Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancers (Basel) ; 15(18)2023 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-37760525

RESUMO

Early detection of lung cancer is crucial for patient survival and treatment. Recent advancements in next-generation sequencing (NGS) analysis enable cell-free DNA (cfDNA) liquid biopsy to detect changes, like chromosomal rearrangements, somatic mutations, and copy number variations (CNVs), in cancer. Machine learning (ML) analysis using cancer markers is a highly promising tool for identifying patterns and anomalies in cancers, making the development of ML-based analysis methods essential. We collected blood samples from 92 lung cancer patients and 80 healthy individuals to analyze the distinction between them. The detection of lung cancer markers Cyfra21 and carcinoembryonic antigen (CEA) in blood revealed significant differences between patients and controls. We performed machine learning analysis to obtain AUC values via Adaptive Boosting (AdaBoost), Multi-Layer Perceptron (MLP), and Logistic Regression (LR) using cancer markers, cfDNA concentrations, and CNV screening. Furthermore, combining the analysis of all multi-omics data for ML showed higher AUC values compared with analyzing each element separately, suggesting the potential for a highly accurate diagnosis of cancer. Overall, our results from ML analysis using multi-omics data obtained from blood demonstrate a remarkable ability of the model to distinguish between lung cancer and healthy individuals, highlighting the potential for a diagnostic model against lung cancer.

2.
Exp Mol Med ; 55(6): 1232-1246, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37258580

RESUMO

SIRT1, a member of the mammalian sirtuin family, is a nicotinamide adenosine dinucleotide (NAD)-dependent deacetylase with key roles in aging-related diseases and cellular senescence. However, the mechanism by which SIRT1 protein homeostasis is controlled under senescent conditions remains elusive. Here, we revealed that SIRT1 protein is significantly downregulated due to ubiquitin-mediated proteasomal degradation during stress-induced premature senescence (SIPS) and that SIRT1 physically associates with anaphase-promoting complex/cyclosome (APC/C), a multisubunit E3 ubiquitin ligase. Ubiquitin-dependent SIRT1 degradation is stimulated by the APC/C coactivator Cdh1 and not by the coactivator Cdc20. We found that Cdh1 depletion impaired the SIPS-promoted downregulation of SIRT1 expression and reduced cellular senescence, likely through SIRT1-driven p53 inactivation. In contrast, AROS, a SIRT1 activator, reversed the SIRT1 degradation induced by diverse stressors and antagonized Cdh1 function through competitive interactions with SIRT1. Furthermore, our data indicate opposite roles for Cdh1 and AROS in the epigenetic regulation of the senescence-associated secretory phenotype genes IL-6 and IL-8. Finally, we demonstrated that pinosylvin restores downregulated AROS (and SIRT1) expression levels in bleomycin-induced mouse pulmonary senescent tissue while repressing bleomycin-promoted Cdh1 expression. Overall, our study provides the first evidence of the reciprocal regulation of SIRT1 stability by APC/C-Cdh1 and AROS during stress-induced premature senescence, and our findings suggest pinosylvin as a potential senolytic agent for pulmonary fibrosis.


Assuntos
Epigênese Genética , Sirtuína 1 , Animais , Camundongos , Ciclossomo-Complexo Promotor de Anáfase/genética , Ciclossomo-Complexo Promotor de Anáfase/metabolismo , Proteínas de Ciclo Celular/metabolismo , Senescência Celular , Sirtuína 1/genética , Sirtuína 1/metabolismo , Ubiquitina/metabolismo , Ubiquitinação
3.
Nutrients ; 15(7)2023 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-37049636

RESUMO

Shikonin, a natural ingredient produced by Lithospermum erythrorhizon, has anti-inflammatory, anti-cancer, and anti-obesity effects. It also inhibits adipocyte differentiation; however, the underlying molecular and epigenetic mechanisms remain unclear. We performed RNA-sequencing of shikonin-treated 3T3-L1 cells. Gene ontology and gene set enrichment analysis showed that shikonin is significantly associated with genes related to adipogenesis, histone modification, and PPARγ. Shikonin treatment downregulated the mRNA expression of PPARγ-responsive genes and rosiglitazone-induced transcriptional activity of PPARγ. Microscale thermophoresis assays showed a KD value 1.4 ± 0.13 µM for binding between shikonin and PPARγ. Glutathione S-transferase pull-down assays exhibited that shikonin blocked the rosiglitazone-dependent association of PPARγ with its coactivator CBP. In addition, shikonin decreased the enrichment of the active histone code H3K4me3 and increased the repressive code H3K27me3 of PPARγ target promoters. Shikonin is a PPARγ antagonist that suppresses adipogenesis by regulating the enrichment of histone codes during adipogenesis. Therefore, it may be used to treat obesity-related disorders via epigenetic changes.


Assuntos
Histonas , PPAR gama , Camundongos , Animais , PPAR gama/genética , PPAR gama/metabolismo , Histonas/metabolismo , Rosiglitazona/metabolismo , Rosiglitazona/farmacologia , Metilação , Adipócitos , Adipogenia , Diferenciação Celular , Células 3T3-L1
4.
Biochem Biophys Res Commun ; 617(Pt 1): 48-54, 2022 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-35679710

RESUMO

We previously demonstrated that kaempferol, a flavonoid present in various herbs, inhibits adipogenesis by repressing peroxisome proliferator-activated receptor γ (PPARγ) activity. Here, we focused on elucidation of the underlying mechanism using genome-wide tools. First, RNA sequencing (RNA-seq) analysis showed downregulation of genes involved in adipogenesis in response to kaempferol. Subsequent ChIP assays revealed that kaempferol regulates the expression of adipogenic (Adipoq, Fabp4, Lpl) genes by modulating enrichment of active H3K4me3 and repressive H3K27me3 histone codes on target promoters. Second, we performed ChIP sequencing analysis of active H3K4me3, and co-analysis with RNA-seq identified PPARγ responsive sites in genes downregulated by kaempferol, in terms of expression and H3K4me3 deposition. Third, direct kaempferol binding to PPARγ, for which the KD value was 44.54 µM, was determined by microscale thermophoresis. Further RT-qPCR and GST pull-down assays demonstrated that kaempferol antagonizes rosiglitazone-induced PPARγ activation and impairs the rosiglitazone-dependent interaction between PPARγ and its coactivator CBP. Overall, our data suggest that kaempferol, as a PPARγ antagonist, mediates epigenetic repression of lipid accumulation by regulating histone methylation, and could serve as a candidate epigenetic drug to treat obesity-related diseases.


Assuntos
Adipogenia , PPAR gama , Células 3T3-L1 , Adipócitos/metabolismo , Animais , Histonas/metabolismo , Quempferóis/farmacologia , Metilação , Camundongos , PPAR gama/genética , PPAR gama/metabolismo , Rosiglitazona
5.
Phytother Res ; 33(9): 2429-2439, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31359554

RESUMO

Previously, we reported that piperine, one of the major pungent components in black pepper, attenuates adipogenesis by repressing PPARγ activity in 3T3-L1 preadipocytes. However, the epigenetic mechanisms underlying this activity remain unexplored. Here, gene set enrichment analysis using microarray data indicated that there was significant downregulation of adipogenesis-associated and PPARγ target genes and upregulation of genes bound with H3K27me3 in response to piperine. As shown by Gene Ontology analysis, the upregulated genes are related to lipid oxidation and polycomb repressive complex 2 (PRC2). Chromatin immunoprecipitation assays revealed that PPARγ (and its coactivators), H3K4me3, and H3K9ac were less enriched at the PPAR response element of three adipogenic genes, whereas increased accumulation of H3K9me2, H3K27me3, and Ezh2 was found, which likely led to the reduced gene expression. Further analysis using three lipolytic genes revealed the opposite enrichment pattern of H3K4me3 and H3K27me3 at the Ezh2 binding site. Treatment with GSK343, an Ezh2 inhibitor, elevated lipolytic gene expression by decreasing the enrichment of H3K27me3 during adipogenesis, which confirms that Ezh2 plays a repressive role in lipolysis. Overall, these results suggest that piperine regulates the expression of adipogenic and lipolytic genes by dynamic regulation of histone modifications, leading to the repression of adipocyte differentiation.


Assuntos
Adipócitos/efeitos dos fármacos , Adipogenia/fisiologia , Alcaloides/uso terapêutico , Benzodioxóis/uso terapêutico , Código das Histonas/fisiologia , Piperidinas/uso terapêutico , Alcamidas Poli-Insaturadas/uso terapêutico , Alcaloides/farmacologia , Benzodioxóis/farmacologia , Diferenciação Celular , Humanos , Piperidinas/farmacologia , Alcamidas Poli-Insaturadas/farmacologia
6.
Biochem Biophys Res Commun ; 508(3): 907-913, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30545639

RESUMO

Additional sex comb-like1 (Asxl1) is known as a chromatin modulator that plays dual functions in transcriptional regulation depending on the cell type. Recent studies using Asxl1 knockout mice revealed that Asxl1 is important for the proliferation and differentiation of hematopoietic progenitor cells, and the development of organs. Although we previously reported Asxl1 as a Sox2 target gene, its function in embryonic stem cells (ESCs) remains largely unknown. For this purpose, we isolated ESCs from the blastocyst inner cell mass of Asxl1-/- mice. Asxl1 deficiency in ESCs exhibited no effect on cell proliferation, expression of core pluripotent transcription factors, or alkaline phosphatase activity, suggesting dispensability of Asxl1 for self-renewal of ESCs. By contrast, the differentiation of Asxl1-/- ESCs was significantly affected as shown by size reductions of embryoid bodies accompanied with apoptosis, aberrant expression of differentiation genes, downregulation of bivalent neurogenesis genes, and abnormal axon formation in neurons. Overall, our findings indicated that Asxl1 played a critical role in regulating genes associated with neural differentiation without affecting self-renewal of mouse ESCs.


Assuntos
Células-Tronco Embrionárias/fisiologia , Neurogênese/genética , Proteínas Repressoras/fisiologia , Animais , Axônios/ultraestrutura , Células Cultivadas , Corpos Embrioides/citologia , Regulação da Expressão Gênica , Técnicas de Inativação de Genes , Camundongos , Proteínas Repressoras/genética
7.
Cell Death Dis ; 9(11): 1118, 2018 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-30389914

RESUMO

Although additional sex combs-like 1 (ASXL1) has been extensively described in hematologic malignancies, little is known about the molecular role of ASXL1 in organ development. Here, we show that Asxl1 ablation in mice results in postnatal lethality due to cyanosis, a respiratory failure. This lung defect is likely caused by higher proliferative potential and reduced expression of surfactant proteins, leading to reduced air space and defective lung maturation. By microarray analysis, we identified E2F1-responsive genes, including Nmyc, as targets repressed by Asxl1. Nmyc and Asxl1 are reciprocally expressed during the fetal development of normal mouse lungs, whereas Nmyc downregulation is impaired in Asxl1-deficient lungs. Together with E2F1 and ASXL1, host cell factor 1 (HCF-1), purified as an Asxl1-bound protein, is recruited to the E2F1-binding site of the Nmyc promoter. The interaction occurs between the C-terminal region of Asxl1 and the N-terminal Kelch domain of HCF-1. Trimethylation (me3) of histone H3 lysine 27 (H3K27) is enriched in the Nmyc promoter upon Asxl1 overexpression, whereas it is downregulated in Asxl1-deleted lung and -depleted A549 cells, similar to H3K9me3, another repressive histone marker. Overall, these findings suggest that Asxl1 modulates proliferation of lung epithelial cells via the epigenetic repression of Nmyc expression, deficiency of which may cause hyperplasia, leading to dyspnea.


Assuntos
Fator de Transcrição E2F1/genética , Repressão Epigenética , Células Epiteliais/metabolismo , Pulmão/metabolismo , Proteína Proto-Oncogênica N-Myc/genética , Proteínas Repressoras/genética , Insuficiência Respiratória/genética , Células A549 , Animais , Fator de Transcrição E2F1/metabolismo , Embrião de Mamíferos , Células Epiteliais/patologia , Feto , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Genes Letais , Células HEK293 , Histonas/genética , Histonas/metabolismo , Fator C1 de Célula Hospedeira/genética , Fator C1 de Célula Hospedeira/metabolismo , Humanos , Pulmão/crescimento & desenvolvimento , Pulmão/patologia , Camundongos , Camundongos Knockout , Proteína Proto-Oncogênica N-Myc/metabolismo , Organogênese/genética , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Repressoras/deficiência , Insuficiência Respiratória/metabolismo , Insuficiência Respiratória/patologia , Transdução de Sinais
8.
Cell Death Dis ; 8(12): 3201, 2017 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-29233982

RESUMO

Peroxisome proliferator-activated receptor γ (PPARγ) is the master regulator of adipocyte differentiation and is closely linked to the development of obesity. Despite great progress in elucidating the transcriptional network of PPARγ, epigenetic regulation of this pathway by histone modification remains elusive. Here, we found that CDK2-associated cullin 1 (CACUL1), identified as a novel SIRT1 interacting protein, directly bound to PPARγ through the co-repressor nuclear receptor (CoRNR) box 2 and repressed the transcriptional activity and adipogenic potential of PPARγ. Upon CACUL1 depletion, less SIRT1 and more LSD1 were recruited to the PPARγ-responsive gene promoter, leading to increased histone H3K9 acetylation, decreased H3K9 methylation, and PPARγ activation during adipogenesis in 3T3-L1 cells. These findings were reversed upon fasting or resveratrol treatment. Further, gene expression profiling using RNA sequencing supported the repressive role of CACUL1 in PPARγ activation and fat accumulation. Finally, we confirmed CACUL1 function in human adipose-derived stem cells. Overall, our data suggest that CACUL1 tightly regulates PPARγ signaling through the mutual opposition between SIRT1 and LSD1, providing insight into its potential use for anti-obesity treatment.


Assuntos
Adipócitos/metabolismo , Adipogenia/genética , Proteínas de Transporte/genética , Epigênese Genética , Histona Desmetilases/genética , PPAR gama/genética , Sirtuína 1/genética , Células 3T3-L1 , Adipócitos/citologia , Adipócitos/efeitos dos fármacos , Adipogenia/efeitos dos fármacos , Animais , Proteínas de Transporte/metabolismo , Diferenciação Celular , Proteínas Correpressoras/genética , Proteínas Correpressoras/metabolismo , Proteínas Culina , Células HCT116 , Células HEK293 , Histona Desmetilases/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Camundongos , PPAR gama/metabolismo , Resveratrol , Análise de Sequência de RNA , Transdução de Sinais , Sirtuína 1/metabolismo , Estilbenos/farmacologia
9.
Sci Rep ; 7(1): 5198, 2017 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-28701722

RESUMO

Although ASXL1 mutations are frequently found in human diseases, including myeloid leukemia, the cell proliferation-associated function of ASXL1 is largely unknown. Here, we explored the molecular mechanism underlying the growth defect found in Asxl1-deficient mouse embryonic fibroblasts (MEFs). We found that Asxl1, through amino acids 371 to 655, interacts with the kinase domain of AKT1. In Asxl1-null MEFs, IGF-1 was unable to induce AKT1 phosphorylation and activation; p27Kip1, which forms a ternary complex with ASXL1 and AKT1, therefore remained unphosphorylated. Hypophosphorylated p27Kip1 is able to enter the nucleus, where it prevents the phosphorylation of Rb; this ultimately leads to the down-regulation of E2F target genes as confirmed by microarray analysis. We also found that senescence-associated (SA) genes were upregulated and that SA ß-gal staining was increased in Asxl1 -/- MEFs. Further, the treatment of an AKT inhibitor not only stimulated nuclear accumulation of p27Kip1 leading to E2F inactivation, but also promoted senescence. Finally, Asxl1 disruption augmented the expression of p16Ink4a as result of the defect in Asxl1-Ezh2 cooperation. Overall, our study provides the first evidence that Asxl1 both activates the AKT-E2F pathway and cooperates with Ezh2 through direct interactions at early embryonic stages, reflecting that Asxl1 disruption causes cellular senescence.


Assuntos
Senescência Celular , Fatores de Transcrição E2F/antagonistas & inibidores , Embrião de Mamíferos/patologia , Proteína Potenciadora do Homólogo 2 de Zeste/antagonistas & inibidores , Fibroblastos/patologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Repressoras/fisiologia , Animais , Proliferação de Células , Células Cultivadas , Fatores de Transcrição E2F/genética , Fatores de Transcrição E2F/metabolismo , Embrião de Mamíferos/metabolismo , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Fibroblastos/metabolismo , Camundongos , Camundongos Knockout , Fosforilação , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
10.
Cancer Lett ; 403: 144-151, 2017 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-28634046

RESUMO

Elevated expression of preferentially expressed antigen in melanoma (PRAME) has been implicated in disease progression in a variety of cancers. However, the mechanisms underlying the transcriptional regulation of PRAME remain largely unexplored. Initially, we observed that PRAME was elevated in proportion to the malignant potential of melanoma cells. From the in silico prediction of PRAME gene structure, we identified the putative myeloid zinc finger 1 (MZF1) binding sites, which overlap with a CpG-rich region located in the first intron. The transcription factor MZF1 increased PRAME expression via its direct binding to the intron DNA. Upon treatment with a DNA methylation inhibitor, 5-aza-2'-deoxycitidine (5-azaC), together with ectopic expression of MZF1, PRAME expression was significantly enhanced at both the protein and mRNA levels. More pronounced MZF1 binding to the PRAME DNA was observed in the presence of 5-azaC. DNA methylation was inversely correlated with PRAME expression in melanoma cells. Finally, we observed that MZF1, like PRAME, promotes the colony-forming ability in melanoma cells. Overall, our findings suggest that MZF1, via stimulation of PRAME expression, may be a potential prognostic and therapeutic target in melanoma.


Assuntos
Antígenos de Neoplasias/metabolismo , Metilação de DNA , Epigênese Genética , Fatores de Transcrição Kruppel-Like/metabolismo , Melanoma/metabolismo , Neoplasias Cutâneas/metabolismo , Antígenos de Neoplasias/genética , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Sítios de Ligação , Proliferação de Células , Ilhas de CpG , Metilação de DNA/efeitos dos fármacos , Metilases de Modificação do DNA/antagonistas & inibidores , Metilases de Modificação do DNA/metabolismo , Decitabina , Inibidores Enzimáticos/farmacologia , Epigênese Genética/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , Células HCT116 , Humanos , Fatores de Transcrição Kruppel-Like/genética , Melanoma/genética , Melanoma/patologia , Regiões Promotoras Genéticas , Ligação Proteica , Interferência de RNA , Transdução de Sinais , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Transcrição Gênica , Transfecção , Regulação para Cima
11.
Biochem Biophys Res Commun ; 454(4): 479-85, 2014 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-25450400

RESUMO

Among the members of the additional sex comb-like (ASXL) family, ASXL3 remains unexplored. Here, we showed that ASXL3 interacts with HP1α and LSD1, leading to transcriptional repression. We determined that ASXL3 depletion augments the ligand-induced transcriptional activities of LXRα and TRß, which were repressed by ASXL3 overexpression. The ligand-dependent interactions of ASXL3 with LXRα and TRß were demonstrated by the GST pull-down and immunoprecipitation analyses. We confirmed that ASXL3 suppresses the expression of LXRα target genes through its recruitment to the LXR-response elements. Finally, we observed that lipid accumulation in Hep3B cells is downregulated upon ASXL3 overexpression but upregulated upon ASXL3 depletion. Overall, our data suggest that ASXL3 is another corepressor of LXRα, promoting to the regulation of lipid homeostasis.


Assuntos
Regulação para Baixo , Receptores X do Fígado/antagonistas & inibidores , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Homólogo 5 da Proteína Cromobox , Células HEK293 , Homeostase , Humanos , Ligantes , Metabolismo dos Lipídeos , Receptores X do Fígado/genética , Receptores X do Fígado/metabolismo , Proteínas Repressoras/deficiência , Proteínas Repressoras/genética , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Células Tumorais Cultivadas
12.
Biochem Biophys Res Commun ; 443(2): 489-94, 2014 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-24321552

RESUMO

Liver X receptor alpha (LXRα), a member of the nuclear receptor superfamily, plays a pivotal role in hepatic cholesterol and lipid metabolism, regulating the expression of genes associated with hepatic lipogenesis. The additional sex comb-like (ASXL) family was postulated to regulate chromatin function. Here, we investigate the roles of ASXL1 and ASXL2 in regulating LXRα activity. We found that ASXL1 suppressed ligand-induced LXRα transcriptional activity, whereas ASXL2 increased LXRα activity through direct interaction in the presence of the ligand. Chromatin immunoprecipitation (ChIP) assays showed ligand-dependent recruitment of ASXLs to ABCA1 promoters, like LXRα. Knockdown studies indicated that ASXL1 inhibits, while ASXL2 increases, lipid accumulation in H4IIE cells, similar to their roles in transcriptional regulation. We also found that ASXL1 expression increases under fasting conditions, and decreases in insulin-treated H4IIE cells and the livers of high-fat diet-fed mice. Overall, these results support the reciprocal role of the ASXL family in lipid homeostasis through the opposite regulation of LXRα.


Assuntos
Hepatócitos/metabolismo , Lipogênese/fisiologia , Receptores Nucleares Órfãos/metabolismo , Proteínas Repressoras/metabolismo , Animais , Linhagem Celular , Regulação da Expressão Gênica , Receptores X do Fígado , Camundongos , Ratos
13.
FEBS Lett ; 587(1): 17-22, 2013 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-23178685

RESUMO

ERα, a critical transcriptional factor for breast cancer proliferation, is regulated by a complex binding repertoire that includes coactivators and corepressors. Here, we identified a novel class of ERα coregulator called CAC1. The CoRNR box of CAC1 was required for the binding to and inactivation of ERα. CAC1 also associated with histone demethylase LSD1 and suppressed LSD1-enhanced ERα activity. CAC1 impaired recruitment of ERα and LSD1 to the ERα-responsive promoter, leading to greater H3K9me3 accumulation. This effect was reversed by CAC1 depletion. Finally, CAC1 increased paclitaxel-induced cell death in ERα-positive MCF7 cells, which are paclitaxel-resistant. Overall, our study provides the first evidence that CAC1, associated with LSD1, functions as an ERα corepressor, implicating a potential antitumor target in ERα-positive breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , Proteínas Correpressoras/metabolismo , Proteínas Culina/metabolismo , Receptor alfa de Estrogênio/metabolismo , Histona Desmetilases/metabolismo , Proteínas de Neoplasias/metabolismo , Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/enzimologia , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proteínas Correpressoras/antagonistas & inibidores , Proteínas Correpressoras/genética , Proteínas Culina/antagonistas & inibidores , Proteínas Culina/genética , Resistencia a Medicamentos Antineoplásicos , Receptor alfa de Estrogênio/genética , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes Reporter/efeitos dos fármacos , Histona Desmetilases/genética , Histonas/metabolismo , Humanos , Proteínas Mutantes/antagonistas & inibidores , Proteínas Mutantes/metabolismo , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Paclitaxel/farmacologia , Regiões Promotoras Genéticas/efeitos dos fármacos , Protamina Quinase/metabolismo , Interferência de RNA , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/metabolismo , Fator Trefoil-1 , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
14.
Biol Pharm Bull ; 35(9): 1525-33, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22975504

RESUMO

Rhizoma Polygonati falcatum (RPF) has been used as a traditional herbal medicine in Asia, because of its anti-hyperglycemic, anti-triglycemic, and anti-tumor activity. In this study, we determined the anti-adipogenic potential of RPF extract and its component kaempferol in 3T3-L1 adipocytes, and the underlying molecular mechanism(s) using microarray analysis. Adipocyte differentiation of 3T3-L1 cells was significantly impaired by RPF extract and kaempferol as monitored by Oil Red O staining and quantitative measurement of lipid accumulation. Additionally, the mRNA expression of adipogenesis genes decreased on treatment with kaempferol. The role of kaempferol at the genome-wide level was further assessed by a microarray approach. Our analysis indicated that kaempferol decreased the expression of adipogenic transcription factors (Pparγ, Cebpß, Srebp1, Rxrß, Lxrß, Rorα) and genes involved in triglyceride biosynthesis (Gpd1, Agpat2, Dgat2), while increasing lipolysis-related genes, such as Tnfα, Lsr, and Cel. Finally, co-transfection assays using luciferase reporter gene and reverse transcription-polymerase chain reaction (RT-PCR) analysis using peroxisome proliferator-activated receptor-γ (PPARγ) target genes indicated that kaempferol significantly repressed rosiglitazone-induced PPARγ transcriptional activity. Overall, our data suggests that kaempferol, a major component of RPF, may be beneficial in obesity, by reducing adipogenesis and balancing lipid homeostasis partly through the down-regulation of PPARγ.


Assuntos
Adipócitos/efeitos dos fármacos , Adipogenia/efeitos dos fármacos , Fármacos Antiobesidade/farmacologia , Medicamentos de Ervas Chinesas/farmacologia , Quempferóis/farmacologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Polygonatum/química , Células 3T3-L1 , Adipócitos/metabolismo , Adipogenia/genética , Animais , Fármacos Antiobesidade/uso terapêutico , Medicamentos de Ervas Chinesas/uso terapêutico , Homeostase , Hipoglicemiantes/farmacologia , Hipoglicemiantes/uso terapêutico , Quempferóis/uso terapêutico , Metabolismo dos Lipídeos/genética , Lipólise/efeitos dos fármacos , Lipólise/genética , Camundongos , Análise em Microsséries , Obesidade/genética , Obesidade/metabolismo , Obesidade/prevenção & controle , PPAR gama/metabolismo , Fitoterapia , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Rizoma , Rosiglitazona , Tiazolidinedionas/farmacologia , Fatores de Transcrição/metabolismo , Triglicerídeos/biossíntese , Triglicerídeos/genética
15.
Biochem Pharmacol ; 84(11): 1501-10, 2012 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-23000915

RESUMO

This study investigated the role of piperine in the transcriptional regulation of liver X receptor α (LXRα) and the effects of dietary piperine on high-fat diet (HFD)-induced hepatic steatosis and insulin resistance in mice. Furthermore, we explored the potential molecular mechanisms through which the protective effects of piperine may work. In the present study, piperine significantly reduced ligand-induced LXRα activity in a dose-dependent manner and gradually disrupted the interaction between ligand-bound LXRα and GST-CBP. In mice, an HFD supplemented with 0.05% piperine (PSD) significantly decreased body and liver weight as well as plasma and hepatic lipid levels. In agreement with our in vitro study, in mice fed an HFD, dietary piperine markedly decreased LXRα mRNA expression and its lipogenic target genes (i.e., SREBP1c, ChREBPα, FAS, and CD36). Piperine also significantly decreased plasma insulin and glucose concentrations, while increasing insulin sensitivity in mice fed an HFD. In addition, piperine downregulated the expression of genes involved in ER stress, including GRP78, activating transcription factor 6, and eukaryotic translation initiation factor 2α, and upregulated GLUT2 translocation from the cytosol to the plasma membrane in the livers of PSD mice. Piperine antagonized LXRα transcriptional activity by abolishing the interaction of ligand-bound LXRα with the co-activator CBP. The effects of piperine on hepatic lipid accumulation were likely regulated via alterations in LXRα-mediated lipogenesis in mice fed an HFD. Dietary piperine also led to reduced ER stress and increased insulin sensitivity and prevented hepatic insulin resistance in mice fed the HFD.


Assuntos
Alcaloides/farmacologia , Benzodioxóis/farmacologia , Gorduras na Dieta/administração & dosagem , Fígado Gorduroso/prevenção & controle , Insulina/metabolismo , Piperidinas/farmacologia , Alcamidas Poli-Insaturadas/farmacologia , Transdução de Sinais , Alcaloides/administração & dosagem , Animais , Sequência de Bases , Benzodioxóis/administração & dosagem , Western Blotting , Primers do DNA , Chaperona BiP do Retículo Endoplasmático , Células HEK293 , Humanos , Camundongos , Piperidinas/administração & dosagem , Reação em Cadeia da Polimerase , Alcamidas Poli-Insaturadas/administração & dosagem
16.
J Agric Food Chem ; 60(15): 3853-60, 2012 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-22463744

RESUMO

This study investigated the antiadipogenic activity of black pepper extract and its constituent piperine in 3T3-L1 preadipocytes as well as the underlying molecular mechanisms. Both black pepper extract and piperine, without affecting cytotoxicity, strongly inhibited the adipocyte differentiation of 3T3-L1 cells. The mRNA expression of the master adipogenic transcription factors, PPARγ, SREBP-1c, and C/EBPß, was markedly decreased. Intriguingly, mRNA levels of PPARγ target genes were also down-regulated. Moreover, a luciferase reporter assay indicated that pipierine significantly represses the rosiglitazone-induced PPARγ transcriptional activity. Finally, GST-pull down assays demonstrated that piperine disrupts the rosiglitazone-dependent interaction between PPARγ and coactivator CBP. Genome-wide analysis using microarray further supports the role of piperine in regulating genes associated with lipid metabolism. Overall, these results suggest that piperine, a major component of black pepper, attenuates fat cell differentiation by down-regulating PPARγ activity as well as suppressing PPARγ expression, thus leading to potential treatment for obesity-related diseases.


Assuntos
Adipogenia/efeitos dos fármacos , Alcaloides/farmacologia , Benzodioxóis/farmacologia , Regulação para Baixo/efeitos dos fármacos , PPAR gama/antagonistas & inibidores , PPAR gama/metabolismo , Piper nigrum/química , Piperidinas/farmacologia , Extratos Vegetais/farmacologia , Alcamidas Poli-Insaturadas/farmacologia , Células 3T3-L1 , Adipócitos/citologia , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Camundongos , PPAR gama/genética
17.
Biochem Biophys Res Commun ; 404(1): 239-44, 2011 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-21110951

RESUMO

Retinoic acid (RA) plays a role in cancer therapy. However, its long-term treatment is hindered by the acquired resistance which is not fully understood. Our previous study indicated that the transcriptional activity of RA receptor (RAR) is enhanced by association of MED25 with CREB-binding protein (CBP) through the PTOV domain, which is also present in prostate tumor over-expressed protein 1 (PTOV1). Here, we show that MED25 and PTOV1 reciprocally regulate RAR transcriptional activity through competitive bindings to CBP and opposite regulation of CBP recruitment to the RA-responsive gene promoter. Finally, we demonstrate that MED25 and PTOV1 differentially modulate RA sensitivity in cancer cells depending on their expression levels, suggesting a potential molecular mechanism underlying RA resistance which frequently emerges during cancer treatments.


Assuntos
Antineoplásicos/farmacologia , Biomarcadores Tumorais/metabolismo , Resistencia a Medicamentos Antineoplásicos/genética , Complexo Mediador/antagonistas & inibidores , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Receptores do Ácido Retinoico/metabolismo , Ativação Transcricional , Tretinoína/farmacologia , Proteína de Ligação a CREB/metabolismo , Linhagem Celular , Cromatina/metabolismo , Imunoprecipitação da Cromatina , Humanos , Complexo Mediador/metabolismo , Neoplasias/genética
18.
J Biol Chem ; 286(2): 1354-63, 2011 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-21047783

RESUMO

Our previous studies have suggested that the mammalian additional sex comb-like 1 protein functions as a coactivator or repressor of retinoic acid receptors in a cell-specific manner. Here, we investigated the roles of additional sex comb-like 1 proteins in regulating peroxisome proliferator-activated receptors (PPARs). In pulldown assays in vitro and in immunoprecipitation assays in vivo, ASXL1 and its paralog, ASXL2, interacted with PPARα and PPARγ. In 3T3-L1 preadipocyte cells, overexpression of ASXL1 inhibited the induction of PPARγ activity by rosiglitazone, as shown by transcription assays, and completely suppressed adipogenesis, as shown by Oil Red O staining. In contrast, overexpression of ASXL2 greatly enhanced rosiglitazone-induced PPARγ activity and enhanced adipogenesis. Deletion of the heterochromatin protein 1 (HP1)-binding domain from ASXL1 caused the mutant protein to enhance adipogenesis similarly to ASXL2, indicating that HP1 binding is required for the adipogenesis-suppressing activity of ASXL1. Adipocyte differentiation was associated with a gradual decrease in ASXL1 expression but did not affect ASXL2 expression. Knockdown of ASXL1 and ASXL2 had reciprocal effects on adipogenesis. In chromatin immunoprecipitation assays in 3T3-L1 cells, ASXL1 occupied the promoter of the PPARγ target gene aP2 together with HP1α and Lys-9-methylated histone H3, whereas ASXL2 occupied the aP2 promoter together with histone-lysine N-methyltransferase MLL1 and Lys-9-acetylated and Lys-4-methylated H3 histones. Finally, microarray analysis demonstrated that ASXL1 represses, whereas ASXL2 increases, the expression of adipogenic genes, most of which are PPARγ targets. These results suggest that members of the additional sex comb-like family provide complex regulation of adipogenesis via differential modulation of PPARγ activity.


Assuntos
Adipogenia/fisiologia , PPAR gama/genética , Proteínas Repressoras/metabolismo , Ativação Transcricional/fisiologia , Células 3T3-L1 , Animais , Cromatina/fisiologia , Homólogo 5 da Proteína Cromobox , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , PPAR gama/metabolismo , Peptídeos , Regiões Promotoras Genéticas/fisiologia , Proteínas Repressoras/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
19.
J Biol Chem ; 285(44): 34269-78, 2010 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-20736163

RESUMO

In most mammalian cells, the retinoic acid receptor (RAR) is nuclear rather than cytoplasmic, regardless of its cognate ligand, retinoic acid (RA). In testis Sertoli cells, however, RAR is retained in the cytoplasm and moves to the nucleus only when RA is supplied. This led us to identify a protein that regulates the translocation of RAR. From yeast two-hybrid screening, we identified a novel RAR-interacting protein called CART1 (cytoplasmic adaptor for RAR and TR). Systematic interaction assays using deletion mutants showed that the C-terminal CoRNR box of CART1 was responsible for the interaction with the NCoR binding region of RAR and TR. Such interaction was impaired in the presence of ligand RA, as further determined by GST pulldown assays in vitro and immunoprecipitation assays in vivo. Fluorescence microscopy showed that unliganded RAR was captured by CART1 in the cytoplasm, whereas liganded RAR was liberated and moved to the nucleus. Overexpression of CART1 blocked the transcriptional repressing activity of unliganded apoRAR, mediated by corepressor NCoR in the nucleus. CART1 siRNA treatment in a mouse Sertoli cell line, TM4, allowed RAR to move to the nucleus and blocked the derepressing function of CART1, suggesting that CART1 might be a cytoplasmic, testis-specific derepressor of RAR.


Assuntos
Citoplasma/metabolismo , Regulação da Expressão Gênica , Receptores do Ácido Retinoico/metabolismo , Receptores dos Hormônios Tireóideos/metabolismo , Fatores de Transcrição/fisiologia , Tretinoína/química , Animais , Núcleo Celular/metabolismo , Humanos , Masculino , Camundongos , Microscopia de Fluorescência/métodos , Células NIH 3T3 , Células de Sertoli/metabolismo , Testículo/metabolismo , Fatores de Transcrição/química , Técnicas do Sistema de Duplo-Híbrido
20.
J Biol Chem ; 285(1): 18-29, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19880879

RESUMO

We previously suggested that ASXL1 (additional sex comb-like 1) functions as either a coactivator or corepressor for the retinoid receptors retinoic acid receptor (RAR) and retinoid X receptor in a cell type-specific manner. Here, we provide clues toward the mechanism underlying ASXL1-mediated repression. Transfection assays in HEK293 or H1299 cells indicated that ASXL1 alone possessing autonomous transcriptional repression activity significantly represses RAR- or retinoid X receptor-dependent transcriptional activation, and the N-terminal portion of ASXL1 is responsible for the repression. Amino acid sequence analysis identified a consensus HP1 (heterochromatin protein 1)-binding site (HP1 box, PXVXL) in that region. Systematic in vitro and in vivo assays revealed that the HP1 box in ASXL1 is critical for the interaction with the chromoshadow domain of HP1. Transcription assays with HP1 box deletion or HP1alpha knockdown indicated that HP1alpha is required for ASXL1-mediated repression. Furthermore, we found a direct interaction of ASXL1 with histone H3 demethylase LSD1 through the N-terminal region nearby the HP1-binding site. ASXL1 binding to LSD1 was greatly increased by HP1alpha, resulting in the formation of a ternary complex. LSD1 cooperates with ASXL1 in transcriptional repression, presumably by removing H3K4 methylation, an active histone mark, but not H3K9 methylation, a repressive histone mark recognized by HP1. This possibility was supported by chromatin immunoprecipitation assays followed by ASXL1 overexpression or knockdown. Overall, this study provides the first evidence that ASXL1 cooperates with HP1 to modulate LSD1 activity, leading to a change in histone H3 methylation and thereby RAR repression.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Histona Desmetilases/metabolismo , Receptores do Ácido Retinoico/metabolismo , Proteínas Repressoras/metabolismo , Transcrição Gênica , Sequência de Aminoácidos , Sítios de Ligação , Linhagem Celular , Homólogo 5 da Proteína Cromobox , Proteínas Correpressoras/metabolismo , Histonas/metabolismo , Humanos , Ligantes , Metilação/efeitos dos fármacos , Dados de Sequência Molecular , Complexos Multiproteicos/metabolismo , Regiões Promotoras Genéticas/genética , Ligação Proteica/efeitos dos fármacos , Receptores do Ácido Retinoico/genética , Proteínas Repressoras/química , Reprodutibilidade dos Testes , Transcrição Gênica/efeitos dos fármacos , Tretinoína/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...