Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Genes Dev ; 32(7-8): 497-511, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29674394

RESUMEN

The metabolite acetyl-coenzyme A (acetyl-CoA) is the required acetyl donor for lysine acetylation and thereby links metabolism, signaling, and epigenetics. Nutrient availability alters acetyl-CoA levels in cancer cells, correlating with changes in global histone acetylation and gene expression. However, the specific molecular mechanisms through which acetyl-CoA production impacts gene expression and its functional roles in promoting malignant phenotypes are poorly understood. Here, using histone H3 Lys27 acetylation (H3K27ac) ChIP-seq (chromatin immunoprecipitation [ChIP] coupled with next-generation sequencing) with normalization to an exogenous reference genome (ChIP-Rx), we found that changes in acetyl-CoA abundance trigger site-specific regulation of H3K27ac, correlating with gene expression as opposed to uniformly modulating this mark at all genes. Genes involved in integrin signaling and cell adhesion were identified as acetyl-CoA-responsive in glioblastoma cells, and we demonstrate that ATP citrate lyase (ACLY)-dependent acetyl-CoA production promotes cell migration and adhesion to the extracellular matrix. Mechanistically, the transcription factor NFAT1 (nuclear factor of activated T cells 1) was found to mediate acetyl-CoA-dependent gene regulation and cell adhesion. This occurs through modulation of Ca2+ signals, triggering NFAT1 nuclear translocation when acetyl-CoA is abundant. The findings of this study thus establish that acetyl-CoA impacts H3K27ac at specific loci, correlating with gene expression, and that expression of cell adhesion genes are driven by acetyl-CoA in part through activation of Ca2+-NFAT signaling.


Asunto(s)
Acetilcoenzima A/metabolismo , Señalización del Calcio , Adhesión Celular , Movimiento Celular , Glioblastoma/metabolismo , Factores de Transcripción NFATC/metabolismo , ATP Citrato (pro-S)-Liasa/metabolismo , Acetilación , Animales , Adhesión Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Glioblastoma/patología , Glucosa/metabolismo , Histonas/metabolismo , Ratones Desnudos
2.
Mol Cell ; 67(2): 252-265.e6, 2017 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-28689661

RESUMEN

While maintaining the integrity of the genome and sustaining bioenergetics are both fundamental functions of the cell, potential crosstalk between metabolic and DNA repair pathways is poorly understood. Since histone acetylation plays important roles in DNA repair and is sensitive to the availability of acetyl coenzyme A (acetyl-CoA), we investigated a role for metabolic regulation of histone acetylation during the DNA damage response. In this study, we report that nuclear ATP-citrate lyase (ACLY) is phosphorylated at S455 downstream of ataxia telangiectasia mutated (ATM) and AKT following DNA damage. ACLY facilitates histone acetylation at double-strand break (DSB) sites, impairing 53BP1 localization and enabling BRCA1 recruitment and DNA repair by homologous recombination. ACLY phosphorylation and nuclear localization are necessary for its role in promoting BRCA1 recruitment. Upon PARP inhibition, ACLY silencing promotes genomic instability and cell death. Thus, the spatial and temporal control of acetyl-CoA production by ACLY participates in the mechanism of DNA repair pathway choice.


Asunto(s)
ATP Citrato (pro-S)-Liasa/metabolismo , Acetilcoenzima A/metabolismo , Proteína BRCA1/metabolismo , Núcleo Celular/enzimología , Roturas del ADN de Doble Cadena , Reparación del ADN por Recombinación , Células A549 , ATP Citrato (pro-S)-Liasa/genética , Acetilación , Animales , Proteína BRCA1/genética , Núcleo Celular/efectos de los fármacos , Femenino , Puntos de Control de la Fase G2 del Ciclo Celular , Inestabilidad Genómica , Glucosa/metabolismo , Células HCT116 , Células HeLa , Histonas/metabolismo , Humanos , Melanoma Experimental/enzimología , Melanoma Experimental/genética , Melanoma Experimental/patología , Ratones Endogámicos C57BL , Fosforilación , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Unión Proteica , Procesamiento Proteico-Postraduccional , Interferencia de ARN , Reparación del ADN por Recombinación/efectos de los fármacos , Puntos de Control de la Fase S del Ciclo Celular , Serina , Factores de Tiempo , Transfección , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo
3.
J Biol Chem ; 289(24): 17078-86, 2014 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-24811171

RESUMEN

Glucocorticoids contribute to adipocyte differentiation by cooperating with transcription factors, such as CCAAT/enhancer-binding protein ß (C/EBPß), to stimulate transcription of the gene encoding peroxisome proliferator-activated receptor (PPARγ), a master regulator of adipogenesis. However, the mechanism of PPARγ gene regulation by glucocorticoids, the glucocorticoid receptor (GR), and its coregulators is poorly understood. Here we show that two GR binding regions (GBRs) in the mouse PPARγ gene were responsive to glucocorticoid, and treatment of 3T3-L1 preadipocytes with glucocorticoid alone induced GR occupancy and chromatin remodeling at PPARγ GBRs, which also contain binding sites for C/EBP and PPARγ proteins. GR recruited cell cycle and apoptosis regulator 1 (CCAR1), a transcription coregulator, to the PPARγ gene GBRs. Notably, CCAR1 was required for GR occupancy and chromatin remodeling at one of the PPARγ gene GBRs. Moreover, depletion of CCAR1 markedly suppressed differentiation of mouse mesenchymal stem cells and 3T3-L1 preadipocytes to mature adipocytes and decreased induction of PPARγ, C/EBPα, and C/EBPδ. Although CCAR1 was required for stimulation of several GR-regulated adipogenic genes in 3T3-L1 preadipocytes by glucocorticoid, it was not required for GR-activated transcription of certain anti-inflammatory genes in human A549 lung epithelial cells. Overall, our results highlighted the novel and specific roles of GR and CCAR1 in adipogenesis.


Asunto(s)
Adipocitos/metabolismo , Adipogénesis , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de Ciclo Celular/metabolismo , Ensamble y Desensamble de Cromatina , Receptores de Glucocorticoides/metabolismo , Transducción de Señal , Adipocitos/citología , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Glucocorticoides/metabolismo , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIH , PPAR gamma/genética , PPAR gamma/metabolismo
4.
J Clin Invest ; 134(17)2024 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-39225101

RESUMEN

Metastasis is the leading cause of cancer-related deaths. It is unclear how intratumor heterogeneity (ITH) contributes to metastasis and how metastatic cells adapt to distant tissue environments. The study of these adaptations is challenged by the limited access to patient material and a lack of experimental models that appropriately recapitulate ITH. To investigate metastatic cell adaptations and the contribution of ITH to metastasis, we analyzed single-cell transcriptomes of matched primary tumors and metastases from patient-derived xenograft models of breast cancer. We found profound transcriptional differences between the primary tumor and metastatic cells. Primary tumors upregulated several metabolic genes, whereas motility pathway genes were upregulated in micrometastases, and stress response signaling was upregulated during progression. Additionally, we identified primary tumor gene signatures that were associated with increased metastatic potential and correlated with patient outcomes. Immune-regulatory control pathways were enriched in poorly metastatic primary tumors, whereas genes involved in epithelial-mesenchymal transition were upregulated in highly metastatic tumors. We found that ITH was dominated by epithelial-mesenchymal plasticity (EMP), which presented as a dynamic continuum with intermediate EMP cell states characterized by specific genes such as CRYAB and S100A2. Elevated expression of an intermediate EMP signature correlated with worse patient outcomes. Our findings identified inhibition of the intermediate EMP cell state as a potential therapeutic target to block metastasis.


Asunto(s)
Neoplasias de la Mama , Transición Epitelial-Mesenquimal , Metástasis de la Neoplasia , Análisis de la Célula Individual , Humanos , Neoplasias de la Mama/patología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Femenino , Animales , Ratones , Regulación Neoplásica de la Expresión Génica , Transcriptoma , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Línea Celular Tumoral
5.
Drug Discov Today Dis Mech ; 10(1-2): e55-e61, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23878588

RESUMEN

As rates of obesity soar in the Unites States and around the world, cancer attributed to obesity has emerged as major threat to public health. The link between obesity and cancer can be attributed in part to the state of chronic inflammation that develops in obesity. Acetyl-CoA production and protein acetylation patterns are highly sensitive to metabolic state and are significantly altered in obesity. In this article, we explore the potential role of nutrient-sensitive lysine acetylation in regulating inflammatory processes in obesity-linked cancer.

6.
Nat Commun ; 13(1): 3671, 2022 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-35760778

RESUMEN

Few patients with triple negative breast cancer (TNBC) benefit from immune checkpoint inhibitors with complete and durable remissions being quite rare. Oncogenes can regulate tumor immune infiltration, however whether oncogenes dictate diminished response to immunotherapy and whether these effects are reversible remains poorly understood. Here, we report that TNBCs with elevated MYC expression are resistant to immune checkpoint inhibitor therapy. Using mouse models and patient data, we show that MYC signaling is associated with low tumor cell PD-L1, low overall immune cell infiltration, and low tumor cell MHC-I expression. Restoring interferon signaling in the tumor increases MHC-I expression. By combining a TLR9 agonist and an agonistic antibody against OX40 with anti-PD-L1, mice experience tumor regression and are protected from new TNBC tumor outgrowth. Our findings demonstrate that MYC-dependent immune evasion is reversible and druggable, and when strategically targeted, may improve outcomes for patients treated with immune checkpoint inhibitors.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Animales , Antígeno B7-H1/metabolismo , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Evasión Inmune , Inmunoterapia , Ratones , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Transducción de Señal , Neoplasias de la Mama Triple Negativas/metabolismo
7.
Nat Microbiol ; 7(10): 1605-1620, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36138165

RESUMEN

Pharmaceuticals have extensive reciprocal interactions with the microbiome, but whether bacterial drug sensitivity and metabolism is driven by pathways conserved in host cells remains unclear. Here we show that anti-cancer fluoropyrimidine drugs inhibit the growth of gut bacterial strains from 6 phyla. In both Escherichia coli and mammalian cells, fluoropyrimidines disrupt pyrimidine metabolism. Proteobacteria and Firmicutes metabolized 5-fluorouracil to its inactive metabolite dihydrofluorouracil, mimicking the major host mechanism for drug clearance. The preTA operon was necessary and sufficient for 5-fluorouracil inactivation by E. coli, exhibited high catalytic efficiency for the reductive reaction, decreased the bioavailability and efficacy of oral fluoropyrimidine treatment in mice and was prevalent in the gut microbiomes of colorectal cancer patients. The conservation of both the targets and enzymes for metabolism of therapeutics across domains highlights the need to distinguish the relative contributions of human and microbial cells to drug efficacy and side-effect profiles.


Asunto(s)
Antineoplásicos , Escherichia coli , Animales , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Bacterias/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Fluorouracilo/metabolismo , Fluorouracilo/farmacología , Humanos , Mamíferos , Redes y Vías Metabólicas , Ratones
8.
Cancers (Basel) ; 13(1)2021 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-33466329

RESUMEN

Targeting a tumor's metabolic dependencies is a clinically actionable therapeutic approach; however, identifying subtypes of tumors likely to respond remains difficult. The use of lipids as a nutrient source is of particular importance, especially in breast cancer. Imaging techniques offer the opportunity to quantify nutrient use in preclinical tumor models to guide development of new drugs that restrict uptake or utilization of these nutrients. We describe a fast and dynamic approach to image fatty acid uptake in vivo and demonstrate its relevance to study both tumor metabolic reprogramming directly, as well as the effectiveness of drugs targeting lipid metabolism. Specifically, we developed a quantitative optical approach to spatially and longitudinally map the kinetics of long-chain fatty acid uptake in in vivo murine models of breast cancer using a fluorescently labeled palmitate molecule, Bodipy FL c16. We chose intra-vital microscopy of mammary tumor windows to validate our approach in two orthotopic breast cancer models: a MYC-overexpressing, transgenic, triple-negative breast cancer (TNBC) model and a murine model of the 4T1 family. Following injection, Bodipy FL c16 fluorescence increased and reached its maximum after approximately 30 min, with the signal remaining stable during the 30-80 min post-injection period. We used the fluorescence at 60 min (Bodipy60), the mid-point in the plateau region, as a summary parameter to quantify Bodipy FL c16 fluorescence in subsequent experiments. Using our imaging platform, we observed a two- to four-fold decrease in fatty acid uptake in response to the downregulation of the MYC oncogene, consistent with findings from in vitro metabolic assays. In contrast, our imaging studies report an increase in fatty acid uptake with tumor aggressiveness (6NR, 4T07, and 4T1), and uptake was significantly decreased after treatment with a fatty acid transport inhibitor, perphenazine, in both normal mammary pads and in the most aggressive 4T1 tumor model. Our approach fills an important gap between in vitro assays providing rich metabolic information at static time points and imaging approaches visualizing metabolism in whole organs at a reduced resolution.

9.
Cell Rep ; 30(10): 3368-3382.e7, 2020 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-32160543

RESUMEN

Tumors that overexpress the MYC oncogene are frequently aneuploid, a state associated with highly aggressive cancers and tumor evolution. However, how MYC causes aneuploidy is not well understood. Here, we show that MYC overexpression induces mitotic spindle assembly defects and chromosomal instability (CIN) through effects on microtubule nucleation and organization. Attenuating MYC expression reverses mitotic defects, even in established tumor cell lines, indicating an ongoing role for MYC in CIN. MYC reprograms mitotic gene expression, and we identify TPX2 to be permissive for spindle assembly in MYC-high cells. TPX2 depletion blocks mitotic progression, induces cell death, and prevents tumor growth. Further elevating TPX2 expression reduces mitotic defects in MYC-high cells. MYC and TPX2 expression may be useful biomarkers to stratify patients for anti-mitotic therapies. Our studies implicate MYC as a regulator of mitosis and suggest that blocking MYC activity can attenuate the emergence of CIN and tumor evolution.


Asunto(s)
Mitosis , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Muerte Celular , Línea Celular Tumoral , Inestabilidad Cromosómica , Citoprotección , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Huso Acromático/metabolismo , Mutaciones Letales Sintéticas
10.
Elife ; 92020 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-33170774

RESUMEN

The Pro47Ser variant of p53 (S47) exists in African-descent populations and is associated with increased cancer risk in humans and mice. Due to impaired repression of the cystine importer Slc7a11, S47 cells show increased glutathione (GSH) accumulation compared to cells with wild -type p53. We show that mice containing the S47 variant display increased mTOR activity and oxidative metabolism, as well as larger size, improved metabolic efficiency, and signs of superior fitness. Mechanistically, we show that mTOR and its positive regulator Rheb display increased association in S47 cells; this is due to an altered redox state of GAPDH in S47 cells that inhibits its ability to bind and sequester Rheb. Compounds that decrease glutathione normalize GAPDH-Rheb complexes and mTOR activity in S47 cells. This study reveals a novel layer of regulation of mTOR by p53, and raises the possibility that this variant may have been selected for in early Africa.


Asunto(s)
Serina-Treonina Quinasas TOR/metabolismo , Proteína p53 Supresora de Tumor/genética , Sustitución de Aminoácidos/genética , Animales , Población Negra/genética , Línea Celular , Glutatión/metabolismo , Glucólisis , Humanos , Mitocondrias/metabolismo , Oxidación-Reducción , Serina-Treonina Quinasas TOR/genética , Proteína p53 Supresora de Tumor/metabolismo
12.
Cell Rep ; 17(4): 1037-1052, 2016 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-27760311

RESUMEN

Mechanisms of metabolic flexibility enable cells to survive under stressful conditions and can thwart therapeutic responses. Acetyl-coenzyme A (CoA) plays central roles in energy production, lipid metabolism, and epigenomic modifications. Here, we show that, upon genetic deletion of Acly, the gene coding for ATP-citrate lyase (ACLY), cells remain viable and proliferate, although at an impaired rate. In the absence of ACLY, cells upregulate ACSS2 and utilize exogenous acetate to provide acetyl-CoA for de novo lipogenesis (DNL) and histone acetylation. A physiological level of acetate is sufficient for cell viability and abundant acetyl-CoA production, although histone acetylation levels remain low in ACLY-deficient cells unless supplemented with high levels of acetate. ACLY-deficient adipocytes accumulate lipid in vivo, exhibit increased acetyl-CoA and malonyl-CoA production from acetate, and display some differences in fatty acid content and synthesis. Together, these data indicate that engagement of acetate metabolism is a crucial, although partial, mechanism of compensation for ACLY deficiency.


Asunto(s)
ATP Citrato (pro-S)-Liasa/metabolismo , Acetatos/metabolismo , Glucosa/metabolismo , ATP Citrato (pro-S)-Liasa/deficiencia , Acetato CoA Ligasa/metabolismo , Acetatos/farmacología , Acetilcoenzima A/metabolismo , Acetilación , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Eliminación de Gen , Histonas/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Lípidos/biosíntesis , Masculino , Ratones , Regulación hacia Arriba/efectos de los fármacos
13.
Mol Cell Oncol ; 2(2): e965620, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-27308412

RESUMEN

Histone acetylation is sensitive to the availability of acetyl-CoA. However, the extent to which metabolic alterations in cancer cells impact tumor histone acetylation has been unclear. Here, we discuss our recent findings that oncogenic AKT1 activation regulates histone acetylation levels in tumors through regulation of acetyl-CoA metabolism.

14.
Cell Rep ; 11(6): 893-901, 2015 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-25937285

RESUMEN

Replication stress induced by nucleotide deficiency plays an important role in cancer initiation. Replication stress in primary cells typically activates the cellular senescence tumor-suppression mechanism. Senescence bypass correlates with development of cancer, a disease characterized by metabolic reprogramming. However, the role of metabolic reprogramming in the cellular response to replication stress has been little explored. Here, we report that ataxia telangiectasia mutated (ATM) plays a central role in regulating the cellular response to replication stress by shifting cellular metabolism. ATM inactivation bypasses senescence induced by replication stress triggered by nucleotide deficiency. This was due to restoration of deoxyribonucleotide triphosphate (dNTP) levels through both upregulation of the pentose phosphate pathway via increased glucose-6-phosphate dehydrogenase (G6PD) activity and enhanced glucose and glutamine consumption. These phenotypes were mediated by a coordinated suppression of p53 and upregulation of c-MYC downstream of ATM inactivation. Our data indicate that ATM status couples replication stress and metabolic reprogramming during senescence.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Reprogramación Celular , Senescencia Celular , Replicación del ADN , Estrés Fisiológico , Línea Celular , Daño del ADN , Técnicas de Silenciamiento del Gen , Glucosa/metabolismo , Glucosafosfato Deshidrogenasa/metabolismo , Glutamina/metabolismo , Humanos , Nucleótidos/metabolismo , Vía de Pentosa Fosfato , Proteínas Proto-Oncogénicas c-myc/metabolismo , Especificidad por Sustrato , Proteína p53 Supresora de Tumor/metabolismo
15.
Cell Metab ; 20(2): 306-319, 2014 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-24998913

RESUMEN

Histone acetylation plays important roles in gene regulation, DNA replication, and the response to DNA damage, and it is frequently deregulated in tumors. We postulated that tumor cell histone acetylation levels are determined in part by changes in acetyl coenzyme A (acetyl-CoA) availability mediated by oncogenic metabolic reprogramming. Here, we demonstrate that acetyl-CoA is dynamically regulated by glucose availability in cancer cells and that the ratio of acetyl-CoA:coenzyme A within the nucleus modulates global histone acetylation levels. In vivo, expression of oncogenic Kras or Akt stimulates histone acetylation changes that precede tumor development. Furthermore, we show that Akt's effects on histone acetylation are mediated through the metabolic enzyme ATP-citrate lyase and that pAkt(Ser473) levels correlate significantly with histone acetylation marks in human gliomas and prostate tumors. The data implicate acetyl-CoA metabolism as a key determinant of histone acetylation levels in cancer cells.


Asunto(s)
Histonas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Acetilcoenzima A/metabolismo , Acetilación/efectos de los fármacos , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Análisis por Conglomerados , Coenzima A/metabolismo , Glioma/metabolismo , Glioma/patología , Glucosa/farmacología , Humanos , Interleucina-3/farmacología , Masculino , Fosforilación/efectos de los fármacos , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Proteínas ras/genética , Proteínas ras/metabolismo
16.
PLoS One ; 5(10): e13754, 2010 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-21060794

RESUMEN

BACKGROUND: Glucocorticoids are potent anti-inflammatory agents commonly used to treat inflammatory diseases. They convey signals through the intracellular glucocorticoid receptor (GR), which upon binding to ligands, associates with genomic glucocorticoid response elements (GREs) to regulate transcription of associated genes. One mechanism by which glucocorticoids inhibit inflammation is through induction of the dual specificity phosphatase-1 (DUSP1, a.k.a. mitogen-activated protein kinase phosphatase-1, MKP-1) gene. METHODOLOGY/PRINCIPAL FINDINGS: We found that glucocorticoids rapidly increased transcription of DUSP1 within 10 minutes in A549 human lung adenocarcinoma cells. Using chromatin immunoprecipitation (ChIP) scanning, we located a GR binding region between -1421 and -1118 upstream of the DUSP1 transcription start site. This region is active in a reporter system, and mutagenesis analyses identified a functional GRE located between -1337 and -1323. We found that glucocorticoids increased DNase I hypersensitivity, reduced nucleosome density, and increased histone H3 and H4 acetylation within genomic regions surrounding the GRE. ChIP experiments showed that p300 was recruited to the DUSP1 GRE, and RNA interference experiments demonstrated that reduction of p300 decreased glucocorticoid-stimulated DUSP1 gene expression and histone H3 hyperacetylation. Furthermore, overexpression of p300 potentiated glucocorticoid-stimulated activity of a reporter gene containing the DUSP1 GRE, and this coactivation effect was compromised when the histone acetyltransferase domain was mutated. ChIP-reChIP experiments using GR followed by p300 antibodies showed significant enrichment of the DUSP1 GRE upon glucocorticoid treatment, suggesting that GR and p300 are in the same protein complex recruited to the DUSP1 GRE. CONCLUSIONS/SIGNIFICANCE: Our studies identified a functional GRE for the DUSP1 gene. Moreover, the transcriptional activation of DUSP1 by glucocorticoids requires p300 and a rapid modification of the chromatin structure surrounding the GRE. Overall, understanding the mechanism of glucocorticoid-induced DUSP1 gene transcription could provide insights into therapeutic approaches against inflammatory diseases.


Asunto(s)
Dexametasona/farmacología , Fosfatasa 1 de Especificidad Dual/genética , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Acetilación , Secuencia de Bases , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Cartilla de ADN , Fosfatasa 1 de Especificidad Dual/metabolismo , Histonas/metabolismo , Humanos , Mutagénesis Sitio-Dirigida , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas
17.
PLoS One ; 5(12): e15188, 2010 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-21187916

RESUMEN

Glucocorticoids play important roles in the regulation of distinct aspects of adipocyte biology. Excess glucocorticoids in adipocytes are associated with metabolic disorders, including central obesity, insulin resistance and dyslipidemia. To understand the mechanisms underlying the glucocorticoid action in adipocytes, we used chromatin immunoprecipitation sequencing to isolate genome-wide glucocorticoid receptor (GR) binding regions (GBRs) in 3T3-L1 adipocytes. Furthermore, gene expression analyses were used to identify genes that were regulated by glucocorticoids. Overall, 274 glucocorticoid-regulated genes contain or locate nearby GBR. We found that many GBRs were located in or nearby genes involved in triglyceride (TG) synthesis (Scd-1, 2, 3, GPAT3, GPAT4, Agpat2, Lpin1), lipolysis (Lipe, Mgll), lipid transport (Cd36, Lrp-1, Vldlr, Slc27a2) and storage (S3-12). Gene expression analysis showed that except for Scd-3, the other 13 genes were induced in mouse inguinal fat upon 4-day glucocorticoid treatment. Reporter gene assays showed that except Agpat2, the other 12 glucocorticoid-regulated genes contain at least one GBR that can mediate hormone response. In agreement with the fact that glucocorticoids activated genes in both TG biosynthetic and lipolytic pathways, we confirmed that 4-day glucocorticoid treatment increased TG synthesis and lipolysis concomitantly in inguinal fat. Notably, we found that 9 of these 12 genes were induced in transgenic mice that have constant elevated plasma glucocorticoid levels. These results suggested that a similar mechanism was used to regulate TG homeostasis during chronic glucocorticoid treatment. In summary, our studies have identified molecular components in a glucocorticoid-controlled gene network involved in the regulation of TG homeostasis in adipocytes. Understanding the regulation of this gene network should provide important insight for future therapeutic developments for metabolic diseases.


Asunto(s)
Adipocitos/citología , Estudio de Asociación del Genoma Completo , Receptores de Glucocorticoides/genética , Triglicéridos/metabolismo , Células 3T3-L1 , Animales , Redes Reguladoras de Genes , Genoma , Glucocorticoides/metabolismo , Homeostasis , Lipólisis , Masculino , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Presión
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA