siRNA to induce knock-down and a specific small-molecule agonist to induce aberrant activation of Esrrg. Both resulted in severe abnormality of early branching events of the ureteric duct. Mouse embryos with a targeted inactivation of Esrrg on both alleles (Esrrg(-/-)) showed agenesis of the renal papilla but normal development of the cortex and remaining medulla. Taken together, these results suggest that Esrrg is required for early branching events of the ureteric duct that occur prior to the onset of nephrogenesis. These findings confirm ESRRG as a strong candidate gene for CAKUT.


Asunto(s)
Médula Renal/embriología , Receptores de Estrógenos/metabolismo , Uréter/embriología , Uréter/metabolismo , Animales , Anomalías Congénitas/embriología , Anomalías Congénitas/genética , Anomalías Congénitas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Riñón/anomalías , Riñón/embriología , Riñón/metabolismo , Enfermedades Renales/congénito , Médula Renal/metabolismo , Ratones , Ratones Noqueados , Organogénesis , Receptores de Estrógenos/genética
13.
Mol Endocrinol ; 24(2): 299-309, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19965931

RESUMEN

Energy production by oxidative metabolism in kidney, stomach, and heart, is primarily expended in establishing ion gradients to drive renal electrolyte homeostasis, gastric acid secretion, and cardiac muscle contraction, respectively. In addition to orchestrating transcriptional control of oxidative metabolism, the orphan nuclear receptor, estrogen-related receptor gamma (ERRgamma), coordinates expression of genes central to ion homeostasis in oxidative tissues. Renal, gastric, and cardiac tissues subjected to genomic analysis of expression in perinatal ERRgamma null mice revealed a characteristic dysregulation of genes involved in transport processes, exemplified by the voltage-gated potassium channel, Kcne2. Consistently, ERRgamma null animals die during the first 72 h of life with elevated serum potassium, reductions in key gastric acid production markers, and cardiac arrhythmia with prolonged QT intervals. In addition, we find altered expression of several genes associated with hypertension in ERRgamma null mice. These findings suggest a potential role for genetic polymorphisms at the ERRgamma locus and ERRgamma modulators in the etiology and treatment of renal, gastric, and cardiac dysfunction.


Asunto(s)
Mucosa Gástrica/metabolismo , Regulación de la Expresión Génica , Corazón/fisiología , Riñón/metabolismo , Miocardio/metabolismo , Potasio/metabolismo , Receptores de Estrógenos/fisiología , Adulto , Animales , Animales Recién Nacidos , Índice de Masa Corporal , Femenino , Estudios de Asociación Genética , Homeostasis , Humanos , Hipertensión/genética , Riñón/patología , Síndrome de QT Prolongado/genética , Masculino , Ratones , Persona de Mediana Edad , Miocardio/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Especificidad de Órganos , Polimorfismo de Nucleótido Simple , Canales de Potasio con Entrada de Voltaje/genética , Canales de Potasio con Entrada de Voltaje/metabolismo , Receptores de Estrógenos/deficiencia , Receptores de Estrógenos/genética , Estómago/patología
14.
Cell Metab ; 9(6): 483-4, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19490901

RESUMEN

In this issue of Cell Metabolism, Liao and colleagues have applied a novel synthetic approach to fight obesity in animals by engineering the plant glyoxylate shunt in vivo (Dean et al., 2009). The idea of introducing an entirely foreign metabolic pathway opens up opportunities for understanding metabolism and creating novel potential therapeutic approaches.


Asunto(s)
Ácidos Grasos/metabolismo , Acetilcoenzima A/metabolismo , Línea Celular Tumoral , Gluconeogénesis , Glioxilatos/metabolismo , Humanos , Isocitratoliasa/genética , Isocitratoliasa/metabolismo , Malato Sintasa/genética , Malato Sintasa/metabolismo , Palmitatos/metabolismo
15.
Proc Natl Acad Sci U S A ; 105(50): 20021-6, 2008 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-19066220

RESUMEN

The nuclear receptor corepressor, silencing mediator of retinoid and thyroid hormone receptors (SMRT), is recruited by a plethora of transcription factors to mediate lineage and signal-dependent transcriptional repression. We generated a knockin mutation in the receptor interaction domain (RID) of SMRT (SMRT(mRID)) that solely disrupts its interaction with nuclear hormone receptors (NHRs). SMRT(mRID) mice are viable and exhibit no gross developmental abnormalities, demonstrating that the reported lethality of SMRT knockouts is determined by non-NHR transcription factors. However, SMRT(mRID) mice exhibit widespread metabolic defects including reduced respiration, altered insulin sensitivity, and 70% increased adiposity. The latter phenotype is illustrated by the observation that SMRT(mRID)-derived MEFs display a dramatically increased adipogenic capacity and accelerated differentiation rate. Collectively, our results demonstrate that SMRT-RID-dependent repression is a key determinant of the adipogenic set point as well as an integrator of glucose metabolism and whole-body metabolic homeostasis.


Asunto(s)
Adipogénesis/genética , Proteínas de Unión al ADN/metabolismo , Proteínas Represoras/metabolismo , Receptores alfa de Hormona Tiroidea/genética , Receptores beta de Hormona Tiroidea/genética , Animales , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/genética , Regulación hacia Abajo , Regulación de la Expresión Génica , Técnicas de Sustitución del Gen , Genes Letales , Glucosa/metabolismo , Homeostasis/genética , Ratones , Ratones Mutantes , Co-Represor 2 de Receptor Nuclear , PPAR gamma/metabolismo , Estructura Terciaria de Proteína , Proteínas Represoras/genética , Hormonas Tiroideas/metabolismo
16.
Arterioscler Thromb Vasc Biol ; 26(10): 2301-7, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16888235

RESUMEN

OBJECTIVE: The lungs of Abcg1-/- mice accumulate macrophage foam cells that contain high levels of unesterified and esterified cholesterol, consistent with a role for ABCG1 in facilitating the efflux of cholesterol from macrophages to high-density lipoprotein (HDL) and other exogenous sterol acceptors. Based on these observations, we investigated whether loss of ABCG1 affects foam cell deposition in the artery wall and the development of atherosclerosis. METHODS AND RESULTS: Bone marrow from wild-type or Abcg1-/- mice was transplanted into Ldlr-/- or ApoE-/- mice. After administration of a high-fat/high-cholesterol diet, plasma and tissue lipid levels and atherosclerotic lesion size were quantified and compared. Surprisingly, transplantation of Abcg1-/- bone marrow cells resulted in a significant reduction in lesion size in both mouse models, despite the fact that lipid levels increased in the lung, spleen, and kidney. Lesions of Ldlr-/- mice transplanted with Abcg1-/- cells contained increased numbers of apoptotic cells. Consistent with this observation, in vitro studies demonstrated that Abcg1-/- macrophages were more susceptible to oxidized low-density lipoprotein (ox-LDL)-dependent apoptosis than Abcg1+/+ cells. CONCLUSIONS: Diet-induced atherosclerosis is impaired when atherosclerotic-susceptible mice are transplanted with Abcg1-/- bone marrow. The demonstration that Abcg1-/- macrophages undergo accelerated apoptosis provides a mechanism to explain the decrease in the atherosclerotic lesions.


Asunto(s)
Apolipoproteínas E/deficiencia , Aterosclerosis/prevención & control , Trasplante de Médula Ósea , Médula Ósea/metabolismo , Hiperlipidemias/metabolismo , Lipoproteínas/deficiencia , Receptores de LDL/deficiencia , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1 , Transportadoras de Casetes de Unión a ATP/genética , Animales , Apoptosis , Aterosclerosis/etiología , Grasas de la Dieta/administración & dosificación , Femenino , Silenciador del Gen , Hiperlipidemias/fisiopatología , Hiperlipidemias/cirugía , Lipoproteínas/genética , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
17.
Nat Med ; 12(9): 1048-55, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16906154

RESUMEN

Hepatic glucose production is crucial for glucose homeostasis, and its dysregulation contributes to the pathogenesis of diabetes. Here, we show that members of the NR4A family of ligand-independent orphan nuclear receptors are downstream mediators of cAMP action in the hormonal control of gluconeogenesis. Hepatic expression of Nur77, Nurr1 and NOR1 is induced by the cAMP axis in response to glucagon and fasting in vivo and is increased in diabetic mice that exhibit elevated gluconeogenesis. Adenoviral expression of Nur77 induces genes involved in gluconeogenesis, stimulates glucose production both in vitro and in vivo, and raises blood glucose levels. Conversely, expression of an inhibitory mutant Nur77 receptor antagonizes gluconeogenic gene expression and lowers blood glucose levels in db/db mice. These results outline a previously unrecognized role for orphan nuclear receptors in the transcriptional control of glucose homeostasis.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Glucosa/metabolismo , Hígado/metabolismo , Receptores Citoplasmáticos y Nucleares/fisiología , Receptores de Esteroides/fisiología , Factores de Transcripción/fisiología , 8-Bromo Monofosfato de Adenosina Cíclica/farmacología , Animales , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Tipo 1/fisiopatología , Diabetes Mellitus Tipo 2/fisiopatología , Glucagón/farmacología , Gluconeogénesis/efectos de los fármacos , Humanos , Hiperglucemia/etiología , Masculino , Ratones , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares , Receptores Citoplasmáticos y Nucleares/genética , Receptores de Esteroides/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
18.
Mol Endocrinol ; 20(4): 786-94, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16339277

