Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 183
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 133(23): 2507-2517, 2019 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-30952671

RESUMEN

CCAAT/enhancer binding protein ε (CEBPE) is an essential transcription factor for granulocytic differentiation. Mutations of CEBPE occur in individuals with neutrophil-specific granule deficiency (SGD), which is characterized by defects in neutrophil maturation. Cebpe-knockout mice also exhibit defects in terminal differentiation of granulocytes, a phenotype reminiscent of SGD. Analysis of DNase I hypersensitive sites sequencing data revealed an open chromatin region 6 kb downstream of the transcriptional start site of Cebpe in murine myeloid cells. We identified an interaction between this +6-kb region and the core promoter of Cebpe using circular chromosome conformation capture sequencing (4C-seq). To understand the role of this putative enhancer in transcriptional regulation of Cebpe, we targeted it using catalytically inactive Cas9 fused to Krüppel-associated box (KRAB) domain and observed a significant downregulation of transcript and protein levels of CEBPE in cells expressing guide RNA targeting the +6-kb region. To further investigate the role of this novel enhancer further in myelopoiesis, we generated mice with deletion of this region using CRISPR/Cas9 technology. Germline deletion of the +6-kb enhancer resulted in reduced levels of CEBPE and its target genes and caused a severe block in granulocytic differentiation. We also identified binding of CEBPA and CEBPE to the +6-kb enhancer, which suggests their role in regulating the expression of Cebpe In summary, we have identified a novel enhancer crucial for regulating expression of Cebpe and required for normal granulocytic differentiation.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/biosíntesis , Diferenciación Celular/genética , Regulación de la Expresión Génica/genética , Granulocitos/metabolismo , Mielopoyesis/genética , Animales , Proteínas Potenciadoras de Unión a CCAAT/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
2.
Nucleic Acids Res ; 46(12): 5996-6008, 2018 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-29733382

RESUMEN

3'-untranslated regions (UTRs) can vary through the use of alternative polyadenylation sites during pre-mRNA processing. Multiple publically available pipelines combining high profiling technologies and bioinformatics tools have been developed to catalog changes in 3'-UTR lengths. In our recent RNA-seq experiments using cells with hyper-activated mammalian target of rapamycin (mTOR), we found that cellular mTOR activation leads to transcriptome-wide alternative polyadenylation (APA), resulting in the activation of multiple cellular pathways. Here, we developed a novel bioinformatics algorithm, IntMAP, which integrates RNA-Seq and PolyA Site (PAS)-Seq data for a comprehensive characterization of APA events. By applying IntMAP to the datasets from cells with hyper-activated mTOR, we identified novel APA events that could otherwise not be identified by either profiling method alone. Several transcription factors including Cebpg (CCAAT/enhancer binding protein gamma) were among the newly discovered APA transcripts, indicating that diverse transcriptional networks may be regulated by mTOR-coordinated APA. The prevention of APA in Cebpg using the CRISPR/cas9-mediated genome editing tool showed that mTOR-driven 3'-UTR shortening in Cebpg is critical in protecting cells from endoplasmic reticulum (ER) stress. Taken together, we present IntMAP as a new bioinformatics algorithm for APA analysis by which we expand our understanding of the physiological role of mTOR-coordinated APA events to ER stress response. IntMAP toolbox is available at http://compbio.cs.umn.edu/IntMAP/.


Asunto(s)
Algoritmos , Estrés del Retículo Endoplásmico/genética , Poliadenilación , Serina-Treonina Quinasas TOR/metabolismo , Regiones no Traducidas 3' , Animales , Proteínas Potenciadoras de Unión a CCAAT/biosíntesis , Proteínas Potenciadoras de Unión a CCAAT/genética , Células Cultivadas , Ratones
3.
Hum Mol Genet ; 26(8): 1522-1534, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28334952

RESUMEN

DNMT1 is recruited to substrate sites by PCNA and UHRF1 to maintain DNA methylation after replication. The cell cycle dependent recruitment of DNMT1 is mediated by the PCNA-binding domain (PBD) and the targeting sequence (TS) within the N-terminal regulatory domain. The TS domain was found to be mutated in patients suffering from hereditary sensory and autonomic neuropathies with dementia and hearing loss (HSANIE) and autosomal dominant cerebellar ataxia deafness and narcolepsy (ADCA-DN) and is associated with global hypomethylation and site specific hypermethylation. With functional complementation assays in mouse embryonic stem cells, we showed that DNMT1 mutations P496Y and Y500C identified in HSANIE patients not only impair DNMT1 heterochromatin association, but also UHRF1 interaction resulting in hypomethylation. Similar DNA methylation defects were observed when DNMT1 interacting domains in UHRF1, the UBL and the SRA domain, were deleted. With cell-based assays, we could show that HSANIE associated mutations perturb DNMT1 heterochromatin association and catalytic complex formation at methylation sites and decrease protein stability in late S and G2 phase. To investigate the neuronal phenotype of HSANIE mutations, we performed DNMT1 rescue assays and could show that cells expressing mutated DNMT1 were prone to apoptosis and failed to differentiate into neuronal lineage. Our results provide insights into the molecular basis of DNMT1 dysfunction in HSANIE patients and emphasize the importance of the TS domain in the regulation of DNA methylation in pluripotent and differentiating cells.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/genética , Diferenciación Celular/genética , ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN/genética , Neuropatías Hereditarias Sensoriales y Autónomas/genética , Animales , Apoptosis/genética , Proteínas Potenciadoras de Unión a CCAAT/biosíntesis , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/biosíntesis , Regulación de la Expresión Génica , Neuropatías Hereditarias Sensoriales y Autónomas/patología , Heterocromatina/genética , Humanos , Ratones , Células Madre Embrionarias de Ratones/metabolismo , Mutación , Neuronas/metabolismo , Neuronas/patología , Dominios Proteicos/genética , Dominios y Motivos de Interacción de Proteínas/genética , Estabilidad Proteica , Ubiquitina-Proteína Ligasas
4.
Prostate ; 79(3): 302-311, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30430607

RESUMEN

BACKGROUND: The transcription factor CCAAT-enhancer-binding protein alpha (CEBPA) is a crucial regulator of cell proliferation and differentiation. Expression levels of CEBPA have been suggested to be prognostic in various tumor types. METHODS: Here, we analyzed the immunohistochemical expression of CEBPA in a tissue microarray containing more than 17 000 prostate cancer specimens with annotated clinical and molecular data including for example TMPRSS2:ERG fusion and PTEN deletion status. RESULTS: Normal prostate glands showed moderate to strong CEBPA staining, while CEBPA expression was frequently reduced (40%) or lost (30%) in prostate cancers. Absence of detectable CEBPA expression was markedly more frequent in ERG negative (45%) as compared to ERG positive cancers (20%, P < 0.0001). Reduced CEBPA expression was linked to unfavorable phenotype (P < 0.0001) and poor prognosis (P = 0.0008). Subgroup analyses revealed, that the prognostic value of CEBPA loss was entirely driven by tumors carrying both TMPRSS2:ERG fusions and PTEN deletions. In this subgroup, CEBPA loss was tightly linked to advanced tumor stage (P < 0.0001), high Gleason grade (P < 0.0001), positive nodal stage (0.0003), and early biochemical recurrence (P = 0.0007), while these associations were absent or markedly diminished in tumors with normal PTEN copy numbers and/or absence of ERG fusion. CONCLUSIONS: CEBPA is down regulated in about one third of prostate cancers, but the clinical impact of CEBPA loss is strictly limited to the subset of about 10% prostate cancers carrying both ERG fusion and deletions of the PTEN tumor suppressor. Our findings challenge the concept that prognostic molecular markers may be generally applicable to all prostate cancers.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/deficiencia , Proteínas de Fusión Oncogénica/metabolismo , Fosfohidrolasa PTEN/deficiencia , Neoplasias de la Próstata/metabolismo , Anciano , Proteínas Potenciadoras de Unión a CCAAT/biosíntesis , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Dosificación de Gen , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Proteínas de Fusión Oncogénica/genética , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Pronóstico , Prostatectomía , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/cirugía , Análisis de Matrices Tisulares
5.
PLoS Genet ; 12(12): e1006474, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27923061

RESUMEN

Increasing energy expenditure through brown adipocyte recruitment is a promising approach to combat obesity. We report here the comprehensive profiling of the epigenome and transcriptome throughout the lineage commitment and differentiation of C3H10T1/2 mesenchymal stem cell line into brown adipocytes. Through direct comparison to datasets from differentiating white adipocytes, we systematically identify stage- and lineage-specific coding genes, lncRNAs and microRNAs. Utilizing chromatin state maps, we also define stage- and lineage-specific enhancers, including super-enhancers, and their associated transcription factor binding motifs and genes. Through these analyses, we found that in brown adipocytes, brown lineage-specific genes are pre-marked by both H3K4me1 and H3K27me3, and the removal of H3K27me3 at the late stage is necessary but not sufficient to promote brown gene expression, while the pre-deposition of H3K4me1 plays an essential role in poising the brown genes for expression in mature brown cells. Moreover, we identify SOX13 as part of a p38 MAPK dependent transcriptional response mediating early brown cell lineage commitment. We also identify and subsequently validate PIM1, SIX1 and RREB1 as novel regulators promoting brown adipogenesis. Finally, we show that SIX1 binds to adipogenic and brown marker genes and interacts with C/EBPα, C/EBPß and EBF2, suggesting their functional cooperation during adipogenesis.


Asunto(s)
Adipogénesis/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas de Homeodominio/genética , Obesidad/genética , Tejido Adiposo Pardo/crecimiento & desarrollo , Tejido Adiposo Pardo/metabolismo , Animales , Autoantígenos/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Proteína beta Potenciadora de Unión a CCAAT/biosíntesis , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/biosíntesis , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Diferenciación Celular/genética , Linaje de la Célula/genética , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Metabolismo Energético/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Células Madre Mesenquimatosas , Ratones , Obesidad/metabolismo , Obesidad/patología , ARN Largo no Codificante/biosíntesis , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Transcriptoma/genética
6.
PLoS Genet ; 11(10): e1005603, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26484536

RESUMEN

The ability of specific neurons to regenerate their axons after injury is governed by cell-intrinsic regeneration pathways. In Caenorhabditis elegans, the JNK and p38 MAPK pathways are important for axon regeneration. Axonal injury induces expression of the svh-2 gene encoding a receptor tyrosine kinase, stimulation of which by the SVH-1 growth factor leads to activation of the JNK pathway. Here, we identify ETS-4 and CEBP-1, related to mammalian Ets and C/EBP, respectively, as transcriptional activators of svh-2 expression following axon injury. ETS-4 and CEBP-1 function downstream of the cAMP and Ca2+-p38 MAPK pathways, respectively. We show that PKA-dependent phosphorylation of ETS-4 promotes its complex formation with CEBP-1. Furthermore, activation of both cAMP and Ca2+ signaling is required for activation of svh-2 expression. Thus, the cAMP/Ca2+ signaling pathways cooperatively activate the JNK pathway, which then promotes axon regeneration.


Asunto(s)
Axones/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas de Caenorhabditis elegans/genética , Regeneración/genética , Factores de Transcripción/genética , Animales , Axones/fisiología , Proteínas Potenciadoras de Unión a CCAAT/biosíntesis , Caenorhabditis elegans/genética , Caenorhabditis elegans/crecimiento & desarrollo , Proteínas de Caenorhabditis elegans/biosíntesis , Señalización del Calcio/genética , AMP Cíclico/genética , AMP Cíclico/metabolismo , Regulación del Desarrollo de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Péptidos y Proteínas de Señalización Intercelular/genética , Sistema de Señalización de MAP Quinasas/genética , Neuronas/metabolismo , Fosforilación , Transducción de Señal/genética , Factores de Transcripción/biosíntesis , Proteínas Quinasas p38 Activadas por Mitógenos/genética
7.
Biochim Biophys Acta ; 1859(2): 348-54, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26704017

RESUMEN

Mammalian Sirtuin proteins (SIRTs) are homologs of yeast Sir2, and characterized as class III histone deacetylases of NAD(+) dependence. Unlike their lower counterparts that are directly involved in the extending of lifespan, mammalian SIRTs mainly function in metabolism and cellular homeostasis, among them, SIRT7 is the least understood. SIRT7 is localized in the nucleus and rich in nucleoli associated with RNA polymerase I, and correlated with cell proliferation. In contrast, SIRT7 has recently been demonstrated to specifically deacetylate H3K18ac in the chromatin, and in most cases represses proliferation. Although MicroRNA as miR-125b has been reported to down-regulate SIRT7 by binding to its 3'UTR, however, how SIRT7 gene is regulated remains unclear. Here, we identified the transcription initiation site of human SIRT7 gene at the upstream 23rd A nucleotide respective to the translational codon, and the SIRT7 is a TATA-less and initiator-less gene. The sequences in the upstream region between -256 and -129 bp are identical with important functions in the three species detected. A C/EBPα responding element is found that binds both C/EBPα and C/EBPß in vitro. We showed TSA induced SIRT7 gene transcription and only the HDAC3, but not its catalytic domain depleted mutant, interacted with C/EBPα to occupy the C/EBPα element and repressed SIRT7 gene in the hepatocellular carcinoma cells. To our knowledge, this is the first report on the regulation mechanism of SIRT7 gene, in which, HDAC3 collaborated with C/EBPα to occupy its responding element in the upstream region of SIRT7 gene and repressed its expression in human cells.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/genética , Carcinoma Hepatocelular/genética , Histona Desacetilasas/genética , Neoplasias Hepáticas/genética , Sirtuinas/genética , Regiones no Traducidas 3' , Animales , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/biosíntesis , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Cromatina/genética , Histona Desacetilasas/biosíntesis , Humanos , Neoplasias Hepáticas/patología , Regiones Promotoras Genéticas , Sirtuinas/biosíntesis
8.
Mol Cell Biochem ; 431(1-2): 103-112, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28285359

RESUMEN

Esophageal adenocarcinoma (EAC) is one of the most common malignancies in the world which is associated the increased prevalence of obesity. In the context of obesity, leptin can directly contribute to progression of EAC. Adiponectin inhibits leptin-induced oncogenic signaling in EAC cells. However, the exact molecular mechanisms linking obesity, adipokines, and EAC remain far from completely understood. In the present study, we tested the role of ubiquitin-like with PHD and ring finger domains 1 (UHRF1) in adiponectin-induced protective effects against leptin-induced EAC cell proliferation. We found that globular adiponectin (gAD) significantly inhibited leptin-induced increase of cell proliferation and decrease of apoptosis in OE 19 cells. Moreover, leptin-induced increase of UHRF1 expression was suppressed by gAD. Compared with normal controls, UHRF1 expression was markedly increased in EAC tissues and cell lines. Silence of UHRF1 increased the expression of cleaved caspase 3 and 9 and Bax, reduced the expression of Bcl-2, promoted apoptosis, and inhibited cell proliferation in OE 19 cells. Overexpression of UHRF1 significantly blocked gAD-induced decrease of cell proliferation and increase of apoptosis in leptin-treated cells. Silence of adiponectin receptor 1/2 (AdipoR1/2) could inhibit gAD-induced decrease of cell proliferation and increase of apoptosis in leptin-treated cells. Silence of AdipoR2, but not AdipoR1, suppressed gAD-induced decrease of UHRF1 expression in leptin-treated cells. The results indicated that gAD inhibited the prooncogenic effects of leptin via AdipoR2-mediated suppression of UHRF1. Our study provides novel insights into the role of UHRF1 in the development of EAC and the mechanism of antitumor effect of gAD.


Asunto(s)
Adenocarcinoma/metabolismo , Adiponectina/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/biosíntesis , Proliferación Celular , Neoplasias Esofágicas/metabolismo , Leptina/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores de Adiponectina/metabolismo , Adenocarcinoma/patología , Línea Celular Tumoral , Neoplasias Esofágicas/patología , Humanos , Ubiquitina-Proteína Ligasas
9.
Anticancer Drugs ; 28(4): 384-391, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28059831

RESUMEN

As the second most common cancer in men around the world, prostate cancer is increasingly gaining more attention. Dihydroartemisinin (DHA) has been proven to be a promising anticancer agent in vitro as well as in vivo in accumulating data. However, the detailed mechanisms of how DHA action in human prostate cancer PC-3 cells remain elusive. This study aimed to investigate the effects of DHA, a novel anticancer agent, by inhibiting the expression of ubiquitin like containing PHD and ring finger 1 (UHRF1) in PC-3 cells. The apoptosis and cell-cycle distribution were detected by flow cytometry. Quantitative real-time PCR was performed to examine both UHRF1 and DNA methyltransferase 1 (DNMT1) expressions at mRNA levels, whereas the expressions of UHRF1, DNMT1, and p16 proteins at protein levels were detected by Western blotting. Methylation levels of p16 CpG islands were determined by bisulfite genomic sequencing. We showed that DHA induced the downregulation of UHRF1 and DNMT1, accompanied by an upregulation of p16 in PC-3 cells. Decreased p16 promoter methylation levels in DHA-treated groups were also observed in PC-3 cells. Furthermore, DHA significantly induced apoptosis and G1/S cell-cycle arrest in PC-3 cells. Our results suggested that downregulation of UHRF1/DNMT1 is upstream to many cellular events, including G1 cell arrest, demethylation of p16, and apoptosis. Together, our study provides new evidence that DHA may serve as a potential therapeutic agent in the treatment of prostate cancer.


Asunto(s)
Artemisininas/farmacología , Proteínas Potenciadoras de Unión a CCAAT/biosíntesis , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Línea Celular Tumoral , Islas de CpG , Inhibidor p16 de la Quinasa Dependiente de Ciclina/biosíntesis , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , ADN (Citosina-5-)-Metiltransferasa 1/biosíntesis , ADN (Citosina-5-)-Metiltransferasa 1/genética , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Metilación de ADN , Regulación hacia Abajo/efectos de los fármacos , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Humanos , Masculino , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Fase S/efectos de los fármacos , Ubiquitina-Proteína Ligasas , Regulación hacia Arriba/efectos de los fármacos
10.
Blood ; 121(1): 159-69, 2013 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-23100311

RESUMEN

Recently, we showed that increased miR-181a expression was associated with improved outcomes in cytogenetically normal acute myeloid leukemia (CN-AML). Interestingly, miR-181a expression was increased in CN-AML patients harboring CEBPA mutations, which are usually biallelic and associate with better prognosis. CEBPA encodes the C/EBPα transcription factor. We demonstrate here that the presence of N-terminal CEBPA mutations and miR-181a expression are linked. Indeed, the truncated C/EBPα-p30 isoform, which is produced from the N-terminal mutant CEBPA gene or from the differential translation of wild-type CEBPA mRNA and is commonly believed to have no transactivation activity, binds to the miR-181a-1 promoter and up-regulates the microRNA expression. Furthermore, we show that lenalidomide, a drug approved for myelodysplastic syndromes and multiple myeloma, enhances translation of the C/EBPα-p30 isoform, resulting in higher miR-181a levels. In xenograft mouse models, ectopic miR-181a expression inhibits tumor growth. Similarly, lenalidomide exhibits antitumorigenic activity paralleled by increased miR-181a expression. This regulatory pathway may explain an increased sensitivity to apoptosis-inducing chemotherapy in subsets of AML patients. Altogether, our data provide a potential explanation for the improved clinical outcomes observed in CEBPA-mutated CN-AML patients, and suggest that lenalidomide treatment enhancing the C/EBPα-p30 protein levels and in turn miR-181a may sensitize AML blasts to chemotherapy.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/fisiología , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Factores Inmunológicos/farmacología , Leucemia Mieloide Aguda/tratamiento farmacológico , MicroARNs/biosíntesis , Proteínas de Neoplasias/biosíntesis , ARN Neoplásico/biosíntesis , Talidomida/análogos & derivados , Adulto , Animales , Antimetabolitos Antineoplásicos/farmacología , Proteínas Potenciadoras de Unión a CCAAT/biosíntesis , Proteínas Potenciadoras de Unión a CCAAT/genética , Línea Celular Tumoral/efectos de los fármacos , Línea Celular Tumoral/metabolismo , Línea Celular Tumoral/trasplante , Citarabina/farmacología , Mutación del Sistema de Lectura , Humanos , Factores Inmunológicos/uso terapéutico , Células K562 , Lenalidomida , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , MicroARNs/genética , Proteínas de Neoplasias/genética , Mutación Puntual , Regiones Promotoras Genéticas/genética , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , Estructura Terciaria de Proteína , ARN Neoplásico/genética , Proteínas Recombinantes de Fusión/fisiología , Talidomida/farmacología , Talidomida/uso terapéutico , Regulación hacia Arriba/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Tumour Biol ; 36(11): 8887-93, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26070868

RESUMEN

Ubiquitin-like with PHD and ring finger domains 1 (UHRF1), known as ICB90 or Np95, has been found to be overexpressed in numerous cancers. In this study, we evaluated the expression level of UHRF1 in ovarian cancer. UHRF1 levels in paired ovarian cancer tissues and adjacent normal tissues from 80 ovarian cancer patients were detected using relative quantitatively PCR and Western blot. Small interfering RNA (siRNA) was introduced in two human ovarian cancer cell lines (SKOV-3 and OVCAR-3) to downregulate the expression of UHRF1. The proliferation of siRNA-treated cells was examined using cell counting kit-8 (CCK-8) assay. The growth of these cells showed a remarkable decrease. Moreover, flow cytometric and Hoechst 33342 assays were used to detect the apoptosis. The diagnostic value of UHRF1 messenger RNA (mRNA) expression in ovarian cancer was estimated by receiver-operator characteristic (ROC) curve. The correlation between UHRF1 mRNA expression and clinicopathologic features of ovarian cancer patients was evaluated by χ (2) test. Our results demonstrated that both UHRF1 mRNA and protein were highly expressed in ovarian cancer tissues and significantly higher than that in adjacent normal tissues. Moreover, the inhibition of UHRF1 may lead to cells to undergo apoptosis. Thus, UHRF1 could act as a new oncogenic factor in ovarian cancer and be a potential molecular target for ovarian cancer gene therapy.


Asunto(s)
Biomarcadores de Tumor/biosíntesis , Proteínas Potenciadoras de Unión a CCAAT/biosíntesis , Proliferación Celular/genética , Neoplasias Ováricas/genética , Apoptosis/genética , Biomarcadores de Tumor/genética , Proteínas Potenciadoras de Unión a CCAAT/genética , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Ováricas/patología , ARN Interferente Pequeño , Ubiquitina-Proteína Ligasas
12.
Mol Cell Biochem ; 400(1-2): 223-32, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25416862

RESUMEN

Ubiquitin-like with PHD and ring finger domains 1 (UHRF1) has been reported as a marker for the differential diagnosis of pancreatic cancer and chronic pancreatitis. However, the expression pattern and biological functions of UHRF1 in the progression of pancreatic cancer are not fully understood. In this study, it was found that the expression of UHRF1 was significantly up-regulated in pancreatic cancer samples compared to their adjacent normal tissues. Meanwhile, the expression of UHRF1 was inversely correlated with the survival of pancreatic cancer patients. Moreover, in the biological function studies, UHRF1 was shown to promote the growth, migration, and metastasis of pancreatic cancer cells in vitro and in vivo. Mechanistically, the expression of UHRF1 was induced by oncogenic Ras in both pancreatic cancer mouse model and cultured cells. Taken together, our study demonstrated that UHRF1 played an oncogenic role in the progression of pancreatic cancer, and UHRF1 might be a promising target for the treatment of pancreatic cancer.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/biosíntesis , Proliferación Celular/genética , Genes ras/genética , Neoplasias Pancreáticas/genética , Animales , Línea Celular Tumoral , Movimiento Celular/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Metástasis de la Neoplasia , Neoplasias Pancreáticas/patología , Ubiquitina-Proteína Ligasas , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Carcinogenesis ; 35(11): 2404-14, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24947179

RESUMEN

Genetic mutations in pancreatic ductal adenocarcinoma (PDAC) with critical roles have been well examined. The recent discovery of alterations in genes encoding histone modifiers suggests their possible roles in the complexity of cancer development. We previously reported loss of heterozygosity of the KDM6B gene, which encodes a histone demethylase for trimethylated histone H3 lysine 27, a repressive chromatin mark, in PDAC cells. In this study, we demonstrated that loss of KDM6B enhanced aggressiveness of PDAC cells. KDM6B has been regarded as a tumor suppressor that mediates oncogenic KRAS-induced senescence. Consistently, KDM6B was highly expressed in pancreatic precancerous lesions (pancreatic intraepithelial neoplasms); then, the expression decreased as the malignant grade progressed. We found that knockdown of KDM6B in PDAC cells promoted tumor sphere formation and increased peritoneal dissemination and liver metastasis in vivo. Microarray and chromatin immunoprecipitation analysis implicated CEBPA for aggressiveness induced by KDM6B knockdown. CEBPA knockdown recapitulated the phenotypic change of PDAC cells after KDM6B knockdown, which was reversed by forced expression of C/EBPα. Moreover, similar protein expression patterns of KDM6B and C/EBPα in human PDAC emphasized their functional correlation. Notably, pharmacological inhibition of the H3K27 methylase EZH2 in PDAC cells inhibited tumor sphere formation along with the upregulation of CEBPA expression, and this effect was impaired in KDM6B knockdown cells, highlighting the role for KDM6B in the activation of CEBPA. Together, our results propose a significant role for the KDM6B-C/EBPα axis in the PDAC phenotype.


Asunto(s)
Adenocarcinoma/genética , Proteínas Potenciadoras de Unión a CCAAT/biosíntesis , Proliferación Celular/genética , Histona Demetilasas con Dominio de Jumonji/genética , Neoplasias Pancreáticas/genética , Adenocarcinoma/patología , Proteínas Potenciadoras de Unión a CCAAT/genética , Línea Celular Tumoral , Proteína Potenciadora del Homólogo Zeste 2 , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Histona Demetilasas/genética , Histonas/genética , Humanos , Histona Demetilasas con Dominio de Jumonji/antagonistas & inhibidores , Neoplasias Pancreáticas/patología , Complejo Represivo Polycomb 2/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas p21(ras) , Proteínas ras/genética
14.
J Biol Chem ; 288(52): 36810-26, 2013 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-24202177

RESUMEN

Δ(9)-Tetrahydrocannabinol (THC), the major bioactive component of marijuana, has been shown to induce functional myeloid-derived suppressor cells (MDSCs) in vivo. Here, we studied the involvement of microRNA (miRNA) in this process. CD11b(+)Gr-1(+) MDSCs were purified from peritoneal exudates of mice administered with THC and used for genome-wide miRNA profiling. Expression of CD31 and Ki-67 confirmed that the THC-MDSCs were immature and proliferating. THC-induced MDSCs exhibited distinct miRNA expression signature relative to various myeloid cells and BM precursors. We identified 13 differentially expressed (>2-fold) miRNA in THC-MDSCs relative to control BM precursors. In silico target prediction for these miRNA and pathway analysis using multiple bioinformatics tools revealed significant overrepresentation of Gene Ontology clusters within hematopoiesis, myeloid cell differentiation, and regulation categories. Insulin-like growth factor 1 signaling involved in cell growth and proliferation, and myeloid differentiation pathways were among the most significantly enriched canonical pathways. Among the differentially expressed, miRNA-690 was highly overexpressed in THC-MDSCs (∼16-fold). Transcription factor CCAAT/enhancer-binding protein α (C/EBPα) was identified as a potential functional target of miR-690. Supporting this, C/EBPα expression was attenuated in THC-MDSCs as compared with BM precursors and exhibited an inverse relation with miR-690. miR-690 knockdown using peptide nucleic acid-antagomiR was able to unblock and significantly increase C/EBPα expression establishing the functional link. Further, CD11b(+)Ly6G(+)Ly6C(+) and CD11b(+)Ly6G(-)Ly6C(+) purified subtypes showed high levels of miR-690 with attenuated C/EBPα expression. Moreover, EL-4 tumor-elicited MDSCs showed increased miR-690 expression. In conclusion, miRNA are significantly altered during the generation of functional MDSC from BM. Select miRNA such as miR-690 targeting genes involved in myeloid expansion and differentiation likely play crucial roles in this process and therefore in cannabinoid-induced immunosuppression.


Asunto(s)
Analgésicos no Narcóticos/farmacología , Proteínas Potenciadoras de Unión a CCAAT/biosíntesis , Dronabinol/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Tolerancia Inmunológica/efectos de los fármacos , Células Mieloides/metabolismo , Animales , Antígenos de Diferenciación/biosíntesis , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/inmunología , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/inmunología , Células Cultivadas , Femenino , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Tolerancia Inmunológica/genética , Ratones , Células Mieloides/citología , Células Mieloides/inmunología
15.
Antimicrob Agents Chemother ; 58(11): 6717-23, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25155608

RESUMEN

Gene expression studies of subcutaneous adipose tissue may help to better understand the mechanisms behind body fat changes in HIV-infected patients who initiate antiretroviral therapy (ART). Here, we evaluated early changes in adipose tissue gene expression and their relationship to fat changes in ART-naive HIV-infected patients randomly assigned to initiate therapy with emtricitabine/tenofovir plus efavirenz (EFV) or ritonavir-boosted lopinavir (LPV/r). Patients had abdominal subcutaneous adipose tissue biopsies at baseline and week 16 and dual-energy-X-ray absorptiometry at baseline and weeks 16 and 48. mRNA changes of 11 genes involved in adipogenesis, lipid and glucose metabolism, mitochondrial energy, and inflammation were assessed through reverse transcription-quantitative PCR (RT-qPCR). Additionally, correlations between gene expression changes and fat changes were evaluated. Fat increased preferentially in the trunk with EFV and in the limbs with LPV/r (P < 0.05). After 16 weeks of exposure to the drug regimen, transcripts of CEBP/A, ADIPOQ, GLUT4, LPL, and COXIV were significantly down-regulated in the EFV arm compared to the LPV/r arm (P < 0.05). Significant correlations were observed between LPL expression change and trunk fat change at week 16 in both arms and between CEBP/A or COXIV change and trunk fat change at the same time point only in the EFV arm and not in the LPV/r arm. When combined with emtricitabine/tenofovir as standard backbone therapy, EFV and LPV/r induced differential early expression of genes involved in adipogenesis and energy metabolism. Moreover, these mRNA expression changes correlated with trunk fat change in the EFV arm. (This was a substudy of a randomized clinical trial [LIPOTAR study] registered at ClinicalTrials.gov under identifier NCT00759070.).


Asunto(s)
Adipogénesis/genética , Benzoxazinas/uso terapéutico , Composición Corporal/efectos de los fármacos , Lopinavir/uso terapéutico , Ritonavir/uso terapéutico , Grasa Subcutánea/citología , Síndrome de Inmunodeficiencia Adquirida/tratamiento farmacológico , Síndrome de Inmunodeficiencia Adquirida/virología , Adenina/análogos & derivados , Adenina/uso terapéutico , Adiponectina/biosíntesis , Adulto , Alquinos , Fármacos Anti-VIH/uso terapéutico , Proteínas Potenciadoras de Unión a CCAAT/biosíntesis , Ciclopropanos , Desoxicitidina/análogos & derivados , Desoxicitidina/uso terapéutico , Combinación de Medicamentos , Emtricitabina , Metabolismo Energético/genética , Femenino , Expresión Génica , Glucosa/metabolismo , Transportador de Glucosa de Tipo 4/biosíntesis , VIH-1/efectos de los fármacos , Humanos , Inflamación/genética , Metabolismo de los Lípidos/genética , Lipoproteína Lipasa/genética , Masculino , Organofosfonatos/uso terapéutico , Inhibidores de la Transcriptasa Inversa/uso terapéutico , Tenofovir
16.
Eur J Immunol ; 43(11): 2854-65, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23881867

RESUMEN

In response to aggravation by activated microglia, IL-13 can significantly enhance ER stress induction, apoptosis, and death via reciprocal signaling through CCAAT/enhancer-binding protein alpha (C/EBP-α) and C/EBP-beta (C/EBP-ß). This reciprocal signaling promotes neuronal survival. Since the induction of cyclooxygenase-2 (COX-2) and peroxisome proliferator-activated receptor gamma/heme oxygenase 1 (PPAR-γ/HO-1) by IL-13 plays a crucial role in the promotion of and protection from activated microglia, respectively; here, we investigated the role of IL-13 in regulating C/EBPs in activated microglia and determined its correlation with neuronal function. The results revealed that IL-13 significantly enhanced C/EBP-α/COX-2 expression and PGE2 production in LPS-treated microglial cells. Paradoxically, IL-13 abolished C/EBP-ß/PPAR-γ/HO-1 expression. IL-13 also enhanced ER stress-evoked calpain activation by promoting the association of C/EBP-ß and PPAR-γ. SiRNA-C/EBP-α effectively reversed the combined LPS-activated caspase-12 activation and IL-13-induced apoptosis. In contrast, siRNA-C/EBP-ß partially increased microglial cell apoptosis. By NeuN immunochemistry and CD11b staining, there was improvement in the loss of CA3 neuronal cells after intrahippocampal injection of IL-13. This suggests that IL-13-enhanced PLA2 activity regulates COX-2/PGE2 expression through C/EBP-α activation. In parallel, ER stress-related calpain downregulates the PPAR-γ/HO-1 pathway via C/EBP-ß and leads to aggravated death of activated microglia via IL-13, thereby preventing cerebral inflammation and neuronal injury.


