Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
1.
BMB Rep ; 57(6): 299-304, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38835116

ABSTRACT

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].


Subject(s)
Antigens, Neoplasm , Melanoma , Promoter Regions, Genetic , Tumor Suppressor Protein p53 , Humans , Tumor Suppressor Protein p53/metabolism , Melanoma/metabolism , Melanoma/genetics , Melanoma/pathology , Antigens, Neoplasm/metabolism , Antigens, Neoplasm/genetics , Cell Line, Tumor , Promoter Regions, Genetic/genetics , Gene Expression Regulation, Neoplastic , Etoposide/pharmacology , Histone Deacetylase 1/metabolism , Down-Regulation/drug effects
2.
Int J Mol Sci ; 25(10)2024 May 08.
Article in English | MEDLINE | ID: mdl-38791157

ABSTRACT

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.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Humans , Animals , Repressor Proteins/metabolism , Repressor Proteins/genetics , Histones/metabolism , Mutation
3.
Sci Rep ; 13(1): 22742, 2023 12 20.
Article in English | MEDLINE | ID: mdl-38123607

ABSTRACT

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.


Subject(s)
Hair Follicle , Spheroids, Cellular , Hair Follicle/metabolism , Hedgehog Proteins/metabolism , Cell Culture Techniques , Stem Cells
4.
Exp Mol Med ; 55(6): 1232-1246, 2023 06.
Article in English | MEDLINE | ID: mdl-37258580

ABSTRACT

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.


Subject(s)
Epigenesis, Genetic , Sirtuin 1 , Animals , Mice , Anaphase-Promoting Complex-Cyclosome/genetics , Anaphase-Promoting Complex-Cyclosome/metabolism , Cell Cycle Proteins/metabolism , Cellular Senescence , Sirtuin 1/genetics , Sirtuin 1/metabolism , Ubiquitin/metabolism , Ubiquitination
5.
Nutrients ; 15(7)2023 Apr 06.
Article in English | MEDLINE | ID: mdl-37049636

ABSTRACT

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.


Subject(s)
Histones , PPAR gamma , Mice , Animals , PPAR gamma/genetics , PPAR gamma/metabolism , Histones/metabolism , Rosiglitazone/metabolism , Rosiglitazone/pharmacology , Methylation , Adipocytes , Adipogenesis , Cell Differentiation , 3T3-L1 Cells
6.
Biol Pharm Bull ; 46(1): 12-18, 2023.
Article in English | MEDLINE | ID: mdl-36596519

ABSTRACT

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.


Subject(s)
Proto-Oncogene Proteins c-myc , Tumor Suppressor Protein p53 , Humans , HCT116 Cells , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Epigenesis, Genetic , Cell Line, Tumor , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism
7.
Biochem Biophys Res Commun ; 617(Pt 1): 48-54, 2022 08 20.
Article in English | MEDLINE | ID: mdl-35679710

ABSTRACT

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.


Subject(s)
Adipogenesis , PPAR gamma , 3T3-L1 Cells , Adipocytes/metabolism , Animals , Histones/metabolism , Kaempferols/pharmacology , Methylation , Mice , PPAR gamma/genetics , PPAR gamma/metabolism , Rosiglitazone
8.
JCI Insight ; 7(10)2022 05 23.
Article in English | MEDLINE | ID: mdl-35603786

ABSTRACT

The survival of motor neuron (SMN) protein is a major component of the pre-mRNA splicing machinery and is required for RNA metabolism. Although SMN has been considered a fundamental gene for the central nervous system, due to its relationship with neuromuscular diseases, such as spinal muscular atrophy, recent studies have also revealed the requirement of SMN in non-neuronal cells in the peripheral regions. Here, we report that the fibro-adipogenic progenitor subpopulation expressing Dpp4 (Dpp4+ FAPs) is required for the neuromuscular system. Furthermore, we also reveal that BRCA1-associated protein-1 (Bap1) is crucial for the stabilization of SMN in FAPs by preventing its ubiquitination-dependent degradation. Inactivation of Bap1 in FAPs decreased SMN levels and accompanied degeneration of the neuromuscular junction, leading to loss of motor neurons and muscle atrophy. Overexpression of the ubiquitination-resistant SMN variant, SMNK186R, in Bap1-null FAPs completely prevented neuromuscular degeneration. In addition, transplantation of Dpp4+ FAPs, but not Dpp4- FAPs, completely rescued neuromuscular defects. Our data reveal the crucial role of Bap1-mediated SMN stabilization in Dpp4+ FAPs for the neuromuscular system and provide the possibility of cell-based therapeutics to treat neuromuscular diseases.


Subject(s)
Muscular Atrophy, Spinal , Neuromuscular Diseases , Animals , Disease Models, Animal , Motor Neurons/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy, Spinal/genetics , Neuromuscular Diseases/genetics
9.
Cell Death Differ ; 29(11): 2151-2162, 2022 11.
Article in English | MEDLINE | ID: mdl-35473985

ABSTRACT

Hematopoiesis occurs within a unique bone marrow (BM) microenvironment, which consists of various niche cells, cytokines, growth factors, and extracellular matrix components. These multiple components directly or indirectly regulate the maintenance and differentiation of hematopoietic stem cells (HSCs). Here we report that BAP1 in BM mesenchymal stromal cells (MSCs) is critical for the maintenance of HSCs and B lymphopoiesis. Mice lacking BAP1 in MSCs show aberrant differentiation of hematopoietic stem and progenitor cells, impaired B lymphoid differentiation, and expansion of myeloid lineages. Mechanistically, BAP1 loss in distinct endosteal MSCs, expressing PRX1 but not LEPR, leads to aberrant expression of genes affiliated with BM niche functions. BAP1 deficiency leads to a reduced expression of pro-hematopoietic factors such as Scf caused by increased H2AK119-ub1 and H3K27-me3 levels on the promoter region of these genes. On the other hand, the expression of myelopoiesis stimulating factors including Csf3 was increased by enriched H3K4-me3 and H3K27-ac levels on their promoter, causing myeloid skewing. Notably, loss of BAP1 substantially blocks B lymphopoiesis and skews the differentiation of hematopoietic precursors toward myeloid lineages in vitro, which is reversed by G-CSF neutralization. Thus, our study uncovers a key role for BAP1 expressed in endosteal MSCs in controlling normal hematopoiesis in mice by modulating expression of various niche factors governing lymphopoiesis and myelopoiesis via histone modifications.


Subject(s)
Lymphopoiesis , Mesenchymal Stem Cells , Mice , Animals , Lymphopoiesis/genetics , Bone Marrow/metabolism , Mesenchymal Stem Cells/metabolism , Hematopoietic Stem Cells/metabolism , Hematopoiesis/genetics , Bone Marrow Cells , Cell Differentiation/genetics , Granulocyte Colony-Stimulating Factor , Epigenesis, Genetic , Stem Cell Niche/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism
10.
Biomedicines ; 9(11)2021 Nov 20.
Article in English | MEDLINE | ID: mdl-34829962

ABSTRACT

Deep learning (DL) is a distinct class of machine learning that has achieved first-class performance in many fields of study. For epigenomics, the application of DL to assist physicians and scientists in human disease-relevant prediction tasks has been relatively unexplored until very recently. In this article, we critically review published studies that employed DL models to predict disease detection, subtype classification, and treatment responses, using epigenomic data. A comprehensive search on PubMed, Scopus, Web of Science, Google Scholar, and arXiv.org was performed following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. Among 1140 initially identified publications, we included 22 articles in our review. DNA methylation and RNA-sequencing data are most frequently used to train the predictive models. The reviewed models achieved a high accuracy ranged from 88.3% to 100.0% for disease detection tasks, from 69.5% to 97.8% for subtype classification tasks, and from 80.0% to 93.0% for treatment response prediction tasks. We generated a workflow to develop a predictive model that encompasses all steps from first defining human disease-related tasks to finally evaluating model performance. DL holds promise for transforming epigenomic big data into valuable knowledge that will enhance the development of translational epigenomics.

11.
Phytother Res ; 33(9): 2429-2439, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31359554

ABSTRACT

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.


Subject(s)
Adipocytes/drug effects , Adipogenesis/physiology , Alkaloids/therapeutic use , Benzodioxoles/therapeutic use , Histone Code/physiology , Piperidines/therapeutic use , Polyunsaturated Alkamides/therapeutic use , Alkaloids/pharmacology , Benzodioxoles/pharmacology , Cell Differentiation , Humans , Piperidines/pharmacology , Polyunsaturated Alkamides/pharmacology
12.
Biochem Biophys Res Commun ; 508(3): 907-913, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30545639

ABSTRACT

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.


Subject(s)
Embryonic Stem Cells/physiology , Neurogenesis/genetics , Repressor Proteins/physiology , Animals , Axons/ultrastructure , Cells, Cultured , Embryoid Bodies/cytology , Gene Expression Regulation , Gene Knockout Techniques , Mice , Repressor Proteins/genetics
13.
Cell Death Dis ; 9(11): 1118, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30389914

ABSTRACT

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.


Subject(s)
E2F1 Transcription Factor/genetics , Epigenetic Repression , Epithelial Cells/metabolism , Lung/metabolism , N-Myc Proto-Oncogene Protein/genetics , Repressor Proteins/genetics , Respiratory Insufficiency/genetics , A549 Cells , Animals , E2F1 Transcription Factor/metabolism , Embryo, Mammalian , Epithelial Cells/pathology , Fetus , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genes, Lethal , HEK293 Cells , Histones/genetics , Histones/metabolism , Host Cell Factor C1/genetics , Host Cell Factor C1/metabolism , Humans , Lung/growth & development , Lung/pathology , Mice , Mice, Knockout , N-Myc Proto-Oncogene Protein/metabolism , Organogenesis/genetics , Promoter Regions, Genetic , Protein Binding , Repressor Proteins/deficiency , Respiratory Insufficiency/metabolism , Respiratory Insufficiency/pathology , Signal Transduction
14.
Aging Cell ; 17(1)2018 02.
Article in English | MEDLINE | ID: mdl-29130578

ABSTRACT

Alzheimer's disease (AD) is the leading cause of dementia in the elderly. Despite decades of study, effective treatments for AD are lacking. Mitochondrial dysfunction has been closely linked to the pathogenesis of AD, but the relationship between mitochondrial pathology and neuronal damage is poorly understood. Sirtuins (SIRT, silent mating type information regulation 2 homolog in yeast) are NAD-dependent histone deacetylases involved in aging and longevity. The objective of this study was to investigate the relationship between SIRT3 and mitochondrial function and neuronal activity in AD. SIRT3 mRNA and protein levels were significantly decreased in AD cerebral cortex, and Ac-p53 K320 was significantly increased in AD mitochondria. SIRT3 prevented p53-induced mitochondrial dysfunction and neuronal damage in a deacetylase activity-dependent manner. Notably, mitochondrially targeted p53 (mito-p53) directly reduced mitochondria DNA-encoded ND2 and ND4 gene expression resulting in increased reactive oxygen species (ROS) and reduced mitochondrial oxygen consumption. ND2 and ND4 gene expressions were significantly decreased in patients with AD. p53-ChIP analysis verified the presence of p53-binding elements in the human mitochondrial genome and increased p53 occupancy of mitochondrial DNA in AD. SIRT3 overexpression restored the expression of ND2 and ND4 and improved mitochondrial oxygen consumption by repressing mito-p53 activity. Our results indicate that SIRT3 dysfunction leads to p53-mediated mitochondrial and neuronal damage in AD. Therapeutic modulation of SIRT3 activity may ameliorate mitochondrial pathology and neurodegeneration in AD.


Subject(s)
Alzheimer Disease/metabolism , Mitochondria/metabolism , Sirtuin 3/metabolism , Tumor Suppressor Protein p53/metabolism , Alzheimer Disease/pathology , Animals , Mice , Neurons/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Sirtuins/metabolism
15.
Cell Death Dis ; 8(12): 3201, 2017 12 11.
Article in English | MEDLINE | ID: mdl-29233982

ABSTRACT

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.


Subject(s)
Adipocytes/metabolism , Adipogenesis/genetics , Carrier Proteins/genetics , Epigenesis, Genetic , Histone Demethylases/genetics , PPAR gamma/genetics , Sirtuin 1/genetics , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/drug effects , Adipogenesis/drug effects , Animals , Carrier Proteins/metabolism , Cell Differentiation , Co-Repressor Proteins/genetics , Co-Repressor Proteins/metabolism , Cullin Proteins , HCT116 Cells , HEK293 Cells , Histone Demethylases/metabolism , Histones/genetics , Histones/metabolism , Humans , Mice , PPAR gamma/metabolism , Resveratrol , Sequence Analysis, RNA , Signal Transduction , Sirtuin 1/metabolism , Stilbenes/pharmacology
16.
Biochem Biophys Res Commun ; 492(1): 89-95, 2017 10 07.
Article in English | MEDLINE | ID: mdl-28802580

ABSTRACT

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.


Subject(s)
N-Acetylglucosaminyltransferases/metabolism , Signal Transduction , Tretinoin/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism , Alloxan/pharmacology , Cell Differentiation/drug effects , Cells, Cultured , Glycosylation/drug effects , Humans , N-Acetylglucosaminyltransferases/antagonists & inhibitors , Structure-Activity Relationship , Tretinoin/pharmacology
17.
Sci Rep ; 7(1): 5198, 2017 07 12.
Article in English | MEDLINE | ID: mdl-28701722

ABSTRACT

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.


Subject(s)
Cellular Senescence , E2F Transcription Factors/antagonists & inhibitors , Embryo, Mammalian/pathology , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Fibroblasts/pathology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Repressor Proteins/physiology , Animals , Cell Proliferation , Cells, Cultured , E2F Transcription Factors/genetics , E2F Transcription Factors/metabolism , Embryo, Mammalian/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Fibroblasts/metabolism , Mice , Mice, Knockout , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction
18.
Cancer Lett ; 403: 144-151, 2017 09 10.
Article in English | MEDLINE | ID: mdl-28634046

ABSTRACT

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.


Subject(s)
Antigens, Neoplasm/metabolism , DNA Methylation , Epigenesis, Genetic , Kruppel-Like Transcription Factors/metabolism , Melanoma/metabolism , Skin Neoplasms/metabolism , Antigens, Neoplasm/genetics , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Binding Sites , Cell Proliferation , CpG Islands , DNA Methylation/drug effects , DNA Modification Methylases/antagonists & inhibitors , DNA Modification Methylases/metabolism , Decitabine , Enzyme Inhibitors/pharmacology , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Kruppel-Like Transcription Factors/genetics , Melanoma/genetics , Melanoma/pathology , Promoter Regions, Genetic , Protein Binding , RNA Interference , Signal Transduction , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Transcription, Genetic , Transfection , Up-Regulation
19.
Cancer Lett ; 376(2): 360-6, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27085459

ABSTRACT

The androgen receptor (AR) plays a critical role in the initiation and progression of prostate cancer (PCa), and thus its regulation is an important tool in PCa therapy. Here, we report that CDK2-associated cullin 1 (CACUL1) directly associates with AR and suppresses AR transcriptional activity. In addition, CACUL1 represses histone demethylase LSD1-mediated AR transactivation by competing with LSD1 for AR binding. Depletion of CACUL1 enhances the LSD1 occupancy of the AR-target promoter, accompanied by decreased accumulation of H3K9me2, a repressive transcriptional marker. CACUL1 and LSD1 oppositely regulate CDX-induced cell death in AR-positive LNCaP and metastatic castrate-resistant LNCaP-LN3 cells. These data suggest that CACUL1 impairs LSD1-mediated activation of AR, thereby implicating it as a potential antitumor target in PCa.


Subject(s)
Cullin Proteins/metabolism , Prostatic Neoplasms, Castration-Resistant/metabolism , Receptors, Androgen/metabolism , Androgen Antagonists/pharmacology , Anilides/pharmacology , Antineoplastic Agents, Hormonal , Cell Line, Tumor , Cullin Proteins/genetics , Gene Expression Regulation, Neoplastic , Histone Demethylases/metabolism , Histones/metabolism , Humans , Male , Methylation , Nitriles/pharmacology , Promoter Regions, Genetic , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Protein Binding , RNA Interference , Receptors, Androgen/drug effects , Receptors, Androgen/genetics , Signal Transduction , Tosyl Compounds/pharmacology , Transcription, Genetic , Transfection
20.
Biochem Biophys Res Commun ; 469(4): 884-90, 2016 Jan 22.
Article in English | MEDLINE | ID: mdl-26718407

ABSTRACT

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.


Subject(s)
Benzamides/administration & dosage , Cellular Senescence/drug effects , E2F1 Transcription Factor/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Naphthols/administration & dosage , Polycomb Repressive Complex 1/metabolism , Retinoblastoma Protein/metabolism , Antineoplastic Agents/administration & dosage , Cell Cycle Checkpoints/drug effects , Drug Antagonism , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Oncogene Protein v-akt/metabolism , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...