RESUMEN

Members of the nuclear hormone receptor superfamily have emerged as important regulators of macrophage gene expression in inflammation and disease. Previous studies have shown that the lipid-activated receptors peroxisomal proliferator-activated receptor and liver X receptor inhibit nuclear factor-kappaB (NF-kappaB) signaling and inflammatory gene expression. We recently identified the NR4A subfamily of orphan nuclear receptors (Nur77/NR4A1, Nurr1/NR4A2, and NOR1/NR4A3) as lipopolysaccharide- and NF-kappaB-responsive genes in macrophages. However, the role of these transcription factors in macrophage gene expression is unknown. We demonstrate here that, in contrast to peroxisomal proliferator-activated receptor and liver X receptor, the role of NR4A receptors in macrophages is proinflammatory. Retroviral expression of Nur77 in macrophages leads to the transcriptional activation of multiple genes involved in inflammation, apoptosis, and cell cycle control. One particularly interesting Nur77-responsive gene is the inducible kinase IKKi/IKKepsilon, an important component of the NF-kappaB signaling pathway. The IKKi promoter contains a functional NR4A binding site and is activated by all three NR4A receptors in transient transfection assays. Consistent with the activation of IKKi, expression of Nur77 in macrophages potentiates the induction of inflammatory gene expression in response to lipopolysaccharide. These results identify a new role for NR4A orphan nuclear receptors in the control of macrophage gene expression during inflammation.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Inflamación/genética , Inflamación/metabolismo , Macrófagos/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Animales , Línea Celular , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Quinasa I-kappa B , Ratones , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal , Transfección
19.
J Biol Chem ; 280(32): 29256-62, 2005 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-15964844

RESUMEN

Oxidized lipids and inflammatory cytokines are believed to play a causal role in atherosclerosis through the regulation of gene expression in macrophages and other cells. Previous work has implicated the nuclear receptors peroxisome proliferator-activated receptor and liver X receptor in the control of lipid-dependent gene expression and inflammation. Here we demonstrate that expression of a third group of nuclear receptors, the NR4A ligand-independent orphan receptors, is highly inducible in macrophages by diverse inflammatory stimuli. Treatment of macrophages with lipopolysaccharide (LPS), cytokines, or oxidized lipids triggers the transcriptional induction of Nur77 (NR4A1), Nurr1 (NR4A2), and NOR1 (NR4A3) expression. Several lines of evidence point to the NF-kappaB signaling pathway as a principal mediator of inducible NR4A expression in macrophages. Analysis of the murine and human Nur77 promoters revealed two highly conserved NF-kappaB response elements. Mutation of these elements inhibited LPS-dependent expression of the Nur77 promoter in transient transfection assays. Furthermore, induction of Nur77 expression by LPS was severely compromised in fibroblasts lacking the three NF-kappaB subunits, Nfkb1, c-Rel, and RelA. Consistent with its ability to be induced by oxidized lipids, Nur77 was expressed in macrophages within human atherosclerotic lesions. These results identified NR4A nuclear receptors as potential transcriptional mediators of inflammatory signals in activated macrophages.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas de Unión al ADN/biosíntesis , Macrófagos/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Receptores Citoplasmáticos y Nucleares/biosíntesis , Receptores de Esteroides/biosíntesis , Receptores de Hormona Tiroidea/biosíntesis , Factores de Transcripción/biosíntesis , Animales , Arteriosclerosis , Línea Celular , Secuencia Conservada , Citoplasma/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Eliminación de Gen , Humanos , Inmunohistoquímica , Inflamación , Ligandos , Metabolismo de los Lípidos , Lipopolisacáridos/metabolismo , Receptores X del Hígado , Ratones , Mutación , FN-kappa B/metabolismo , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares , Receptores Nucleares Huérfanos , Oxígeno/metabolismo , Regiones Promotoras Genéticas , ARN/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de Esteroides/genética , Elementos de Respuesta , Transducción de Señal , Factores de Tiempo , Factores de Transcripción/genética , Transcripción Genética , Transfección
20.
Cell ; 119(2): 299-309, 2004 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-15479645

RESUMEN

The liver X receptors (LXRs) are nuclear receptors with established roles in the regulation of lipid metabolism. We now show that LXR signaling not only regulates macrophage cholesterol metabolism but also impacts antimicrobial responses. Mice lacking LXRs are highly susceptible to infection with the intracellular bacteria Listeria monocytogenes (LM). Bone marrow transplant studies point to altered macrophage function as the major determinant of susceptibility. LXR-null macrophages undergo accelerated apoptosis when challenged with LM and exhibit defective bacterial clearance in vivo. These defects result, at least in part, from loss of regulation of the antiapoptotic factor SPalpha, a direct target for regulation by LXRalpha. Expression of LXRalpha or SPalpha in macrophages inhibits apoptosis in the setting of LM infection. Our results demonstrate that LXR-dependent gene expression plays an unexpected role in innate immunity and suggest that common nuclear receptor pathways mediate macrophage responses to modified lipoproteins and intracellular pathogens.


Asunto(s)
Colesterol/metabolismo , Regulación de la Expresión Génica , Inmunidad Innata/fisiología , Macrófagos/inmunología , Receptores Citoplasmáticos y Nucleares/metabolismo , Transducción de Señal/fisiología , Animales , Trasplante de Médula Ósea , Supervivencia Celular , Células Cultivadas , Proteínas de Unión al ADN , Perfilación de la Expresión Génica , Listeria monocytogenes/metabolismo , Listeriosis/metabolismo , Receptores X del Hígado , Macrófagos/citología , Macrófagos/microbiología , Macrófagos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores Nucleares Huérfanos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Tasa de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA