Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
1.
Int J Mol Sci ; 25(10)2024 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-38791157

RESUMO

The additional sex combs-like (ASXL) family, a mammalian homolog of the additional sex combs (Asx) of Drosophila, has been implicated in transcriptional regulation via chromatin modifications. Abnormal expression of ASXL family genes leads to myelodysplastic syndromes and various types of leukemia. De novo mutation of these genes also causes developmental disorders. Genes in this family and their neighbor genes are evolutionary conserved in humans and mice. This review provides a comprehensive summary of epigenetic regulations associated with ASXL family genes. Their expression is commonly regulated by DNA methylation at CpG islands preceding transcription starting sites. Their proteins primarily engage in histone tail modifications through interactions with chromatin regulators (PRC2, TrxG, PR-DUB, SRC1, HP1α, and BET proteins) and with transcription factors, including nuclear hormone receptors (RAR, PPAR, ER, and LXR). Histone modifications associated with these factors include histone H3K9 acetylation and methylation, H3K4 methylation, H3K27 methylation, and H2AK119 deubiquitination. Recently, non-coding RNAs have been identified following mutations in the ASXL1 or ASXL3 gene, along with circular ASXLs and microRNAs that regulate ASXL1 expression. The diverse epigenetic regulations linked to ASXL family genes collectively contribute to tumor suppression and developmental processes. Our understanding of ASXL-regulated epigenetics may provide insights into the development of therapeutic epigenetic drugs.


Assuntos
Metilação de DNA , Epigênese Genética , Humanos , Animais , Proteínas Repressoras/metabolismo , Proteínas Repressoras/genética , Histonas/metabolismo , Mutação
2.
Biol Pharm Bull ; 46(1): 12-18, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36596519

RESUMO

Although bromodomain and extraterminal (BET) inhibitors (BETis) have anti-tumor potential, the underlying molecular mechanism is poorly understood. We found that BETis effectively repressed cell growth via G1/S arrest and migration of HCT116 cells in a p53-independent manner. BETis increased the expression of p21WAF1 and repressed the expression of E2F target genes. Consistent with this, retinoblastoma protein (Rb) phosphorylation was downregulated by BETis, supporting E2F inactivation. To investigate the epigenetic mechanism, chromatin immunoprecipitation (ChIP) assays were employed using the E2F1 target gene c-MYC. Following BETi treatment, recruitment of phosphorylated Rb, BRD2, and MLL2 to the c-MYC promoter was reduced, whereas recruitment of unphosphorylated Rb and EZH2 was increased. Consequently, decreased H4K5/K12ac and H3K4me3 accumulation but increased H3K27me3 accumulation were observed. Overall, this study suggests that BETis may be useful for the treatment of colorectal cancer via epigenetic regulation of the E2F1/c-MYC axis, leading to growth arrest in a p53-independent manner.


Assuntos
Proteínas Proto-Oncogênicas c-myc , Proteína Supressora de Tumor p53 , Humanos , Células HCT116 , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Epigênese Genética , Linhagem Celular Tumoral , Fator de Transcrição E2F1/genética , Fator de Transcrição E2F1/metabolismo
3.
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
4.
Mol Cell ; 51(2): 200-10, 2013 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-23850490

RESUMO

Despite the importance of retinoic acid (RA) signaling and histone monoubiquitination in determining cell fate, the underlying mechanism linking the two processes is poorly explored. We describe that additional sex comb-like 1 (ASXL1) represses RA receptor activity by cooperating with BRCA1-associated protein 1 (BAP1), which contains the ubiquitin C-terminal hydrolase (UCH) domain. Both the UCH- and ASXL1-binding domains of BAP1 were required for cooperation. In contrast to Drosophila BAP1, mammalian BAP1 cleaved ubiquitin from histone H2B. As supported by BAP1 mutants, ASXL1 was critical for BAP1 recruitment to chromatin and its activation therein. ASXL1 requirement was supported using Asxl1 null mice embryonic fibroblasts. Both ASXL1 and BAP1 were downregulated during RA-induced P19 cell differentiation with concomitant increase of ubiquitinated H2B, leading to activation of Hox genes. Our data demonstrate the critical role of ASXL1 cooperation with BAP1 in cell differentiation through the regulation of RA signaling associated with H2B ubiquitination.


Assuntos
Regulação da Expressão Gênica , Histonas/metabolismo , Proteínas Repressoras/fisiologia , Tretinoína/farmacologia , Proteínas Supressoras de Tumor/metabolismo , Ubiquitina Tiolesterase/metabolismo , Ubiquitina/metabolismo , Animais , Sítios de Ligação , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Células Cultivadas , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Knockout , Regiões Promotoras Genéticas/genética , Transdução de Sinais , Transcrição Gênica/genética , Proteínas Supressoras de Tumor/genética , Ubiquitina Tiolesterase/genética , Ubiquitinação
5.
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
6.
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
7.
Biochem Biophys Res Commun ; 492(1): 89-95, 2017 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-28802580

RESUMO

BRCA1-associated protein 1 (BAP1) has been implicated in diverse biological functions, including tumor suppression. However, its regulation via glycosylation and its role in embryonic stem (ES) cells are poorly defined. BAP1 was recently reported to interact with O-linked N-acetylglucosamine (O-GlcNAc) transferase (OGT). Here, we confirmed the physical interaction and investigated its functional significance. The O-GlcNAcylation of BAP1, which requires OGT, was examined in vivo and in vitro, and was proven using alloxan, an OGT inhibitor. OGT promoted the BAP1-induced repression of retinoic acid (RA)-induced RA receptor (RAR) activation. The repressive activity of BAP1 was relieved by alloxan but exacerbated by PUGNAc, an O-GlcNAcase (OGA) inhibitor. Finally, we addressed the role of O-GlcNAcylation in the RA-induced differentiation of murine ES cells. Alkaline phosphatase staining revealed the cooperation of RA and alloxan for impairing the pluripotency of ES cells. This cooperation was also observed by measuring the size of embryonic bodies and the expression of Sox2, a pluripotency marker. Overall, our data suggest that OGT-mediated O-GlcNAcylation of BAP1 prefers the maintenance of pluripotency, whereas its inhibition facilitates RA-induced differentiation in ES cells.


Assuntos
N-Acetilglucosaminiltransferases/metabolismo , Transdução de Sinais , Tretinoína/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Ubiquitina Tiolesterase/metabolismo , Aloxano/farmacologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Glicosilação/efeitos dos fármacos , Humanos , N-Acetilglucosaminiltransferases/antagonistas & inibidores , Relação Estrutura-Atividade , Tretinoína/farmacologia
8.
Biochem Biophys Res Commun ; 469(4): 884-90, 2016 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-26718407

RESUMO

Although tyrosine kinase inhibitor (TKI) therapies are highly effective in the treatment of chronic myeloid leukemia (CML), frequent recurrence limits their usage and demands new approaches for CML therapy. Stress-induced premature senescence (SIPS) is considered a potential anticancer treatment, but the underlying mechanism remains elusive. Here, we report that Sirtinol, a known SIRT1 inhibitor, induces premature senescence and growth arrest in K562 CML cells. Chromobox homolog 8 (CBX8) suppresses the Sirtinol-induced premature senescence, which is reversed by CBX8 knockdown. Upon Sirtinol treatment, the phosphorylation of AKT1, p27KIP1 and RB is severely downregulated. However, CBX8 overexpression enhances phosphorylation and, thereby, promotes the transcriptional activity of E2F1, both of which are impaired upon CBX depletion. These data suggest that CBX8 modulates SIPS through the RB-E2F1 pathway in CML cells and provide important insight into its application in CML treatment.


Assuntos
Benzamidas/administração & dosagem , Senescência Celular/efeitos dos fármacos , Fator de Transcrição E2F1/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Naftóis/administração & dosagem , Complexo Repressor Polycomb 1/metabolismo , Proteína do Retinoblastoma/metabolismo , Antineoplásicos/administração & dosagem , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Antagonismo de Drogas , Humanos , Células K562 , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Proteína Oncogênica v-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos
9.
Carcinogenesis ; 36(12): 1580-9, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26464195

RESUMO

Combination chemotherapy for the treatment of pancreatic cancer commonly employs gemcitabine with an EGFR inhibitor such as erlotinib. Here, we show that the retinoic acid derivative, ABPN, exhibits more potent anticancer effects than erlotinib, while exhibiting less toxicity toward noncancerous human control cells. Low micromolar concentrations of ABPN induced apoptosis in BxPC3 and HPAC pancreatic cancer cell lines, concomitant with a reduction in phosphorylated EGFR as well as decreased ErbB3, Met and BRUCE protein levels. The degradation of ErbB3 is a result of proteasomal degradation, possibly due to the ABPN-dependent upregulation of Nrdp1. Administration of ABPN showed significant reductions in tumor size when tested using a mouse xenograft model, with higher potency than erlotinib at the same concentration. Analysis of the tumors demonstrated that ABPN treatment suppressed ErbB3 and Met and induced Nrdp1 in vivo. The data suggest that ABPN may be more suitable in combination chemotherapy with gemcitabine than the more widely used EGFR inhibitor, erlotinib.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Pancreáticas/tratamento farmacológico , Retinoides/farmacologia , Ativação Transcricional/efeitos dos fármacos , Ubiquitina-Proteína Ligases/genética , Animais , Apoptose , Linhagem Celular Tumoral , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Sinergismo Farmacológico , Cloridrato de Erlotinib/farmacologia , Fluoruracila/farmacologia , Humanos , Masculino , Camundongos Nus , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , Proteínas Proto-Oncogênicas c-met/metabolismo , Receptor ErbB-3/metabolismo , Transdução de Sinais , Tretinoína/farmacologia , Carga Tumoral/efeitos dos fármacos , Ubiquitina-Proteína Ligases/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Gencitabina
10.
Biochem Biophys Res Commun ; 466(3): 560-6, 2015 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-26385183

RESUMO

Additional sex comb-like (ASXL) family proteins are chromatin factors that function in transcriptional activation and repression. However, the underlying mechanisms and biological implications have not been well established. Here, we identified a LIM domain-containing protein, Wilms tumor 1-interacting protein (WTIP), as an ASXL1-binding partner. Biochemical assays confirmed an interaction between the murine homologs Asxl1 and Wtip. The suppressive role of Wtip in WT1 function and the expression of Wtip in kidney podocytes prompted us to investigate the role of Asxl1 in the kidney using Asxl1-null mice. In homozygous Asxl1(-/-) embryos, defects in kidney size and glomerular podocyte morphology were observed. Furthermore, up-regulation of Wt1/Wtip target genes was observed in the kidneys of Asxl1-null embryos. Overall, these findings implicate Asxl1 in the maintenance of podocyte structure via its association with Wtip and in the regulation of WT1 signaling during early kidney development.


Assuntos
Proteínas de Transporte/metabolismo , Podócitos/citologia , Podócitos/metabolismo , Proteínas Repressoras/metabolismo , Animais , Proteínas de Transporte/genética , Proteínas Correpressoras , Proteínas do Citoesqueleto , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Células HEK293 , Células HeLa , Humanos , Rim/anormalidades , Rim/embriologia , Rim/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Gravidez , Proteínas Repressoras/deficiência , Proteínas Repressoras/genética , Transdução de Sinais , Técnicas do Sistema de Duplo-Híbrido , Regulação para Cima , Proteínas WT1
11.
Biochim Biophys Acta ; 1831(4): 698-708, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23228689

RESUMO

To investigate the underlying mechanism of targets of cyanidin, a flavonoid, which exhibits potent anti-atherogenic activities in vitro and in vivo, a natural chemical library that identified potent agonistic activity between cyanidin and peroxisome proliferator-activated receptors (PPAR) was performed. Cyanidin induced transactivation activity in all three PPAR subtypes in a reporter gene assay and time-resolved fluorescence energy transfer analyses. Cyanidin also bound directly to all three subtypes, as assessed by surface plasmon resonance experiments, and showed the greatest affinity to PPARα. These effects were confirmed by measuring the expression of unique genes of each PPAR subtype. Cyanidin significantly reduced cellular lipid concentrations in lipid-loaded steatotic hepatocytes. In addition, transcriptome profiling in lipid-loaded primary hepatocytes revealed that the net effects of stimulation with cyanidin on lipid metabolic pathways were similar to those elicited by hypolipidemic drugs. Cyanidin likely acts as a physiological PPARα agonist and potentially for PPARß/δ and γ, and reduces hepatic lipid concentrations by rewiring the expression of genes involved in lipid metabolic pathways.


Assuntos
Antocianinas/farmacologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , PPAR alfa/agonistas , Animais , Células CHO , Células Cultivadas , Cricetinae , Células Hep G2 , Humanos , Metabolismo dos Lipídeos/efeitos dos fármacos , PPAR gama/agonistas , PPAR beta/agonistas
12.
Biochem Biophys Res Commun ; 444(4): 605-10, 2014 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-24486547

RESUMO

Transcriptional activity of the retinoic acid receptor (RAR) is regulated by diverse binding partners, including classical corepressors and coactivators, in response to its ligand retinoic acid (RA). Recently, we identified a novel corepressor of RAR called the retinoic acid resistance factor (RaRF) (manuscript submitted). Here, we report how adenovirus E1A stimulates RAR activity by associating with RaRF. Based on immunoprecipitation (IP) assays, E1A interacts with RaRF through the conserved region 2 (CR2), which is also responsible for pRb binding. The first coiled-coil domain of RaRF was sufficient for this interaction. An in vitro glutathione-S-transferase (GST) pull-down assay was used to confirm the direct interaction between E1A and RaRF. Further fluorescence microscopy indicated that E1A and RaRF were located in the nucleoplasm and nucleolus, respectively. However, RaRF overexpression promoted nucleolar translocation of E1A from the nucleoplasm. Both the RA-dependent interaction of RAR with RaRF and RAR translocation to the nucleolus were disrupted by E1A. RaRF-mediated RAR repression was impaired by wild-type E1A, but not by the RaRF binding-defective E1A mutant. Taken together, our data suggest that E1A is sequestered to the nucleolus by RaRF through a specific interaction, thereby leaving RAR in the nucleoplasm for transcriptional activation.


Assuntos
Infecções por Adenoviridae/metabolismo , Adenoviridae/fisiologia , Proteínas E1A de Adenovirus/metabolismo , Nucléolo Celular/virologia , Interações Hospedeiro-Patógeno , Receptores do Ácido Retinoico/metabolismo , Proteínas Repressoras/metabolismo , Adenoviridae/química , Adenoviridae/metabolismo , Infecções por Adenoviridae/virologia , Proteínas E1A de Adenovirus/análise , Linhagem Celular , Nucléolo Celular/metabolismo , Humanos , Mapas de Interação de Proteínas , Estrutura Terciária de Proteína , Receptores do Ácido Retinoico/análise , Proteínas Repressoras/análise , Tretinoína/metabolismo
13.
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
14.
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
15.
BMB Rep ; 57(6): 299-304, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38835116

RESUMO

Upregulation of PRAME (preferentially expressed antigen of melanoma) has been implicated in the progression of a variety of cancers, including melanoma. The tumor suppressor p53 is a transcriptional regulator that mediates cell cycle arrest and apoptosis in response to stress signals. Here, we report that PRAME is a novel repressive target of p53. This was supported by analysis of melanoma cell lines carrying wild-type p53 and human melanoma databases. mRNA expression of PRAME was downregulated by p53 overexpression and activation using DNA-damaging agents, but upregulated by p53 depletion. We identified a p53-responsive element (p53RE) in the promoter region of PRAME. Luciferase and ChIP assays showed that p53 represses the transcriptional activity of the PRAME promoter and is recruited to the p53RE together with HDAC1 upon etoposide treatment. The functional significance of p53 activationmediated PRAME downregulation was demonstrated by measuring colony formation and p27 expression in melanoma cells. These data suggest that p53 activation, which leads to PRAME downregulation, could be a therapeutic strategy in melanoma cells. [BMB Reports 2024; 57(6): 299-304].


Assuntos
Antígenos de Neoplasias , Melanoma , Regiões Promotoras Genéticas , Proteína Supressora de Tumor p53 , Humanos , Proteína Supressora de Tumor p53/metabolismo , Melanoma/metabolismo , Melanoma/genética , Melanoma/patologia , Antígenos de Neoplasias/metabolismo , Antígenos de Neoplasias/genética , Linhagem Celular Tumoral , Regiões Promotoras Genéticas/genética , Regulação Neoplásica da Expressão Gênica , Etoposídeo/farmacologia , Histona Desacetilase 1/metabolismo , Regulação para Baixo/efeitos dos fármacos
16.
Biol Pharm Bull ; 36(6): 944-51, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23727915

RESUMO

The herbal flavonoid quercetin inhibits the growth of various cancer cells, but how it affects human cancer cells, particularly lung cancer cells, is unclear. We investigated the anticancer activity of quercetin and the underlying molecular mechanisms in non-small cell lung cancer (NSCLC) cells. Quercetin strongly inhibited cell proliferation, and increased sub-G1 and apoptotic cell populations regardless of p53 status. Quercetin-induced apoptosis was verified by caspase cleavage, Hoechst staining, trypan blue exclusion, and DNA fragmentation assays. Microarray analysis using H460 cells indicated that quercetin increased the expression of genes associated with death receptor signaling tumor necrosis factor-related apoptosis-inducing ligand receptor (TRAILR), caspase-10, interleukin (IL) 1R DNA fragmentation faotor 45 (DFF45), tumor necrosis factor receptor (TNFR) 1, FAS, inhibitor of kappaBalpha (IκBα)) and cell cycle inhibition growth arrest and DNA-damage inducible 45 (GADD45), p21(Cip1)), but decreased the expression of genes involved in nuclear factor (NF)-kappaB activation (NF-κB, IKKα). Further validation assays confirmed that quercetin inhibited growth by suppressing NF-κB and by increasing the expression of death receptors and cell cycle inhibitors. Taken together, these findings suggest that quercetin may be useful in the prevention and therapy of NSCLC.


Assuntos
Antineoplásicos/farmacologia , Carcinoma Pulmonar de Células não Pequenas/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias Pulmonares/genética , NF-kappa B/antagonistas & inibidores , Quercetina/farmacologia , Apoptose/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Perfilação da Expressão Gênica , Humanos , Neoplasias Pulmonares/metabolismo , NF-kappa B/genética , NF-kappa B/metabolismo , Transdução de Sinais/efeitos dos fármacos
17.
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
18.
Sci Rep ; 13(1): 22742, 2023 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-38123607

RESUMO

Hair loss caused by malfunction of the hair follicle stem cells (HFSCs) and physical damage to the skin is difficult to recover from naturally. To overcome these obstacles to hair follicle (HF) regeneration, it is essential to understand the three-dimensional (3D) microenvironment and interactions of various cells within the HFs. Therefore, 3D cell culture technology has been used in HF regeneration research; specifically, multicellular spheroids have been generally adapted to mimic the 3D volumetric structure of the HF. In this study, we culture HF-derived cells, which are mainly composed of HFSCs, in the form of 3D spheroids using a microwell array and discuss the effects of the 3D cellular environment on HF morphogenesis by expression measurements of Sonic hedgehog signaling and stem cell markers in the HF spheroids. Additionally, the influences of microwell depth on HF spheroid formation and biological conditions were investigated. The biomolecular diffusion and convective flow in the microwell were predicted using computational fluid dynamics, which allows analysis of the physical stimulations occurring on the spheroid at the micro-scale. Although a simple experimental method using the microwell array was adopted in this study, the results provide fundamental insights into the physiological phenomena of HFs in the 3D microenvironment, and the numerical analysis is expected to shed light on the investigation of the geometric parameters of the microwell system.


Assuntos
Folículo Piloso , Esferoides Celulares , Folículo Piloso/metabolismo , Proteínas Hedgehog/metabolismo , Técnicas de Cultura de Células , Células-Tronco
19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA