Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Exp Med ; 220(4)2023 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-36787127

RESUMEN

Non-alcoholic steatohepatitis (NASH) is a global health concern without treatment. The challenge in finding effective therapies is due to the lack of good mouse models and the complexity of the disease, characterized by gene-environment interactions. We tested the susceptibility of seven mouse strains to develop NASH. The severity of the clinical phenotypes observed varied widely across strains. PWK/PhJ mice were the most prone to develop hepatic inflammation and the only strain to progress to NASH with extensive fibrosis, while CAST/EiJ mice were completely resistant. Levels of mitochondrial transcripts and proteins as well as mitochondrial function were robustly reduced specifically in the liver of PWK/PhJ mice, suggesting a central role of mitochondrial dysfunction in NASH progression. Importantly, the NASH gene expression profile of PWK/PhJ mice had the highest overlap with the human NASH signature. Our study exposes the limitations of using a single mouse genetic background in metabolic studies and describes a novel NASH mouse model with features of the human NASH.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Ratones , Humanos , Animales , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Ratones Endogámicos C57BL , Hígado/metabolismo , Cirrosis Hepática/metabolismo , Ratones Endogámicos , Mitocondrias/genética , Mitocondrias/metabolismo , Modelos Animales de Enfermedad
2.
JCI Insight ; 8(3)2023 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-36752209

RESUMEN

Acute kidney failure and chronic kidney disease are global health issues steadily rising in incidence and prevalence. Animal models on a single genetic background have so far failed to recapitulate the clinical presentation of human nephropathies. Here, we used a simple model of folic acid-induced kidney injury in 7 highly diverse mouse strains. We measured plasma and urine parameters, as well as renal histopathology and mRNA expression data, at 1, 2, and 6 weeks after injury, covering the early recovery and long-term remission. We observed an extensive strain-specific response ranging from complete resistance of the CAST/EiJ to high sensitivity of the C57BL/6J, DBA/2J, and PWK/PhJ strains. In susceptible strains, the severe early kidney injury was accompanied by the induction of mitochondrial stress response (MSR) genes and the attenuation of NAD+ synthesis pathways. This is associated with delayed healing and a prolonged inflammatory and adaptive immune response 6 weeks after insult, heralding a transition to chronic kidney disease. Through a thorough comparison of the transcriptomic response in mouse and human disease, we show that critical metabolic gene alterations were shared across species, and we highlight the PWK/PhJ strain as an emergent model of transition from acute kidney injury to chronic disease.


Asunto(s)
Lesión Renal Aguda , Insuficiencia Renal Crónica , Humanos , Ratones , Animales , Ratones Endogámicos C57BL , NAD , Ratones Endogámicos DBA
3.
Trends Endocrinol Metab ; 29(8): 560-570, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29866502

RESUMEN

Growth differentiation factor-15 (GDF15) is a circulating protein that has been implicated in multiple biological processes, including energy homeostasis, body weight regulation, and cachexia driven by cancer and chronic disease. The potential to target GDF15 in the treatment of energy-intake disorders, including obesity and anorexia, is an area of intense investigation, but has been limited by the lack of an identified receptor, signaling mechanism, and target tissue. GDNF family receptor α-like (GFRAL) was recently identified as the neuronal brainstem receptor responsible for mediating the anorectic actions of GDF15. Herein, we provide a brief overview of GDF15 biology with a focus on energy homeostasis, and highlight the implications of the recent receptor identification to this field and beyond.


Asunto(s)
Anorexia , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial , Factor 15 de Diferenciación de Crecimiento , Obesidad , Animales , Anorexia/tratamiento farmacológico , Anorexia/metabolismo , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Factor 15 de Diferenciación de Crecimiento/agonistas , Factor 15 de Diferenciación de Crecimiento/antagonistas & inhibidores , Factor 15 de Diferenciación de Crecimiento/metabolismo , Humanos , Obesidad/tratamiento farmacológico , Obesidad/metabolismo
4.
Nat Med ; 23(10): 1150-1157, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28846097

RESUMEN

Growth differentiation factor 15 (GDF15), a distant member of the transforming growth factor (TGF)-ß family, is a secreted protein that circulates as a 25-kDa dimer. In humans, elevated GDF15 correlates with weight loss, and the administration of GDF15 to mice with obesity reduces body weight, at least in part, by decreasing food intake. The mechanisms through which GDF15 reduces body weight remain poorly understood, because the cognate receptor for GDF15 is unknown. Here we show that recombinant GDF15 induces weight loss in mice fed a high-fat diet and in nonhuman primates with spontaneous obesity. Furthermore, we find that GDF15 binds with high affinity to GDNF family receptor α-like (GFRAL), a distant relative of receptors for a distinct class of the TGF-ß superfamily ligands. Gfral is expressed in neurons of the area postrema and nucleus of the solitary tract in mice and humans, and genetic deletion of the receptor abrogates the ability of GDF15 to decrease food intake and body weight in mice. In addition, diet-induced obesity and insulin resistance are exacerbated in GFRAL-deficient mice, suggesting a homeostatic role for this receptor in metabolism. Finally, we demonstrate that GDF15-induced cell signaling requires the interaction of GFRAL with the coreceptor RET. Our data identify GFRAL as a new regulator of body weight and as the bona fide receptor mediating the metabolic effects of GDF15, enabling a more comprehensive assessment of GDF15 as a potential pharmacotherapy for the treatment of obesity.


Asunto(s)
Ingestión de Alimentos/efectos de los fármacos , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Factor 15 de Diferenciación de Crecimiento/genética , Obesidad/metabolismo , Pérdida de Peso/efectos de los fármacos , Animales , Dieta Alta en Grasa , Ingestión de Alimentos/genética , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Factor 15 de Diferenciación de Crecimiento/metabolismo , Factor 15 de Diferenciación de Crecimiento/farmacología , Humanos , Macaca fascicularis , Ratones , Ratones Noqueados , Pérdida de Peso/genética
5.
J Clin Invest ; 127(4): 1451-1462, 2017 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-28240605

RESUMEN

Obesity causes insulin resistance, and PPARγ ligands such as rosiglitazone are insulin sensitizing, yet the mechanisms remain unclear. In C57BL/6 (B6) mice, obesity induced by a high-fat diet (HFD) has major effects on visceral epididymal adipose tissue (eWAT). Here, we report that HFD-induced obesity in B6 mice also altered the activity of gene regulatory elements and genome-wide occupancy of PPARγ. Rosiglitazone treatment restored insulin sensitivity in obese B6 mice, yet, surprisingly, had little effect on gene expression in eWAT. However, in subcutaneous inguinal fat (iWAT), rosiglitazone markedly induced molecular signatures of brown fat, including the key thermogenic gene Ucp1. Obesity-resistant 129S1/SvImJ mice (129 mice) displayed iWAT browning, even in the absence of rosiglitazone. The 129 Ucp1 locus had increased PPARγ binding and gene expression that were preserved in the iWAT of B6x129 F1-intercrossed mice, with an imbalance favoring the 129-derived alleles, demonstrating a cis-acting genetic difference. Thus, B6 mice have genetically defective Ucp1 expression in iWAT. However, when Ucp1 was activated by rosiglitazone, or by iWAT browning in cold-exposed or young mice, expression of the B6 version of Ucp1 was no longer defective relative to the 129 version, indicating epigenomic rescue. These results provide a framework for understanding how environmental influences like drugs can affect the epigenome and potentially rescue genetically determined disease phenotypes.


Asunto(s)
Epigénesis Genética , Obesidad/metabolismo , PPAR gamma/fisiología , Animales , Dieta Alta en Grasa/efectos adversos , Hipoglucemiantes/farmacología , Grasa Intraabdominal/metabolismo , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Unión Proteica , Elementos Reguladores de la Transcripción , Rosiglitazona , Grasa Subcutánea Abdominal/metabolismo , Tiazolidinedionas/farmacología , Activación Transcripcional , Transcriptoma , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
6.
EMBO Mol Med ; 6(9): 1124-32, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25007801

RESUMEN

Macrophages are key immune cells found in atherosclerotic plaques and critically shape atherosclerotic disease development. Targeting the functional repertoire of macrophages may hold novel approaches for future atherosclerosis management. Here, we describe a previously unrecognized role of the epigenomic enzyme Histone deacetylase 3 (Hdac3) in regulating the atherosclerotic phenotype of macrophages. Using conditional knockout mice, we found that myeloid Hdac3 deficiency promotes collagen deposition in atherosclerotic lesions and thus induces a stable plaque phenotype. Also, macrophages presented a switch to anti-inflammatory wound healing characteristics and showed improved lipid handling. The pro-fibrotic phenotype was directly linked to epigenetic regulation of the Tgfb1 locus upon Hdac3 deletion, driving smooth muscle cells to increased collagen production. Moreover, in humans, HDAC3 was the sole Hdac upregulated in ruptured atherosclerotic lesions, Hdac3 associated with inflammatory macrophages, and HDAC3 expression inversely correlated with pro-fibrotic TGFB1 expression. Collectively, we show that targeting the macrophage epigenome can improve atherosclerosis outcome and we identify Hdac3 as a potential novel therapeutic target in cardiovascular disease.


Asunto(s)
Aterosclerosis/genética , Histona Desacetilasas/fisiología , Macrófagos/fisiología , Acetilación , Animales , Aterosclerosis/inmunología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Colágeno/metabolismo , Epigénesis Genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Humanos , Metabolismo de los Lípidos/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo
7.
Nature ; 504(7478): 153-7, 2013 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-24185009

RESUMEN

The development and severity of inflammatory bowel diseases and other chronic inflammatory conditions can be influenced by host genetic and environmental factors, including signals derived from commensal bacteria. However, the mechanisms that integrate these diverse cues remain undefined. Here we demonstrate that mice with an intestinal epithelial cell (IEC)-specific deletion of the epigenome-modifying enzyme histone deacetylase 3 (HDAC3(ΔIEC) mice) exhibited extensive dysregulation of IEC-intrinsic gene expression, including decreased basal expression of genes associated with antimicrobial defence. Critically, conventionally housed HDAC3(ΔIEC) mice demonstrated loss of Paneth cells, impaired IEC function and alterations in the composition of intestinal commensal bacteria. In addition, HDAC3(ΔIEC) mice showed significantly increased susceptibility to intestinal damage and inflammation, indicating that epithelial expression of HDAC3 has a central role in maintaining intestinal homeostasis. Re-derivation of HDAC3(ΔIEC) mice into germ-free conditions revealed that dysregulated IEC gene expression, Paneth cell homeostasis and intestinal barrier function were largely restored in the absence of commensal bacteria. Although the specific mechanisms through which IEC-intrinsic HDAC3 expression regulates these complex phenotypes remain to be determined, these data indicate that HDAC3 is a critical factor that integrates commensal-bacteria-derived signals to calibrate epithelial cell responses required to establish normal host-commensal relationships and maintain intestinal homeostasis.


Asunto(s)
Regulación de la Expresión Génica , Histona Desacetilasas/metabolismo , Homeostasis , Mucosa Intestinal/enzimología , Intestinos/microbiología , Simbiosis , Adulto , Animales , Bacterias/genética , Colitis Ulcerosa/enzimología , Colitis Ulcerosa/genética , Colitis Ulcerosa/microbiología , Enfermedad de Crohn/enzimología , Enfermedad de Crohn/genética , Enfermedad de Crohn/microbiología , Femenino , Eliminación de Gen , Perfilación de la Expresión Génica , Histona Desacetilasas/genética , Humanos , Mucosa Intestinal/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Células de Paneth/citología , Células de Paneth/metabolismo , ARN Ribosómico 16S/genética , Transducción de Señal
8.
Mol Cell ; 52(6): 769-82, 2013 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-24268577

RESUMEN

Histone deacetylases (HDACs) are believed to regulate gene transcription by catalyzing deacetylation reactions. HDAC3 depletion in mouse liver upregulates lipogenic genes and results in severe hepatosteatosis. Here we show that pharmacologic HDAC inhibition in primary hepatocytes causes histone hyperacetylation but does not upregulate expression of HDAC3 target genes. Meanwhile, deacetylase-dead HDAC3 mutants can rescue hepatosteatosis and repress lipogenic genes expression in HDAC3-depleted mouse liver, demonstrating that histone acetylation is insufficient to activate gene transcription. Mutations abolishing interactions with the nuclear receptor corepressor (NCOR or SMRT) render HDAC3 nonfunctional in vivo. Additionally, liver-specific knockout of NCOR, but not SMRT, causes metabolic and transcriptomal alterations resembling those of mice without hepatic HDAC3, demonstrating that interaction with NCOR is essential for deacetylase-independent function of HDAC3. These findings highlight nonenzymatic roles of a major HDAC in transcriptional regulation in vivo and warrant reconsideration of the mechanism of action of HDAC inhibitors.


Asunto(s)
Hepatocitos/enzimología , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Metabolismo de los Lípidos , Hígado/enzimología , Co-Represor 1 de Receptor Nuclear/metabolismo , Transcripción Genética , Acetilación , Animales , Hígado Graso/enzimología , Hígado Graso/genética , Perfilación de la Expresión Génica/métodos , Genotipo , Células HEK293 , Hepatocitos/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/química , Histona Desacetilasas/deficiencia , Histona Desacetilasas/genética , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/genética , Hígado/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Modelos Moleculares , Mutación , Co-Represor 1 de Receptor Nuclear/genética , Co-Represor 2 de Receptor Nuclear/genética , Co-Represor 2 de Receptor Nuclear/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Conformación Proteica , Relación Estructura-Actividad , Transcripción Genética/efectos de los fármacos , Transfección
9.
J Mol Endocrinol ; 51(3): T115-40, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24096517

RESUMEN

The nuclear receptor superfamily includes many receptors, identified based on their similarity to steroid hormone receptors but without a known ligand. The study of how these receptors are diversely regulated to interact with genomic regions to control a plethora of biological processes has provided critical insight into development, physiology, and the molecular pathology of disease. Here we provide a compendium of these so-called orphan receptors and focus on what has been learned about their modes of action, physiological functions, and therapeutic promise.


Asunto(s)
Receptores Nucleares Huérfanos/metabolismo , Factores de Transcripción/metabolismo , Animales , Humanos
10.
Proc Natl Acad Sci U S A ; 110(46): 18656-61, 2013 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-24167256

RESUMEN

Adipose tissue is an important metabolic organ, the dysfunction of which is associated with the development of obesity, diabetes mellitus, and cardiovascular disease. The nuclear receptor peroxisome proliferator-activated receptor gamma (PPARγ) is considered the master regulator of adipocyte differentiation and function. Although its cell-autonomous role in adipogenesis has been clearly demonstrated in cell culture, previous fat-specific knockouts of the murine PPARγ gene did not demonstrate a dramatic phenotype in vivo. Here, using Adipoq-Cre mice to drive adipose-specific recombination, we report a unique fat-specific PPARγ knockout (PPARγ FKO) mouse model with almost no visible brown and white adipose tissue at age 3 mo. As a consequence, PPARγ FKO mice had hugely enlarged pancreatic islets, massive fatty livers, and dramatically elevated levels of blood glucose and serum insulin accompanied by extreme insulin resistance. PPARγ FKO mice also exhibited delayed hair coat formation associated with absence of dermal fat, disrupted mammary gland development with loss of mammary fat pads, and high bone mass with loss of bone marrow fat, indicating the critical roles of adipose PPARγ in these tissues. Together, our data reveal the necessity of fat PPARγ in adipose formation, whole-body metabolic homeostasis, and normal development of fat-containing tissues.


Asunto(s)
Adipocitos/metabolismo , Resistencia a la Insulina/genética , Obesidad/metabolismo , PPAR gamma/deficiencia , Animales , Compuestos Azo , Immunoblotting , Inmunohistoquímica , Resistencia a la Insulina/fisiología , Ratones , PPAR gamma/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Microtomografía por Rayos X
11.
Mol Endocrinol ; 27(1): 127-34, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23192980

RESUMEN

Adipose-specific gene deletion in mice is crucial in determining gene function in adipocyte homeostasis and the development of obesity. We noted 100% mortality when the Hdac3 gene was conditionally deleted using Fabp4-Cre mice, the most commonly used model of adipose-targeted Cre recombinase. However, this surprising result was not reproduced using other models of adipose targeting of Cre, including a novel Retn-Cre mouse. These findings underscore the need for caution when interpreting data obtained using Fabp4-Cre mice and should encourage the use of additional or alternative adipose-targeting Cre mouse models before drawing conclusions about in vivo adipocyte-specific functions.


Asunto(s)
Tejido Adiposo/enzimología , Modelos Animales de Enfermedad , Eliminación de Gen , Histona Desacetilasas/genética , Obesidad/enzimología , Tejido Adiposo/fisiopatología , Animales , Epidídimo/enzimología , Epidídimo/fisiopatología , Proteínas de Unión a Ácidos Grasos/genética , Genes Letales , Ingeniería Genética , Masculino , Ratones , Ratones Transgénicos , Obesidad/genética , Especificidad de Órganos , Fenotipo
12.
Proc Natl Acad Sci U S A ; 109(35): 14140-5, 2012 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-22891325

RESUMEN

Sustained Toll-like receptor (TLR) stimulation continuously activates antimicrobial genes but paradoxically represses inflammatory genes. This phenomenon, termed TLR tolerance, is essential for preventing fatal inflammatory conditions such as sepsis, but its underlying mechanisms are unclear. We report here that NF-κB binding nucleic acids of gene promoters are tolerogenic motifs, which selectively recruit an NcoR-Hdac3-deacetylated-p50 repressosome to inflammatory genes. Genome-wide analyses of TLR4-induced genes revealed that NF-κB motifs were the only regulatory elements significantly enriched in tolerizable genes. Mutating the NF-κB motifs of tolerizable genes converted them into nontolerizable ones, whereas inserting NF-κB binding motifs into nontolerizable genes conferred the tolerance. Although NF-κB p50 was essential for assembling the repressosome, genetic disruption of the NcoR-Hdac3 interaction alone was sufficient to completely abolish TLR4 tolerance and to render mice vulnerable to sepsis. Thus, the specificity of TLR tolerance is dictated by evolutionally conserved nucleic acid motifs that bound by NF-κB and the NcoR repressosome.


Asunto(s)
Tolerancia Inmunológica/inmunología , Subunidad p50 de NF-kappa B/inmunología , Co-Represor 1 de Receptor Nuclear/inmunología , Receptor Toll-Like 4/inmunología , Acetilación , Secuencias de Aminoácidos/inmunología , Animales , Células de la Médula Ósea/citología , Línea Celular , Expresión Génica/inmunología , Histona Desacetilasas/inmunología , Histona Desacetilasas/metabolismo , Tolerancia Inmunológica/genética , Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , Macrófagos/citología , Ratones , Ratones Endogámicos C57BL , Subunidad p50 de NF-kappa B/metabolismo , Co-Represor 1 de Receptor Nuclear/genética , Co-Represor 1 de Receptor Nuclear/metabolismo , Choque Séptico/inmunología , Choque Séptico/prevención & control , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
13.
Genes Dev ; 26(7): 657-67, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22474260

RESUMEN

The nuclear receptor Rev-erbα regulates circadian rhythm and metabolism, but its effects are modest and it has been considered to be a secondary regulator of the cell-autonomous clock. Here we report that depletion of Rev-erbα together with closely related Rev-erbß has dramatic effects on the cell-autonomous clock as well as hepatic lipid metabolism. Mouse embryonic fibroblasts were rendered arrhythmic by depletion of both Rev-erbs. In mouse livers, Rev-erbß mRNA and protein levels oscillate with a diurnal pattern similar to that of Rev-erbα, and both Rev-erbs are recruited to a remarkably similar set of binding sites across the genome, enriched near metabolic genes. Depletion of both Rev-erbs in liver synergistically derepresses several metabolic genes as well as genes that control the positive limb of the molecular clock. Moreover, deficiency of both Rev-erbs causes marked hepatic steatosis, in contrast to relatively subtle changes upon loss of either subtype alone. These findings establish the two Rev-erbs as major regulators of both clock function and metabolism, displaying a level of subtype collaboration that is unusual among nuclear receptors but common among core clock proteins, protecting the organism from major perturbations in circadian and metabolic physiology.


Asunto(s)
Ritmo Circadiano , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/genética , Receptores Citoplasmáticos y Nucleares/genética , Proteínas Represoras/genética , Animales , Células Cultivadas , Regulación de la Expresión Génica , Genoma , Histona Desacetilasas/metabolismo , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Co-Represor 1 de Receptor Nuclear/genética , Co-Represor 1 de Receptor Nuclear/metabolismo , 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/metabolismo , ARN Mensajero/genética , Receptores Citoplasmáticos y Nucleares/deficiencia , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteínas Represoras/deficiencia , Proteínas Represoras/metabolismo
14.
Genes Dev ; 25(23): 2480-8, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-22156208

RESUMEN

Macrophages, a key cellular component of inflammation, become functionally polarized in a signal- and context-specific manner. Th2 cytokines such as interleukin 4 (IL-4) polarize macrophages to a state of alternative activation that limits inflammation and promotes wound healing. Alternative activation is mediated by a transcriptional program that is influenced by epigenomic modifications, including histone acetylation. Here we report that macrophages lacking histone deacetylase 3 (HDAC3) display a polarization phenotype similar to IL-4-induced alternative activation and, furthermore, are hyperresponsive to IL-4 stimulation. Throughout the macrophage genome, HDAC3 deacetylates histone tails at regulatory regions, leading to repression of many IL-4-regulated genes characteristic of alternative activation. Following exposure to Schistosoma mansoni eggs, a model of Th2 cytokine-mediated disease that is limited by alternative activation, pulmonary inflammation was ameliorated in mice lacking HDAC3 in macrophages. Thus, HDAC3 functions in alternative activation as a brake whose release could be of benefit in the treatment of multiple inflammatory diseases.


Asunto(s)
Epigénesis Genética , Histona Desacetilasas/genética , Activación de Macrófagos/genética , Macrófagos/metabolismo , Animales , Histona Desacetilasas/metabolismo , Interleucina-4/genética , Interleucina-4/metabolismo , Macrófagos/inmunología , Ratones , Ratones Endogámicos , Neumonía/enzimología , Neumonía/inmunología , Neumonía/parasitología , Schistosoma mansoni , Células Th2/inmunología , Células Th2/metabolismo
15.
Circ Res ; 109(11): 1240-9, 2011 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-21959220

RESUMEN

RATIONALE: The development of the cardiac outflow tract (OFT) and great vessels is a complex process that involves coordinated regulation of multiple progenitor cell populations. Among these populations, neural crest cells make important contributions to OFT formation and aortic arch remodeling. Although numerous signaling pathways, including Notch, have been implicated in this process, the role of epigenetics in OFT development remains largely unexplored. OBJECTIVE: Because histone deacetylases (Hdacs) play important roles in the epigenetic regulation of mammalian development, we have investigated the function of Hdac3, a class I Hdac, during cardiac neural crest development in mouse. METHODS AND RESULTS: Using 2 neural crest drivers, Wnt1-Cre and Pax3(Cre), we show that loss of Hdac3 in neural crest results in perinatal lethality and cardiovascular abnormalities, including interrupted aortic arch type B, aortic arch hypoplasia, double-outlet right ventricle, and ventricular septal defect. Affected embryos are deficient in aortic arch artery smooth muscle during midgestation, despite intact neural crest cell migration and preserved development of other cardiac and truncal neural crest derivatives. The Hdac3-dependent block in smooth muscle differentiation is cell autonomous and is associated with downregulation of the Notch ligand Jagged1, a key driver of smooth muscle differentiation in the aortic arch arteries. CONCLUSIONS: These results indicate that Hdac3 plays a critical and specific regulatory role in the neural crest-derived smooth muscle lineage and in formation of the OFT.


Asunto(s)
Corazón Fetal/enzimología , Cardiopatías Congénitas/enzimología , Histona Desacetilasas/fisiología , Músculo Liso/patología , Cresta Neural/patología , Timo/anomalías , Médula Suprarrenal/embriología , Animales , Aorta Torácica/anomalías , Diferenciación Celular/fisiología , Linaje de la Célula , Movimiento Celular , Ventrículo Derecho con Doble Salida/embriología , Ventrículo Derecho con Doble Salida/enzimología , Ventrículo Derecho con Doble Salida/genética , Femenino , Corazón Fetal/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Cardiopatías Congénitas/embriología , Cardiopatías Congénitas/genética , Defectos del Tabique Interventricular/embriología , Defectos del Tabique Interventricular/enzimología , Defectos del Tabique Interventricular/genética , Ventrículos Cardíacos/embriología , Ventrículos Cardíacos/enzimología , Histona Desacetilasas/deficiencia , Histona Desacetilasas/genética , Masculino , Ratones , Ratones Transgénicos , Factor de Transcripción PAX3 , Factores de Transcripción Paired Box/fisiología , Receptores Notch/fisiología , Proteína Wnt1/fisiología
16.
J Biol Chem ; 286(38): 33301-9, 2011 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-21808063

RESUMEN

Many human diseases result from the influence of the nutritional environment on gene expression. The environment interacts with the genome by altering the epigenome, including covalent modification of nucleosomal histones. Here, we report a novel and dramatic influence of diet on the phenotype and survival of mice in which histone deacetylase 3 (Hdac3) is deleted postnatally in heart and skeletal muscle. Although embryonic deletion of myocardial Hdac3 causes major cardiomyopathy that reduces survival, we found that excision of Hdac3 in heart and muscle later in development leads to a much milder phenotype and does not reduce survival when mice are fed normal chow. Remarkably, upon switching to a high fat diet, the mice begin to die within weeks and display signs of severe hypertrophic cardiomyopathy and heart failure. Down-regulation of myocardial mitochondrial bioenergetic genes, specifically those involved in lipid metabolism, precedes the full development of cardiomyopathy, suggesting that HDAC3 is important in maintaining proper mitochondrial function. These data suggest that loss of the epigenomic modifier HDAC3 causes dietary lethality by compromising the ability of cardiac mitochondria to respond to changes of nutritional environment. In addition, this study provides a mouse model for diet-inducible heart failure.


Asunto(s)
Dieta/efectos adversos , Eliminación de Gen , Histona Desacetilasas/genética , Músculo Esquelético/enzimología , Músculo Esquelético/patología , Miocardio/enzimología , Miocardio/patología , Animales , Animales Recién Nacidos , Grasas de la Dieta/efectos adversos , Ecocardiografía , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Genes Mitocondriales/genética , Histona Desacetilasas/metabolismo , Humanos , Integrasas/metabolismo , Metabolismo de los Lípidos , Ratones , Ratones Noqueados , Músculo Esquelético/fisiopatología
17.
Science ; 331(6022): 1315-9, 2011 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-21393543

RESUMEN

Disruption of the circadian clock exacerbates metabolic diseases, including obesity and diabetes. We show that histone deacetylase 3 (HDAC3) recruitment to the genome displays a circadian rhythm in mouse liver. Histone acetylation is inversely related to HDAC3 binding, and this rhythm is lost when HDAC3 is absent. Although amounts of HDAC3 are constant, its genomic recruitment in liver corresponds to the expression pattern of the circadian nuclear receptor Rev-erbα. Rev-erbα colocalizes with HDAC3 near genes regulating lipid metabolism, and deletion of HDAC3 or Rev-erbα in mouse liver causes hepatic steatosis. Thus, genomic recruitment of HDAC3 by Rev-erbα directs a circadian rhythm of histone acetylation and gene expression required for normal hepatic lipid homeostasis.


Asunto(s)
Relojes Circadianos , Ritmo Circadiano , Hígado Graso/metabolismo , Genoma , Histona Desacetilasas/metabolismo , Metabolismo de los Lípidos , Hígado/metabolismo , Animales , Sitios de Unión , Inmunoprecipitación de Cromatina , Trastornos Cronobiológicos/genética , Trastornos Cronobiológicos/metabolismo , ADN/metabolismo , Epigénesis Genética , Regulación de la Expresión Génica , Histonas/metabolismo , Homeostasis , Lipogénesis/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Co-Represor 1 de Receptor Nuclear/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 , ARN Polimerasa II/metabolismo , Regulación hacia Arriba
18.
Blood ; 117(9): 2681-90, 2011 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-21205929

RESUMEN

The NR4A subfamily of nuclear receptors (NR4A1, NR4A2, and NR4A3) function as transcription factors that transduce diverse extracellular signals into altered gene transcription to coordinate apoptosis, proliferation, cell cycle arrest, and DNA repair. We previously discovered that 2 of these receptors, NR4A1 and NR4A3, are potent tumor suppressors of acute myeloid leukemia (AML); they are silenced in human AML, and abrogation of both genes in mice leads to rapid postnatal development of AML. Reduced expression of NR4As is also a common feature of myelodysplastic syndromes (MDSs). Here we show that reduced gene dosage of NR4A1 and NR4A3 in hypoallelic (NR4A1(+/-)NR4A3(-/-) or NR4A1(-/-)NR4A3(+/-)) mice below a critical threshold leads to a chronic myeloid malignancy that closely recapitulates the pathologic features of mixed myelodysplastic/myeloproliferative neoplasms (MDS/MPNs) with progression to AML in rare cases. Enhanced proliferation and excessive apoptosis of hematopoietic stem cells and myeloid progenitors, together with elevated DNA damage, contribute to MDS/MPN disease. We identify the myeloid tumor suppressor genes Egr1 and JunB and the DNA damage checkpoint kinase, polo-like kinase 2 (Plk2) as deregulated genes whose disrupted signaling probably contributes to MDS/MPN. These mice provide a novel model to elucidate the molecular pathogenesis of MDS/MPN and for therapeutic evaluation.


Asunto(s)
Proteínas de Unión al ADN/genética , Dosificación de Gen/genética , Síndromes Mielodisplásicos/genética , Trastornos Mieloproliferativos/genética , Proteínas del Tejido Nervioso/genética , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Receptores de Esteroides/genética , Receptores de Hormona Tiroidea/genética , Alelos , Animales , Apoptosis , Compartimento Celular , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Daño del ADN , Progresión de la Enfermedad , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Humanos , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos C57BL , Síndromes Mielodisplásicos/patología , Células Progenitoras Mieloides/patología , Trastornos Mieloproliferativos/patología , Fenotipo , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas
19.
J Neurosci ; 31(2): 764-74, 2011 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-21228185

RESUMEN

Gene expression is dynamically regulated by chromatin modifications on histone tails, such as acetylation. In general, histone acetylation promotes transcription, whereas histone deacetylation negatively regulates transcription. The interplay between histone acetyltranserases and histone deacetylases (HDACs) is pivotal for the regulation of gene expression required for long-term memory processes. Currently, very little is known about the role of individual HDACs in learning and memory. We examined the role of HDAC3 in long-term memory using a combined genetic and pharmacologic approach. We used HDAC3-FLOX genetically modified mice in combination with adeno-associated virus-expressing Cre recombinase to generate focal homozygous deletions of Hdac3 in area CA1 of the dorsal hippocampus. To complement this approach, we also used a selective inhibitor of HDAC3, RGFP136 [N-(6-(2-amino-4-fluorophenylamino)-6-oxohexyl)-4-methylbenzamide]. Immunohistochemistry showed that focal deletion or intrahippocampal delivery of RGFP136 resulted in increased histone acetylation. Both the focal deletion of HDAC3 as well as HDAC3 inhibition via RGFP136 significantly enhanced long-term memory in a persistent manner. Next we examined expression of genes implicated in long-term memory from dorsal hippocampal punches using quantitative reverse transcription-PCR. Expression of nuclear receptor subfamily 4 group A, member 2 (Nr4a2) and c-fos was significantly increased in the hippocampus of HDAC3-FLOX mice compared with wild-type controls. Memory enhancements observed in HDAC3-FLOX mice were abolished by intrahippocampal delivery of Nr4a2 small interfering RNA, suggesting a mechanism by which HDAC3 negatively regulates memory formation. Together, these findings demonstrate a critical role for HDAC3 in the molecular mechanisms underlying long-term memory formation.


Asunto(s)
Benzamidas/farmacología , Histona Desacetilasas/fisiología , Memoria a Largo Plazo/fisiología , Acetilación , Animales , Hipocampo/enzimología , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/biosíntesis , Histona Desacetilasas/genética , Histonas/metabolismo , Memoria a Largo Plazo/efectos de los fármacos , Ratones , Ratones Mutantes , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/biosíntesis , Eliminación de Secuencia , Percepción Espacial/efectos de los fármacos , Percepción Espacial/fisiología
20.
Mol Cell Biol ; 30(9): 2078-89, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20176806

RESUMEN

The nuclear receptor peroxisome proliferator activator receptor gamma (PPARgamma) is the target of antidiabetic thiazolidinedione drugs, which improve insulin resistance but have side effects that limit widespread use. PPARgamma is required for adipocyte differentiation, but it is also expressed in other cell types, notably macrophages, where it influences atherosclerosis, insulin resistance, and inflammation. A central question is whether PPARgamma binding in macrophages occurs at genomic locations the same as or different from those in adipocytes. Here, utilizing chromatin immunoprecipitation and high-throughput sequencing (ChIP-seq), we demonstrate that PPARgamma cistromes in mouse adipocytes and macrophages are predominantly cell type specific. In thioglycolate-elicited macrophages, PPARgamma colocalizes with the hematopoietic transcription factor PU.1 in areas of open chromatin and histone acetylation, near a distinct set of immune genes in addition to a number of metabolic genes shared with adipocytes. In adipocytes, the macrophage-unique binding regions are marked with repressive histone modifications, typically associated with local chromatin compaction and gene silencing. PPARgamma, when introduced into preadipocytes, bound only to regions depleted of repressive histone modifications, where it increased DNA accessibility, enhanced histone acetylation, and induced gene expression. Thus, the cell specificity of PPARgamma function is regulated by cell-specific transcription factors, chromatin accessibility, and histone marks. Our data support the existence of an epigenomic hierarchy in which PPARgamma binding to cell-specific sites not marked by repressive marks opens chromatin and leads to local activation marks, including histone acetylation.


Asunto(s)
Adipocitos/metabolismo , Macrófagos/metabolismo , Especificidad de Órganos , PPAR gamma/metabolismo , Células 3T3-L1 , Acetilación , Animales , Secuencia de Bases , Sitios de Unión , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Cromatina/metabolismo , ADN/metabolismo , Histonas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Unión Proteica , Transporte de Proteínas , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo , Activación Transcripcional/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...