Asunto(s)
Apoptosis , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Interleucina-13/metabolismo , Microglía/metabolismo , Neuronas/fisiología , Animales , Proteína beta Potenciadora de Unión a CCAAT/biosíntesis , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/biosíntesis , Proteínas Potenciadoras de Unión a CCAAT/genética , Calpaína/metabolismo , Caspasa 12/metabolismo , Células Cultivadas , Ciclooxigenasa 2/metabolismo , Dinoprostona/biosíntesis , Hemo-Oxigenasa 1/biosíntesis , Lipopolisacáridos , Proteínas de la Membrana/biosíntesis , PPAR gamma/metabolismo , Interferencia de ARN , ARN Interferente Pequeño , Ratas , Transducción de Señal
17.
Molecules ; 19(11): 18632-55, 2014 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-25405284

RESUMEN

Resveratrol is a non-flavonoid polyphenol which belongs to the stilbenes group and is produced naturally in several plants in response to injury or fungal attack. Resveratrol has been recently reported as preventing obesity. The present review aims to compile the evidence concerning the potential mechanisms of action which underlie the anti-obesity effects of resveratrol, obtained either in cultured cells lines and animal models. Published studies demonstrate that resveratrol has an anti-adipogenic effect. A good consensus concerning the involvement of a down-regulation of C/EBPα and PPARγ in this effect has been reached. Also, in vitro studies have demonstrated that resveratrol can increase apoptosis in mature adipocytes. Furthermore, different metabolic pathways involved in triacylglycerol metabolism in white adipose tissue have been shown to be targets for resveratrol. Both the inhibition of de novo lipogenesis and adipose tissue fatty acid uptake mediated by lipoprotein lipase play a role in explaining the reduction in body fat which resveratrol induces. As far as lipolysis is concerned, although this compound per se seems to be unable to induce lipolysis, it increases lipid mobilization stimulated by ß-adrenergic agents. The increase in brown adipose tissue thermogenesis, and consequently the associated energy dissipation, can contribute to explaining the body-fat lowering effect of resveratrol. In addition to its effects on adipose tissue, resveratrol can also acts on other organs and tissues. Thus, it increases mitochondriogenesis and consequently fatty acid oxidation in skeletal muscle and liver. This effect can also contribute to the body-fat lowering effect of this molecule.


Asunto(s)
Adipocitos/metabolismo , Antiinflamatorios no Esteroideos/uso terapéutico , Apoptosis/efectos de los fármacos , Lipogénesis/efectos de los fármacos , Obesidad , Estilbenos/uso terapéutico , Adipocitos/patología , Proteínas Potenciadoras de Unión a CCAAT/biosíntesis , Regulación hacia Abajo/efectos de los fármacos , Ácidos Grasos/metabolismo , Humanos , Hígado/metabolismo , Hígado/patología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Obesidad/patología , PPAR gamma/biosíntesis , Resveratrol
18.
Am J Physiol Gastrointest Liver Physiol ; 304(4): G420-7, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23257919

RESUMEN

Obesity, inflammation, and insulin resistance are closely linked. Substance P (SP), via its neurokinin 1 receptor (NK1R), mediates inflammatory and, possibly, neuroendocrine processes. We examined SP effects on lipid storage and cytokine production in 3T3-L1 adipocytes and adipose tissues. 3T3-L1 adipocytes and preadipocytes express NK1R, and 8 days of SP supplementation during differentiation to 3T3-L1 preadipocytes decreased lipid droplet accumulation. SP (10 nM, 24 h) increased lipolysis in primary adipocytes (138 ± 7%, P < 0.05) and reduced fatty acid uptake (-31 ± 7%, P < 0.05) and mRNA expression of the differentiation-related transcription factors peroxisome proliferator-activated receptor-γ type 2 (-64 ± 2%, P < 0.001) and CCAAT enhancer-binding protein (CEBP)-α (-65 ± 2%, P < 0.001) and the lipid storage genes fatty acid-binding protein type 4 (-59 ± 2%, P < 0.001) and diacylglycerol O-acyltransferase-1 (-45 ± 2%, P < 0.01) in 3T3-L1 adipocytes, while CD36, a fatty acid transporter (+82 ± 19%, P < 0.01), was augmented. SP increased secretion of complement C3 (148 ± 15%, P < 0.04), monocyte chemoattractant protein-1 (156 ± 16%, P < 0.03), and keratinocyte-derived chemokine (148 ± 18%, P = 0.045) in 3T3-L1 adipocytes and monocyte chemoattractant protein-1 (496 ± 142%, P < 0.02) and complement C3 (152 ± 25%, P < 0.04) in adipose tissue and primary adipocytes, respectively. These SP effects were accompanied by downregulation of insulin receptor substrate 1 (-82 ± 2%, P < 0.01) and GLUT4 (-76 ± 2%, P < 0.01) mRNA expression, and SP acutely blocked insulin-mediated stimulation of fatty acid uptake and Akt phosphorylation. Although adiponectin secretion was unchanged, mRNA expression was decreased (-86 ± 8%, P < 0.001). In humans, NK1R expression correlates positively with plasma insulin, fatty acid, and complement C3 and negatively with adiponectin, CEBPα, CEBPß, and peroxisome proliferator-activated receptor-γ mRNA expression in omental, but not subcutaneous, adipose tissue. Our results suggest that, beyond its neuroendocrine and inflammatory effects, SP could also be involved in targeting adipose tissue and influencing insulin resistance.


Asunto(s)
Adipocitos/metabolismo , Adipoquinas/biosíntesis , Sustancia P/fisiología , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Animales , Proteínas Potenciadoras de Unión a CCAAT/biosíntesis , Diferenciación Celular/efectos de los fármacos , Quimiocina CCL2 , Complemento C3/metabolismo , Diacilglicerol O-Acetiltransferasa/metabolismo , Proteínas de Unión a Ácidos Grasos/biosíntesis , Ácidos Grasos/metabolismo , Humanos , Proteínas Sustrato del Receptor de Insulina/biosíntesis , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Ratones , PPAR gamma/biosíntesis , Receptores de Neuroquinina-1/biosíntesis
19.
Cytokine ; 64(1): 138-45, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23911203

RESUMEN

We previously revealed that endogenous bone morphogenetic protein (Bmp) activity is required for lipid accumulation in 3T3-L1 adipocytes. The present study characterized the role of endogenous Bmp activity in preadipocytes. Endogenous Bmp activity was monitored by analyzing the level of phosphorylation of Smad1/5/8, downstream molecules in the Bmp pathway. Higher levels of phosphorylated Smad1/5/8 were detected in adipogenic cells but not in non-adipogenic cells prior to differentiation induction. The inhibition of the Bmp pathway during this period decreased the expression of Pparγ2 and C/ebpα, which are transcription factors responsible for adipocyte differentiation. The expression of these transcription factors were also down-regulated by Bmp4 knockdown. In addition, endogenous Bmp4 was required for the repression of Intrleukin-11 expression. Endogenous Bmp4 in preadipocytes is indispensable for the onset of the adipogenic program, and may help to maintain the preadipocytic state during adipocyte differentiation.


Asunto(s)
Adipocitos/citología , Adipogénesis , Proteína Morfogenética Ósea 4/metabolismo , Células 3T3 , Adipocitos/metabolismo , Animales , Proteína Morfogenética Ósea 2/antagonistas & inhibidores , Proteína Morfogenética Ósea 2/metabolismo , Proteína Morfogenética Ósea 4/biosíntesis , Proteína Morfogenética Ósea 4/genética , Proteínas Potenciadoras de Unión a CCAAT/biosíntesis , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Bovinos , Diferenciación Celular , Línea Celular , Linaje de la Célula , Interleucina-11/biosíntesis , Ratones , PPAR gamma/biosíntesis , PPAR gamma/metabolismo , Fosforilación/efectos de los fármacos , Pirazoles/farmacología , Pirimidinas/farmacología , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Proteína Smad8/metabolismo , Activación Transcripcional/efectos de los fármacos
20.
J Neuroinflammation ; 9: 165, 2012 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-22776069

RESUMEN

BACKGROUND: In physiological conditions, it is postulated that neurons control microglial reactivity through a series of inhibitory mechanisms, involving either cell contact-dependent, soluble-factor-dependent or neurotransmitter-associated pathways. In the current study, we focus on CD200R1, a microglial receptor involved in one of these cell contact-dependent mechanisms. CD200R1 activation by its ligand, CD200 (mainly expressed by neurons in the central nervous system),is postulated to inhibit the pro-inflammatory phenotype of microglial cells, while alterations in CD200-CD200R1 signalling potentiate this phenotype. Little is known about the regulation of CD200R1 expression in microglia or possible alterations in the presence of pro-inflammatory stimuli. METHODS: Murine primary microglial cultures, mixed glial cultures from wild-type and CCAAT/enhancer binding protein ß (C/EBPß)-deficient mice, and the BV2 murine cell line overexpressing C/EBPß were used to study the involvement of C/EBPß transcription factor in the regulation of CD200R1 expression in response to a proinflammatory stimulus (lipopolysaccharide (LPS)). Binding of C/EBPß to the CD200R1 promoter was determined by quantitative chromatin immunoprecipitation (qChIP). The involvement of histone deacetylase 1 in the control of CD200R1 expression by C/EBPß was also determined by co-immunoprecipitation and qChIP. RESULTS: LPS treatment induced a decrease in CD200R1 mRNA and protein expression in microglial cells, an effect that was not observed in the absence of C/EBPß. C/EBPß overexpression in BV2 cells resulted in a decrease in basal CD200R1 mRNA and protein expression. In addition, C/EBPß binding to the CD200R1 promoter was observed in LPS-treated but not in control glial cells, and also in control BV2 cells overexpressing C/EBPß. Finally, we observed that histone deacetylase 1 co-immunoprecipitated with C/EBPß and showed binding to a C/EBPß consensus sequence of the CD200R1 promoter in LPS-treated glial cells. Moreover, histone deacetylase 1 inhibitors reversed the decrease in CD200R1 expression induced by LPS treatment. CONCLUSIONS: CD200R1 expression decreases in microglial cells in the presence of a pro-inflammatory stimulus, an effect that is regulated, at least in part, by C/EBPß. Histone deacetylase 1 may mediate C/EBPß inhibition of CD200R1 expression, through a direct effect on C/EBPß transcriptional activity and/or on chromatin structure.


Asunto(s)
Antígenos de Superficie/biosíntesis , Proteínas Potenciadoras de Unión a CCAAT/biosíntesis , Regulación de la Expresión Génica , Microglía/metabolismo , Receptores de Superficie Celular/antagonistas & inhibidores , Receptores de Superficie Celular/biosíntesis , Animales , Antígenos de Superficie/genética , Proteína beta Potenciadora de Unión a CCAAT , Proteínas Potenciadoras de Unión a CCAAT/genética , Células Cultivadas , Técnicas de Cocultivo , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Orexina , Unión Proteica/fisiología , Receptores de Superficie Celular/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA