Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta Mol Cell Res ; 1871(3): 119659, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38216089

ABSTRACT

The effects of EGCG on the selective death of cancer cells by modulating antioxidant pathways through autophagy were explored in various normal and cancer cells. EGCG positively regulated the p62-KEAP1-NRF2-HO-1 pathway in normal cells, while negatively regulating it in cancer cells, leading to selective apoptotic death of cancer cells. In EGCG-treated MRC5 cells (EGCG-MRC5), autophagic flux was blocked, which was accompanied by the formation of p62-positive aggregates. However, EGCG-treated HeLa cells (EGCG-HeLa) showed incomplete autophagic flux and no aggregate formation. The levels of P-ULK1 S556 and S758 increased in EGCG-MRC5 through AMPK-mTOR cooperative interaction. In contrast, EGCG treatment in HeLa cells led to AMPK-induced mTOR inactivation, resulting in abrogation of P-ULK1 S556 and S758 levels. AMPK knockout in EGCG-HeLa restored positive regulation of the p62-mediated pathway, which was accompanied by increased P-mTOR S2448 and P-ULK1 S758 levels. Knockdown of 67LR in EGCG-HeLa abolished AMPK activity but did not restore the p62-mediated pathway. Surprisingly, both AMPK knockout and 67LR knockdown in EGCG-HeLa markedly increased cell viability, despite differential regulation of the antioxidant enzyme HO-1. In conclusion, EGCG induces the selective death of cancer cells through the modulation of at least two autophagy-dependent and independent regulatory pathways: negative regulation involves the mTOR-ULK1 (S556 and S758)-p62-KEAP1-NRF2-HO-1 axis via AMPK activation, whereas positive regulation occurs through the 67LR-AMPK axis.


Subject(s)
Antioxidants , Neoplasms , Humans , Antioxidants/pharmacology , Kelch-Like ECH-Associated Protein 1 , AMP-Activated Protein Kinases/genetics , HeLa Cells , NF-E2-Related Factor 2/genetics , Autophagy , TOR Serine-Threonine Kinases/genetics , Neoplasms/drug therapy , Neoplasms/genetics
2.
BMB Rep ; 56(2): 120-125, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36593106

ABSTRACT

Karyopherin-α3 (KPNA3), a karyopherin- α isoform, is intimately associated with metastatic progression via epithelial-mesenchymal transition (EMT). However, the molecular mechanism underlying how KPNA3 acts as an EMT inducer remains to be elucidated. In this report, we identified that KPNA3 was significantly upregulated in cancer cells, particularly in triple-negative breast cancer, and its knockdown resulted in the suppression of cell proliferation and metastasis. The comprehensive transcriptome analysis from KPNA3 knockdown cells indicated that KPNA3 is involved in the regulation of numerous EMTrelated genes, including the downregulation of GATA3 and E-cadherin and the up-regulation of HAS2. Moreover, it was found that KPNA3 EMT-mediated metastasis can be achieved by TGF-ß or AKT signaling pathways; this suggests that the novel independent signaling pathways KPNA3-TGF-ß-GATA3-HAS2/E-cadherin and KPNA3-AKT-HAS2/E-cadherin are involved in the EMT-mediated progress of TNBC MDA-MB-231 cells. These findings provide new insights into the divergent EMT inducibility of KPNA3 according to cell and cancer type. [BMB Reports 2023; 56(2): 120-125].


Subject(s)
Breast Neoplasms , Triple Negative Breast Neoplasms , alpha Karyopherins , Female , Humans , alpha Karyopherins/metabolism , Cadherins/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Triple Negative Breast Neoplasms/metabolism
3.
Genes Genomics ; 44(11): 1353-1361, 2022 11.
Article in English | MEDLINE | ID: mdl-35951156

ABSTRACT

BACKGROUND: Ubiquitin-like with PHD and RING finger domains 1 (UHRF1) is upregulated in colon cancer cells and associated with silencing tumor suppressor genes (TSGs) to promote colon cancer cell proliferation. OBJECTIVE: To investigate epigenetic modification of UHRF1 by TIP60. Whether UHRF1 acetylation by TIP60 can induce cell proliferation in colon cancer cells. METHODS: Acetylation sites of UHRF1 by TIP60 was predicted by ASEB (Acetylation Set Enrichment Based) method and identified by immunoprecipitation assay using anti-pan-acetyl lysine antibody and in vitro acetylation assay. Based on this method, UHRF1 acetylation-deficient mimic 4KR (K644R, K646R, K648R, K650R) mutant was generated to investigate effects of UHRF1 acetylation by TIP60. shRNA system was used to generate stable knockdown cell line of UHRF1. With transient transfection of UHRF1 WT and 4KR, the effects of UHRF1 4KR mutant on Jun dimerization protein 2 (JDP2) gene expression, cell proliferation and cell cycle were investigated by RT-qPCR and FACS analysis in shUHRF1 colon cancer cell line. RESULTS: Downregulation of TIP60-mediated UHRF1 acetylation is correlated with suppressed cell cycle progression. Acetylation-deficient mimic of UHRF1 showed poor cell growth through increased expression of JDP2 gene. CONCLUSIONS: Acetylation of UHRF1 4K residues by TIP60 is important for colon cancer cell growth. Furthermore, upregulated JDP2 expression by acetylation-deficient mutant of UHRF1 might be an important epigenetic target for colon cancer cell proliferation.


Subject(s)
CCAAT-Enhancer-Binding Proteins , Colonic Neoplasms , Lysine Acetyltransferase 5 , Ubiquitin-Protein Ligases , Acetylation , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Proliferation/genetics , Colonic Neoplasms/genetics , DNA Methylation , Humans , Lysine/genetics , Lysine/metabolism , Lysine Acetyltransferase 5/genetics , Lysine Acetyltransferase 5/metabolism , RNA, Small Interfering , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitins/genetics , Ubiquitins/metabolism
4.
BMB Rep ; 55(11): 541-546, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35880433

ABSTRACT

The repair of DNA double-strand breaks (DSBs) by homologous recombination (HR) is crucial for maintaining genomic integrity and is involved in numerous fundamental biological processes. Post-translational modifications by proteins play an important role in regulating DNA repair. Here, we report that the methyltransferase SET7 regulates HR-mediated DSB repair by methylating TIP60, a histone acetyltransferase and tumor suppressor involved in gene expression and protein stability. We show that SET7 targets TIP60 for methylation at K137, which facilitates DSB repair by promoting HR and determines cell viability against DNA damage. Interestingly, TIP60 demethylation is catalyzed by LSD1, which affects HR efficiency. Taken together, our findings reveal the importance of TIP60 methylation status by SET7 and LSD1 in the DSB repair pathway. [BMB Reports 2022; 55(11): 541-546].


Subject(s)
DNA Breaks, Double-Stranded , Histones , Methylation , Histones/metabolism , DNA Repair , Protein Processing, Post-Translational , DNA/metabolism , Histone Demethylases/metabolism
5.
Mol Cells ; 44(7): 444-457, 2021 Jul 31.
Article in English | MEDLINE | ID: mdl-34253692

ABSTRACT

Although the mechanism of chronic myeloid leukemia (CML) initiation through BCR/ABL oncogene has been well characterized, CML cell differentiation into erythroid lineage cells remains poorly understood. Using CRISPR-Cas9 screening, we identify Chromobox 8 (CBX8) as a negative regulator of K562 cell differentiation into erythrocytes. CBX8 is degraded via proteasomal pathway during K562 cell differentiation, which activates the expression of erythroid differentiation-related genes that are repressed by CBX8 in the complex of PRC1. During the differentiation process, the serine/threonine-protein kinase PIM1 phosphorylates serine 196 on CBX8, which contributes to CBX8 reduction. When CD235A expression levels are analyzed, the result reveals that the knockdown of PIM1 inhibits K562 cell differentiation. We also identify TRIM28 as another interaction partner of CBX8 by proteomic analysis. Intriguingly, TRIM28 maintains protein stability of CBX8 and TRIM28 loss significantly induces proteasomal degradation of CBX8, resulting in an acceleration of erythroid differentiation. Here, we demonstrate the involvement of the CBX8-TRIM28 axis during CML cell differentiation, suggesting that CBX8 and TRIM28 are promising novel targets for CML research.


Subject(s)
Erythroid Cells/metabolism , Tripartite Motif-Containing Protein 28/metabolism , Cell Differentiation , Humans
6.
Mol Brain ; 14(1): 106, 2021 07 03.
Article in English | MEDLINE | ID: mdl-34217333

ABSTRACT

Histone modifications are a key mechanism underlying the epigenetic regulation of gene expression, which is critically involved in the consolidation of multiple forms of memory. However, the roles of histone modifications in cerebellum-dependent motor learning and memory are not well understood. To test whether changes in histone methylation are involved in cerebellar learning, we used heterozygous Kdm3b knockout (Kdm3b+/-) mice, which show reduced lysine 9 on histone 3 (H3K9) demethylase activity. H3K9 di-methylation is significantly increased selectively in the granule cell layer of the cerebellum of Kdm3b+/- mice. In the cerebellum-dependent optokinetic response (OKR) learning, Kdm3b+/- mice show deficits in memory consolidation, whereas they are normal in basal oculomotor performance and OKR acquisition. In addition, RNA-seq analyses revealed that the expression levels of several plasticity-related genes were altered in the mutant cerebellum. Our study suggests that active regulation of histone methylation is critical for the consolidation of cerebellar motor memory.


Subject(s)
Cerebellum/physiology , Haploinsufficiency/genetics , Jumonji Domain-Containing Histone Demethylases/genetics , Memory Consolidation/physiology , Motor Activity/physiology , Animals , Gene Expression Regulation , Histones/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Lysine/metabolism , Male , Methylation , Mice, Inbred C57BL
7.
Biochem Biophys Res Commun ; 561: 120-127, 2021 07 05.
Article in English | MEDLINE | ID: mdl-34023776

ABSTRACT

Epigenetic dysregulation has been strongly implicated in carcinogenesis and is one of the mechanisms that contribute to the development of lung cancer. Using genome-wide CRISPR/Cas9 library screening, we showed SET domain-containing protein 1A (SETD1A) is an essential epigenetic modifier of the proliferation of NSCLC H1299 cells. Depletion of SETD1A strikingly inhibited the proliferation of NSCLC cells. IHC staining and bioinformatics showed that SETD1A is upregulated in lung cancer. Kaplan-Meier survival analysis indicated that high expression of SETD1A is associated with poor prognosis of patients with NSCLC. We revealed that loss of SETD1A inhibits DNA replication and induces replication stress accompanied by impaired fork progression. In addition, transcription of CDC7 and TOP1, which are involved in replication origin activation and fork progression, respectively, was significantly reduced by knockdown of SETD1A. Taken together, these findings demonstrated SETD1A is a critical epigenetic modifier of NSCLC cell proliferation by promoting the transcription of a subset of DNA replication-associated genes.


Subject(s)
Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/pathology , Histone-Lysine N-Methyltransferase/metabolism , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , CRISPR-Cas Systems , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Computational Biology/methods , DNA Replication , Databases, Genetic , Gene Expression Regulation, Neoplastic , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/genetics , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , S Phase
8.
Exp Mol Med ; 53(2): 250-263, 2021 02.
Article in English | MEDLINE | ID: mdl-33564100

ABSTRACT

The demethylation of histone lysine residues, one of the most important modifications in transcriptional regulation, is associated with various physiological states. KDM2B is a demethylase of histones H3K4, H3K36, and H3K79 and is associated with the repression of transcription. Here, we present a novel mechanism by which KDM2B demethylates serum response factor (SRF) K165 to negatively regulate muscle differentiation, which is counteracted by the histone methyltransferase SET7. We show that KDM2B inhibited skeletal muscle differentiation by inhibiting the transcription of SRF-dependent genes. Both KDM2B and SET7 regulated the balance of SRF K165 methylation. SRF K165 methylation was required for the transcriptional activation of SRF and for the promoter occupancy of SRF-dependent genes. SET7 inhibitors blocked muscle cell differentiation. Taken together, these data indicate that SRF is a nonhistone target of KDM2B and that the methylation balance of SRF as maintained by KDM2B and SET7 plays an important role in muscle cell differentiation.


Subject(s)
Cell Differentiation , F-Box Proteins/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Muscle, Skeletal/metabolism , Serum Response Factor/metabolism , Binding Sites , Biomarkers , Cell Differentiation/genetics , Cell Line , Cells, Cultured , F-Box Proteins/genetics , Gene Expression Regulation , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Methylation , Models, Biological , Muscle, Skeletal/cytology , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/metabolism , Protein Binding , Response Elements , Transcription, Genetic
9.
Cell Prolif ; 53(11): e12920, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33029857

ABSTRACT

OBJECTIVES: The level of histone H3 lysine 79 methylation is regulated by the cell cycle and involved in cell proliferation. KDM2B is an H3K79 demethylase. Proliferating cell nuclear antigen (PCNA) is a component of the DNA replication machinery. This study aimed at elucidating a molecular link between H3K79me recognition of PCNA and cell cycle control. MATERIALS AND METHODS: We generated KDM2B-depleted 293T cells and histone H3-K79R mutant-expressing 293T cells. Western blots were primarily utilized to examine the H3K79me level and its effect on subsequent PCNA dissociation from chromatin. We applied IP, peptide pull-down, isothermal titration calorimetry (ITC) and ChIP experiments to show the PCNA binding towards methylated H3K79 and DNA replication origins. Flow cytometry, MTT, iPOND and DNA fibre assays were used to assess the necessity of KDM2B for DNA replication and cell proliferation. RESULTS: We revealed that KDM2B-mediated H3K79 demethylation regulated cell cycle progression. We found that PCNA bound chromatin in an H3K79me-dependent manner during S phase. KDM2B was responsible for the timely dissociation of PCNA from chromatin, allowing to efficient DNA replication. Depletion of KDM2B aberrantly enriched chromatin with PCNA and caused slow dissociation of residual PCNA, leading to a negative effect on cell proliferation. CONCLUSIONS: We suggested a novel interaction between PCNA and H3K79me. Thus, our findings provide a new mechanism of KDM2B in regulation of DNA replication and cell proliferation.


Subject(s)
DNA Replication , F-Box Proteins/metabolism , Histones/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Cell Cycle , Cell Proliferation , Chromatin , Demethylation , HEK293 Cells , Humans , S Phase
10.
Commun Biol ; 3(1): 462, 2020 08 21.
Article in English | MEDLINE | ID: mdl-32826945

ABSTRACT

The human myelogenous leukemic cell line, K562 undergoes erythroid differentiation by exposure to hemin. Here, we uncovered NSD2 as an innate erythroid differentiation-related factor through a genome-wide CRISPR library screen and explored the regulatory role of NSD2 during myeloid leukemia cell differentiation. We found that NSD2 stability was disrupted by poly-ubiquitination in differentiated K562 cells. Proteomic analysis revealed an interaction between NSD2 and an E3 ubiquitin ligase, BRCA1, which ubiquitylates NSD on K292. Depletion of BRCA1 stabilized NSD2 protein and suppressed K562 cell differentiation. Furthermore, BRCA1 protein level was decreased in bone marrow tumor, while NSD2 level was elevated. Surprisingly, among BRCA1 mutation(s) discovered in lymphoma patients, BRCA1 K1183R prevented its translocation into the nucleus, failed to reduce NSD2 protein levels in hemin-treated K562 cells and eventually disrupted cell differentiation. Our results indicate the regulation of NSD2 stability by BRCA1-mediated ubiquitination as a potential therapeutic target process in multiple myeloma.


Subject(s)
BRCA1 Protein/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Leukemia/metabolism , Proteasome Endopeptidase Complex/metabolism , Repressor Proteins/metabolism , Biomarkers , Cell Differentiation , Cell Line, Tumor , Cells, Cultured , Epigenesis, Genetic , Gene Expression Regulation, Leukemic , Hemin/metabolism , Histone-Lysine N-Methyltransferase/genetics , Humans , K562 Cells , Leukemia/etiology , Leukemia/pathology , Neoplasm Grading , Protein Binding , Proteolysis , Repressor Proteins/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
11.
Cell Rep ; 32(4): 107958, 2020 07 28.
Article in English | MEDLINE | ID: mdl-32726623

ABSTRACT

UHRF1 is a key regulator in DNA methylation maintenance. It binds histone H3K9me2/3 and hemi-methylated DNA and recruits DNMT1 to DNA replication forks during S phase. However, the regulatory mechanism of hemi-methylated DNA binding activity of UHRF1 remains unknown. In this study, we reveal that acetylation of UHRF1 is regulated by PCAF and HDAC1. We show that UHRF1 acetylation at K490 attenuates its binding affinity to hemi-methylated DNA. We analyze genome-wide DNA methylation and gene-expression patterns using stable cell lines and discover that cells where the endogenous UHRF1 is replaced with an acetyl-mimetic (UHRF1 K490Q) mutant show deficiencies in inherited DNA methylation and show different gene-expression patterns in genes related to cell survival. These results reveal that precise regulation of UHRF1 acetylation is required to maintain DNA methylation during cell division and control cell survival.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , DNA Methylation/physiology , Ubiquitin-Protein Ligases/metabolism , Acetylation , CCAAT-Enhancer-Binding Proteins/physiology , DNA/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HCT116 Cells , HEK293 Cells , Histone Deacetylase 1/metabolism , Histones/metabolism , Humans , K562 Cells , Nuclear Proteins/metabolism , Protein Binding , Protein Processing, Post-Translational , Ubiquitin-Protein Ligases/physiology , p300-CBP Transcription Factors/metabolism
12.
PLoS One ; 15(7): e0236403, 2020.
Article in English | MEDLINE | ID: mdl-32716961

ABSTRACT

Autophagy, a self-degradative physiological process, is critical for homeostasis maintenance and energy source balancing in response to various stresses, including nutrient deprivation. It is a highly conserved catabolic process in eukaryotes and is indispensable for cell survival as it involves degradation of unessential or excessive components and their subsequent recycling as building blocks for the synthesis of necessary molecules. Although the dysregulation of autophagy has been reported to broadly contribute to various diseases, including cancers and neurodegenerative diseases, the molecular mechanisms underlying the epigenetic regulation of autophagy are poorly elucidated. Here, we report that the level of lysine demethylase 3B (KDM3B) increases in nutrient-deprived HCT116 cells, a colorectal carcinoma cell line, resulting in transcriptional activation of the autophagy-inducing genes. KDM3B was found to enhance the transcription by demethylating H3K9me2 on the promoter of these genes. Furthermore, we observed that the depletion of KDM3B inhibited the autophagic flux in HCT116 cells. Collectively, these data suggested the critical role of KDM3B in the regulation of autophagy-related genes via H3K9me2 demethylation and induction of autophagy in nutrient-starved HCT116 cells.


Subject(s)
Autophagy/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Transcriptional Activation/genetics , Amino Acids/deficiency , Autophagy/drug effects , Epigenesis, Genetic/drug effects , HCT116 Cells , HEK293 Cells , Histones/metabolism , Humans , Promoter Regions, Genetic , Proteasome Endopeptidase Complex/metabolism , Protein Processing, Post-Translational/drug effects , Proteolysis/drug effects , Sirolimus/pharmacology , Transcriptional Activation/drug effects , Valosin Containing Protein/metabolism
13.
BMB Rep ; 53(2): 112-117, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31964471

ABSTRACT

A recent study suggested that methylation of ubiquitin-like with PHD and RING finger domain 1 (UHRF1) is regulated by SET7 and lysine-specific histone demethylase 1A (LSD1) and is essential for homologous recombination (HR). The study demonstrated that SET7-mediated methylation of UHRF1 promotes polyubiquitination of proliferating cell nuclear antigen (PCNA), inducing HR. However, studies on mediators that interact with and recruit UHRF1 to damaged lesions are needed to elucidate the mechanism of UHRF1 methylationinduced HR. Here, we identified that poly [ADP-ribose] polymerase 1 (PARP1) interacts with damage-induced methylated UHRF1 specifically and mediates UHRF1 to induce HR progression. Furthermore, cooperation of UHRF1-PARP1 is essential for cell viability, suggesting the importance of the interaction of UHRF1-PARP1 for damage tolerance in response to damage. Our data revealed that PARP1 mediates the HR mechanism, which is regulated by UHRF1 methylation. The data also indicated the significant role of PARP1 as a mediator of UHRF1 methylation-correlated HR pathway. [BMB Reports 2020; 53(2): 112-117].


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , DNA Damage/genetics , Homologous Recombination/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Ubiquitin-Protein Ligases/metabolism , CCAAT-Enhancer-Binding Proteins/chemistry , CCAAT-Enhancer-Binding Proteins/genetics , Cell Survival/genetics , DNA Damage/drug effects , DNA Methylation/drug effects , G2 Phase Cell Cycle Checkpoints/drug effects , G2 Phase Cell Cycle Checkpoints/genetics , HCT116 Cells , HEK293 Cells , Humans , Hydrogen Peroxide/pharmacology , Poly (ADP-Ribose) Polymerase-1/genetics , Protein Binding , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics
14.
Anim Cells Syst (Seoul) ; 23(5): 311-317, 2019.
Article in English | MEDLINE | ID: mdl-31700696

ABSTRACT

Acute myeloid leukemia (AML) is the most common type of leukemia in adults. Owing to the chemotherapy associated side effects and toxicity, it is necessary to find a new mechanism, which can identify new potential therapeutic targets at the molecular level. Here, we identified new target genes that are induced during the TPA-induced HL-60 cell differentiation by ChIP-seq and microarray data analysis. Using q-PCR and ChIP assay, we confirmed that the target genes including USP3, USP35, TCF4, and SGK1 are upregulated during TPA-mediated HL-60 cell differentiation. Levels of USP3, one of the deubiquitinating enzymes (DUBs), increased by TPA treatment, resulting in the reduction of H2AK119ub levels. In addition, we revealed that depletion of USP3 inhibits TPA-mediated leukemia cell differentiation q-PCR and FACS analysis. Taken together, our data indicate that USP3 promotes TPA-mediated leukemia cell differentiation via regulating H2AK119ub levels.

15.
Biochem Biophys Res Commun ; 508(2): 576-582, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30514438

ABSTRACT

Hepatocellular carcinoma (HCC) is the fifth most common cancer and the third most lethal cancer worldwide. Although gene mutations associated with HCC development have been intensively studied, how epigenetic factors specifically modulate the functional properties of HCC by regulating target gene expression is unclear. Here we demonstrated the overexpression of KDM3B in liver tissue of HCC patients using public RNA-seq data. Ablation of KDM3B by CRISPR/Cas9 retarded the cell cycle and proliferation of hepatocarcinoma HepG2 cells. Approximately 30% of KDM3B knockout cells exhibited mitotic spindle multipolarity as a chromosome instability (CIN) phenotype. RNA-seq analysis of KDM3B knockout revealed significantly down-regulated expression of cell cycle related genes, especially cell proliferation factor CDC123. Furthermore, the expression level of Cyclin D1 was reduced in KDM3B knockout by proteosomal degradation without any change in the expression of CCND1, which encodes Cyclin D1. The results implicate KDM3B as a crucial epigenetic factor in cell cycle regulation that manipulates chromatin dynamics and transcription in HCC, and identifies a potential gene therapy target for effective treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Gene Regulatory Networks , Genes, cdc/genetics , Jumonji Domain-Containing Histone Demethylases/physiology , Liver Neoplasms/genetics , Carcinoma, Hepatocellular/metabolism , Cell Cycle Proteins/metabolism , Cyclin D1/metabolism , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Humans , Jumonji Domain-Containing Histone Demethylases/analysis , Jumonji Domain-Containing Histone Demethylases/genetics , Liver/metabolism , Liver Neoplasms/metabolism , Transcription, Genetic
16.
Nucleic Acids Res ; 47(4): 1692-1705, 2019 02 28.
Article in English | MEDLINE | ID: mdl-30535125

ABSTRACT

Posttranslational modifications of the Forkhead family transcription factor, FOXO1, have been known to have important regulatory implications in its diverse activities. Several types of modifications of FOXO1, including acetylation, phosphorylation, and ubiquitination, have been reported. However, lysine methylation of FOXO1 has not yet been identified. Here, we reported that FOXO1 is methylated by G9a at K273 residue in vitro and in vivo. Methylation of FOXO1 by G9a increased interaction between FOXO1 and a specific E3 ligase, SKP2, and decreased FOXO1 protein stability. In addition, G9a expression was increased by insulin and resulted in insulin-mediated FOXO1 degradation by K273 methylation. Tissue array analysis indicated that G9a was overexpressed and FOXO1 levels decreased in human colon cancer. Cell proliferation assays revealed that G9a-mediated FOXO1 methylation increased colon cancer cell proliferation. Fluorescence-activated cell sorting (FACS) analysis indicated that apoptosis rates were higher in the presence of FOXO1 than in FOXO1 knock-out cells. Furthermore, we found that G9a protein levels were elevated and FOXO1 protein levels were decreased in human colon cancer patients tissue samples. Here, we report that G9a specific inhibitor, BIX-01294, can regulate cell proliferation and apoptosis by inhibiting G9a-mediated FOXO1 methylation.


Subject(s)
Colonic Neoplasms/genetics , Forkhead Box Protein O1/genetics , Histocompatibility Antigens/genetics , Histone-Lysine N-Methyltransferase/genetics , S-Phase Kinase-Associated Proteins/genetics , Apoptosis/genetics , Azepines/pharmacology , CRISPR-Cas Systems/genetics , Cell Proliferation/drug effects , Colonic Neoplasms/pathology , DNA Methylation/genetics , Female , Flow Cytometry , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockout Techniques , HCT116 Cells , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Humans , Male , Quinazolines/pharmacology , Tissue Array Analysis , Ubiquitination/genetics
17.
Nucleic Acids Res ; 47(1): 184-196, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30357346

ABSTRACT

Ubiquitin-like with PHD and RING finger domains 1 (UHRF1) is a key epigenetic regulator of DNA methylation maintenance and heterochromatin formation. The roles of UHRF1 in DNA damage repair also have been emphasized in recent years. However, the regulatory mechanism of UHRF1 remains elusive. In this study, we showed that UHRF1 is methylated by SET7 and demethylation is catalyzed by LSD1. In addition, methylation of UHRF1 is induced in response to DNA damage and its phosphorylation in S phase is a prerequisite for interaction with SET7. Furthermore, UHRF1 methylation catalyzes the conjugation of polyubiquitin chains to PCNA and promotes homologous recombination for DNA repair. SET7-mediated UHRF1 methylation is also shown to be essential for cell viability against DNA damage. Our data revealed the regulatory mechanism underlying the UHRF1 methylation status by SET7 and LSD1 in double-strand break repair pathway.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , DNA Breaks, Double-Stranded , DNA Methylation/genetics , Histone Demethylases/genetics , Histone-Lysine N-Methyltransferase/genetics , DNA Damage/genetics , DNA Repair/genetics , Heterochromatin/genetics , Humans , Phosphorylation , Proliferating Cell Nuclear Antigen/genetics , Protein Binding/genetics , S Phase/genetics , Ubiquitin-Protein Ligases
18.
Sci Rep ; 8(1): 17075, 2018 Nov 19.
Article in English | MEDLINE | ID: mdl-30451935

ABSTRACT

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has not been fixed in the paper.

19.
PLoS One ; 13(8): e0202935, 2018.
Article in English | MEDLINE | ID: mdl-30142192

ABSTRACT

The human myeloid leukemia cell line HL-60 differentiate into monocytes following treatment with 12-O-tetradecanoylphorbol-13-acetate (TPA). However, the mechanism underlying the differentiation of these cells in response to TPA has not been fully elucidated. In this study, we performed ChIP-seq profiling of RNA Pol II, HDAC2, Acetyl H3 (AcH3), and H3K27me3 and analyzed differential chromatin state changes during TPA-induced differentiation of HL-60 cells. We focused on atypically active genes, which showed enhanced H3 acetylation despite increased HDAC2 recruitment. We found that HDAC2 positively regulates the expression of these genes in a histone deacetylase activity-independent manner. HDAC2 interacted with and recruited paired box 5 (PAX5) to the promoters of the target genes and regulated HL-60 cell differentiation by PAX5-mediated gene activation. Taken together, these data elucidated the specific-chromatin status during HL-60 cell differentiation following TPA exposure and suggested that HDAC2 can activate transcription of certain genes through interactions with PAX5 in a deacetylase activity-independent pathway.


Subject(s)
Cell Differentiation/drug effects , Cell Differentiation/genetics , Histone Deacetylase 2/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Transcriptional Activation/drug effects , Chromatin/drug effects , Chromatin/metabolism , HL-60 Cells , Humans , PAX5 Transcription Factor/metabolism
20.
Oncogene ; 37(48): 6212-6224, 2018 11.
Article in English | MEDLINE | ID: mdl-30013191

ABSTRACT

The histone methyltransferase multiple myeloma SET domain protein (MMSET/WHSC1) is highly expressed in diverse tumor types, and its expression appears to be involved in cell proliferation. In this study, we report that MMSET interacts with and methylates Aurora kinase A (AURKA). We show that MMSET-mediated methylation of AURKA induces interaction with p53 as well as enhanced kinase activity of AURKA, which results in the proteasomal degradation of p53. MMSET-mediated p53 degradation increases cell proliferation and results in oncogenic activity. Furthermore, knockdown of MMSET potently inhibits tumorigenic cells and renders them sensitive to growth inhibition by the therapeutic drug, alisertib (AURKA inhibitor). Taken together, our results suggest that MMSET is a regulator of p53 stability via methylation of AURKA in proliferating cells and might be a potential therapeutic target in solid tumors.


Subject(s)
Apoptosis/genetics , Aurora Kinase A/genetics , Cell Proliferation/genetics , Histone-Lysine N-Methyltransferase/genetics , Repressor Proteins/genetics , Tumor Suppressor Protein p53/genetics , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/genetics , HCT116 Cells , Humans , Methylation , Neoplasms/genetics , Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...