Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Bioorg Med Chem ; 99: 117603, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38246115

ABSTRACT

NHE5, an isoform of the Na+/H+ exchanger (NHE) protein, is an ion-transporting membrane protein that regulates intracellular pH and is highly expressed in colorectal adenocarcinoma. Therefore, we hypothesized that NHE5 inhibitors can be used as anticancer drugs. However, because NHE1 is ubiquitously expressed in all cells, it is extremely important to demonstrate its selective inhibitory activity against NHE5. We used amiloride, an NHE non-selective inhibitor, as a lead compound and created UTX-143, which has NHE5-selective inhibitory activity, using a structure-activity relationship approach. UTX-143 showed selective cytotoxic effects on cancer cells and reduced the migratory and invasive abilities of cancer cells. These results suggest a new concept wherein drugs exhibit cancer-specific cytotoxic effects through selective inhibition of NHE5 and the possibility of UTX-143 as a lead NHE5-selective inhibitor.


Subject(s)
Amiloride , Sodium , Amiloride/pharmacology , Sodium/metabolism , Sodium-Hydrogen Exchangers/metabolism , Membrane Proteins/metabolism , Hydrogen , Hydrogen-Ion Concentration
2.
Biochem Biophys Res Commun ; 669: 19-29, 2023 08 20.
Article in English | MEDLINE | ID: mdl-37262949

ABSTRACT

ASH2L (Absent-Small-Homeotic-2-Like protein) is a core subunit of the COMPASS (COMplex of Proteins ASsociated with Set1) complex, the most notable writer of the methylation of histone H3 lysine 4 (H3K4). The COMPASS complex regulates active promoters or enhancers for gene expression, and its dysfunction is associated with aberrant development and disease. Here, we demonstrated that ASH2L mediated the cell invasion and migration activity of triple-negative breast cancer cells through the interaction with the COMPASS components and the target genomic regions. Transcriptome analysis indicated a potential correlation between ASH2L and the genes involved in inflammatory/immune responses. Among them, we found that the intrinsic expression of IL1B (interleukin 1 beta), an essential proinflammatory gene, was directly regulated by ASH2L. These results revealed a novel role of ASH2L on the maintenance of breast cancer malignancy possibly through H3K4 methylation of the target inflammatory/immune responsive genes.


Subject(s)
Histones , Triple Negative Breast Neoplasms , Humans , Histones/metabolism , Methylation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Lysine/metabolism , Triple Negative Breast Neoplasms/genetics , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Epigenesis, Genetic , Transcription Factors/genetics , Transcription Factors/metabolism
3.
J Biol Chem ; 298(11): 102554, 2022 11.
Article in English | MEDLINE | ID: mdl-36183833

ABSTRACT

N6-methyladenosine (m6A) is the most common internal chemical modification of mRNAs involved in many pathological processes including various cancers. In this study, we investigated the m6A-dependent regulation of JUN and JUNB transcription factors (TFs) during transforming growth factor-beta-induced epithelial-mesenchymal transition (EMT) of A549 and LC2/ad lung cancer cell lines, as the function and regulation of these TFs within this process remains to be clarified. We found that JUN and JUNB played an important and nonredundant role in the EMT-inducing gene expression program by regulating different mesenchymal genes and that their expressions were controlled by methyltransferase-like 3 (METTL3) m6A methyltransferase. METTL3-mediated regulation of JUN expression is associated with the translation process of JUN protein but not with the stability of JUN protein or mRNA, which is in contrast with the result of m6A-mediated regulation of JUNB mRNA stability. We identified the specific m6A motifs responsible for the regulation of JUN and JUNB in EMT within 3'UTR of JUN and JUNB. Furthermore, we discovered that different m6A reader proteins interacted with JUN and JUNB mRNA and controlled m6A-dependent expression of JUN protein and JUNB mRNA. These results demonstrate that the different modes of m6A-mediated regulation of JUN and JUNB TFs provide critical input in the gene regulatory network during transforming growth factor-beta-induced EMT of lung cancer cells.


Subject(s)
Epithelial-Mesenchymal Transition , Lung Neoplasms , Humans , Epithelial-Mesenchymal Transition/genetics , Transforming Growth Factor beta/metabolism , Methylation , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , RNA, Messenger/genetics , Transcription Factor AP-1/metabolism , Methyltransferases/genetics , Methyltransferases/metabolism , Transforming Growth Factors/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Proc Natl Acad Sci U S A ; 118(43)2021 10 26.
Article in English | MEDLINE | ID: mdl-34663724

ABSTRACT

Although it is held that proinflammatory changes precede the onset of breast cancer, the underlying mechanisms remain obscure. Here, we demonstrate that FRS2ß, an adaptor protein expressed in a small subset of epithelial cells, triggers the proinflammatory changes that induce stroma in premalignant mammary tissues and is responsible for the disease onset. FRS2ß deficiency in mouse mammary tumor virus (MMTV)-ErbB2 mice markedly attenuated tumorigenesis. Importantly, tumor cells derived from MMTV-ErbB2 mice failed to generate tumors when grafted in the FRS2ß-deficient premalignant tissues. We found that colocalization of FRS2ß and the NEMO subunit of the IκB kinase complex in early endosomes led to activation of nuclear factor-κB (NF-κB), a master regulator of inflammation. Moreover, inhibition of the activities of the NF-κB-induced cytokines, CXC chemokine ligand 12 and insulin-like growth factor 1, abrogated tumorigenesis. Human breast cancer tissues that express higher levels of FRS2ß contain more stroma. The elucidation of the FRS2ß-NF-κB axis uncovers a molecular link between the proinflammatory changes and the disease onset.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms/etiology , Breast Neoplasms/metabolism , Mammary Neoplasms, Experimental/etiology , Mammary Neoplasms, Experimental/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Animals , Breast Neoplasms/immunology , Carcinogenesis , Cytokines/metabolism , Female , Humans , Inflammation/etiology , Inflammation/metabolism , Mammary Neoplasms, Experimental/immunology , Mammary Tumor Virus, Mouse , Mice , Mice, Knockout , NF-kappa B/metabolism , Pregnancy , Receptor, ErbB-2/metabolism , Retroviridae Infections , Tumor Microenvironment/immunology , Tumor Virus Infections
6.
Biochem Biophys Rep ; 27: 101072, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34381878

ABSTRACT

Matrix metalloproteinase (MMP)-2 and MMP-9, also known as gelatinases or type IV collagenases, are recognized as major contributors to the proteolytic degradation of extracellular matrix during tumor invasion. Latent MMP-2 (proMMP-2) is activated by membrane type 1 MMP (MT1-MMP) on the cell surface of tumor cells. We previously reported that cell-bound proMMP-9 is activated by the MT1-MMP/MMP-2 axis in HT1080 cells treated with concanavalin A in the presence of exogenous proMMP-2. However, the regulatory mechanism of proMMP-9 activation remains largely unknown. Transforming growth factor (TGF)-ß1 is frequently overexpressed in tumor tissues and is associated with tumor aggressiveness and poor prognosis. In this study, we examined the role of TGF-ß1 on MT1-MMP-mediated proMMP-9 activation using human oral squamous cell carcinoma cells. TGF-ß1 significantly increased the expression of MMP-9. By adding exogenous proMMP-2, TGF-ß1-induced proMMP-9 was activated during collagen gel culture, which was suppressed by the inhibition of TGF-ß1 signaling or MT1-MMP activity. This MT1-MMP-mediated proMMP-9 activation was needed to facilitate TGF-ß1-induced cell invasion into collagen gel. Thus, TGF-ß1 may facilitate MT1-MMP-mediated MMP-9 activation and thereby stimulate invasion of tumor cells in collaboration with MT1-MMP and MMP-2.

7.
J Biol Chem ; 296: 100213, 2021.
Article in English | MEDLINE | ID: mdl-33779563

ABSTRACT

Polycomb repressive complex-1 (PRC1) induces transcriptional repression by regulating monoubiquitination of lysine 119 of histone H2A (H2AK119) and as such is involved in a number of biological and pathological processes including cancer development. Previously we demonstrated that PRC2, which catalyzes the methylation of histone H3K27, has an essential function in TGF-ß-induced epithelial-mesenchymal transition (EMT) of lung and pancreatic cancer cell lines. Since the cooperative activities of PRC1 and PRC2 are thought to be important for transcriptional repression in EMT program, we investigated the role of KDM2B, a member of PRC1 complex, on TGF-ß-induced EMT in this study. Knockdown of KDM2B inhibited TGF-ß-induced morphological conversion of the cells and enhanced cell migration and invasion potentials as well as the expression changes of EMT-related marker genes. Overexpression of KDM2B influenced the expression of several epithelial marker genes such as CDH1, miR200a, and CGN and enhanced the effects of TGF-ß. Mechanistic investigations revealed that KDM2B specifically recognized the regulatory regions of CDH1, miR200a, and CGN genes and induced histone H2AK119 monoubiquitination as a component of PRC1 complex, thereby mediating the subsequent EZH2 recruitment and histone H3K27 methylation process required for gene repression. Studies using KDM2B mutants confirmed that its DNA recognition property but not its histone H3 demethylase activity was indispensable for its function during EMT. This study demonstrated the significance of the regulation of histone H2A ubiquitination in EMT process and provided the possibility to develop novel therapeutic strategies for the treatment of cancer metastasis.


Subject(s)
Epithelial-Mesenchymal Transition/drug effects , F-Box Proteins/metabolism , Histones/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Lung Neoplasms/pathology , Pancreatic Neoplasms/pathology , Transforming Growth Factor beta/pharmacology , Antigens, CD/metabolism , Cadherins/metabolism , Cell Line, Tumor , Enhancer of Zeste Homolog 2 Protein/metabolism , F-Box Proteins/genetics , Gene Expression Regulation, Neoplastic , Histones/genetics , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism
8.
Sci Rep ; 10(1): 16678, 2020 10 07.
Article in English | MEDLINE | ID: mdl-33028834

ABSTRACT

The aggressive invasiveness of malignant mesothelioma limits cancer therapy, however, the molecular mechanisms underlying the invasiveness remain largely unknown. Here we found that the matrix metalloproteinase-2 (MMP-2) was required for the invasion of mesothelioma cells in the collagen matrix and the gene expression of MMP-2 was correlated with the invasive phenotype. The MMP-2 gene expression was regulated by DNA and histone methylation around the transcription start site, implicating the involvement of the polycomb repressive complex (PRC). Knockdown of PRC component chromobox 6 (CBX6) promoted MMP-2 expression and invasion of mesothelioma cells. Transcriptome analysis suggested that CBX6 regulates sets of genes involved in cancer cell migration and metastasis. In invasive but not non-invasive cells, CBX6 was constantly unstable owing to ubiquitination and protein degradation. In human tissues, CBX6 localized in the nuclei of normal mesothelium and benign mesothelioma, but the nuclear staining of CBX6 was lost in malignant mesothelioma. These results suggest involvement of proteasomal degradation of CBX6 in mesothelioma progression.


Subject(s)
Matrix Metalloproteinase 2/metabolism , Mesothelioma, Malignant/metabolism , Polycomb-Group Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Epigenesis, Genetic , Gene Expression Profiling , Gene Knockdown Techniques , Humans , Matrix Metalloproteinase 2/genetics , Mesothelioma, Malignant/genetics , Mesothelioma, Malignant/pathology , Neoplasm Invasiveness/pathology , Polycomb-Group Proteins/genetics
9.
Nat Commun ; 11(1): 4607, 2020 09 14.
Article in English | MEDLINE | ID: mdl-32929081

ABSTRACT

Drug tolerance is the basis for acquired resistance to epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) including osimertinib, through mechanisms that still remain unclear. Here, we show that while AXL-low expressing EGFR mutated lung cancer (EGFRmut-LC) cells are more sensitive to osimertinib than AXL-high expressing EGFRmut-LC cells, a small population emerge osimertinib tolerance. The tolerance is mediated by the increased expression and phosphorylation of insulin-like growth factor-1 receptor (IGF-1R), caused by the induction of its transcription factor FOXA1. IGF-1R maintains association with EGFR and adaptor proteins, including Gab1 and IRS1, in the presence of osimertinib and restores the survival signal. In AXL-low-expressing EGFRmut-LC cell-derived xenograft and patient-derived xenograft models, transient IGF-1R inhibition combined with continuous osimertinib treatment could eradicate tumors and prevent regrowth even after the cessation of osimertinib. These results indicate that optimal inhibition of tolerant signals combined with osimertinib may dramatically improve the outcome of EGFRmut-LC.


Subject(s)
Acrylamides/therapeutic use , Aniline Compounds/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mutation/genetics , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, IGF Type 1/antagonists & inhibitors , Acrylamides/pharmacology , Aged, 80 and over , Aniline Compounds/pharmacology , Animals , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Survival/drug effects , ErbB Receptors/genetics , Gene Expression Regulation, Neoplastic/drug effects , Hepatocyte Nuclear Factor 3-alpha/metabolism , Humans , Imidazoles/pharmacology , Lung Neoplasms/pathology , Male , Mice , Models, Biological , Phosphorylation/drug effects , Pyrazines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Up-Regulation/drug effects , Axl Receptor Tyrosine Kinase
10.
Biochem Biophys Res Commun ; 524(1): 150-155, 2020 03 26.
Article in English | MEDLINE | ID: mdl-31982139

ABSTRACT

N6-Methyladenosine (m6A) is the most common internal chemical modification of mRNAs involved in many pathological processes including various cancers. In this study, we investigated the role of m6A methyltransferase METTL3 in TGF-ß-induced epithelial-mesenchymal transition (EMT) of lung cancer cell lines. The expression of METTL3 and m6A RNA modification were increased during TGF-ß-induced EMT of A549 and LC2/ad lung cancer cells. Knockdown of METTL3 inhibited TGF-ß-induced morphological conversion of the cells, enhanced cell migration potential and the expression changes of EMT-related marker genes such as CDH1/E-cadherin, FN1/Fibronectin and VIM/Vimentin. Mechanistic investigations revealed that METTL3 knockdown decreased the m6A modification, total mRNA level and mRNA stability of JUNB, one of the important transcriptional regulators of EMT. Over-expression of JUNB partially rescued the inhibitory effects of METTL3 knockdown in the EMT phenotypes. This study demonstrates that m6A methyltransferase METTL3 is indispensable for TGF-ß-induced EMT of lung cancer cells through the regulation of JUNB.


Subject(s)
Epithelial-Mesenchymal Transition/drug effects , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Methyltransferases/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta/pharmacology , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Lung Neoplasms/genetics , Methyltransferases/genetics , Phenotype , RNA Stability/drug effects , Transcription Factors/genetics
11.
J Biol Chem ; 293(47): 18016-18030, 2018 11 23.
Article in English | MEDLINE | ID: mdl-30262664

ABSTRACT

Long noncoding RNAs (lncRNAs) are important regulatory molecules in various biological and pathological processes, including cancer development. We have previously shown that the MEG3 lncRNA plays an essential role in transforming growth factor-ß (TGF-ß)-induced epithelial-mesenchymal transition (EMT) of human lung cancer cells. In this study, we investigated the function of another lncRNA, MEG8, which shares the DLK1-DIO3 locus with MEG3, in the regulation of EMT. MEG8 lncRNA expression was immediately induced during TGF-ß-mediated EMT of A549 and LC2/ad lung cancer and Panc1 pancreatic cancer cell lines. MEG8 overexpression specifically suppressed the expression of microRNA-34a and microRNA-203 genes, resulting in up-regulation of SNAIL family transcriptional repressor 1 (SNAI1) and SNAI2 transcription factors, which repressed expression of cadherin 1 (CDH1)/E-cadherin. Mechanistic investigations revealed that MEG8 associates with enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2) protein and induces its recruitment to the regulatory regions of the two microRNA genes for histone H3 methylation and transcriptional repression. Interestingly, expression of both MEG8 and MEG3, but not each individually, could induce EMT-related cell morphological changes and increased cell motility in the absence of TGF-ß by activating the gene expression program required for EMT. MEG8 knockdown indicated that endogenous MEG8 lncRNA is indispensable for TGF-ß-induced EMT in A549 lung cancer and Panc1 pancreatic cancer cells. Our findings indicate that MEG8 lncRNA significantly contributes to epigenetic EMT induction and increase our understanding of the lncRNA-mediated regulatory mechanisms involved in malignant progression of cancer.


Subject(s)
Epithelial-Mesenchymal Transition , Lung Neoplasms/genetics , Lung Neoplasms/physiopathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/physiopathology , RNA, Long Noncoding/genetics , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Epigenesis, Genetic , Epithelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Pancreatic Neoplasms/metabolism , RNA, Long Noncoding/metabolism , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , Transforming Growth Factor beta1
12.
Biochem Biophys Res Commun ; 490(4): 1407-1413, 2017 09 02.
Article in English | MEDLINE | ID: mdl-28698146

ABSTRACT

Histone methylation is associated with various biological and pathological processes including cancer development. KDM6A is a candidate tumor suppressor gene that encodes a histone H3 lysine 27 (H3K27) demethylase. In this study, we discovered that ectopic expression of KDM6A antagonized TGF-ß-induced epithelial-mesenchymal transition (EMT) and cell migration of lung cancer cell lines through its demethylase activity. KDM6A counteracted TGF-ß-dependent changes in the expression of EMT-related genes such as CDH1/E-cadherin, FN1/Fibronectin, ZEB family and microRNA-200 family. Mechanistic investigations revealed that KDM6A inhibited the recruitment of EZH2 histone H3K27 methyltransferase and H3K27 methylation on the regulatory regions of the target genes such as CDH1 and microRNA-200 family. Knockdown of KDM6A did not proceed EMT by itself, but influenced the expression of specific target genes critical for EMT, suggesting that endogenous KDM6A was involved in EMT-inducing transcriptional program. This study demonstrated a novel regulatory role of KDM6A histone demethylase in the epigenetic control of EMT process in lung cancer cells.


Subject(s)
Epigenesis, Genetic , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Histone Demethylases/genetics , Transforming Growth Factor beta/genetics , A549 Cells , Animals , Antigens, CD , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Cell Movement , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Fibronectins/genetics , Fibronectins/metabolism , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Histone Demethylases/metabolism , Humans , Lentivirus/genetics , Lentivirus/metabolism , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Transforming Growth Factor beta/metabolism , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism
13.
J Biol Chem ; 292(1): 82-99, 2017 Jan 06.
Article in English | MEDLINE | ID: mdl-27852821

ABSTRACT

Histone methylation is implicated in a number of biological and pathological processes, including cancer development. In this study, we investigated the molecular mechanism for the recruitment of Polycomb repressive complex-2 (PRC2) and its accessory component, JARID2, to chromatin, which regulates methylation of lysine 27 of histone H3 (H3K27), during epithelial-mesenchymal transition (EMT) of cancer cells. The expression of MEG3 long noncoding RNA (lncRNA), which could interact with JARID2, was clearly increased during transforming growth factor-ß (TGF-ß)-induced EMT of human lung cancer cell lines. Knockdown of MEG3 inhibited TGF-ß-mediated changes in cell morphology and cell motility characteristic of EMT and counteracted TGF-ß-dependent changes in the expression of EMT-related genes such as CDH1, ZEB family, and the microRNA-200 family. Overexpression of MEG3 influenced the expression of these genes and enhanced the effects of TGF-ß in their expressions. Chromatin immunoprecipitation (ChIP) revealed that MEG3 regulated the recruitment of JARID2 and EZH2 and histone H3 methylation on the regulatory regions of CDH1 and microRNA-200 family genes for transcriptional repression. RNA immunoprecipitation and chromatin isolation by RNA purification assays indicated that MEG3 could associate with JARID2 and the regulatory regions of target genes to recruit the complex. This study demonstrated a crucial role of MEG3 lncRNA in the epigenetic regulation of the EMT process in lung cancer cells.


Subject(s)
Epigenesis, Genetic/genetics , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Antigens, CD , Apoptosis , Blotting, Western , Cadherins/genetics , Cadherins/metabolism , Cell Proliferation , Chromatin/genetics , Fluorescent Antibody Technique , Histones/genetics , Humans , Immunoprecipitation , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Tumor Cells, Cultured
14.
Biochimie ; 123: 20-31, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26783998

ABSTRACT

DOT1L is a histone H3 lysine 79 (H3K79) methyltransferase mainly implicated in leukemia. Here we analyzed the function of DOT1L in breast cancer cells. The expression of DOT1L was up-regulated in malignant breast cancer tissues. Over-expression of DOT1L significantly increased the sphere formation and the cell migration activities of MCF7 breast cancer cell line. In contrast, knockdown of DOT1L reduced the cell migration activity of MDA-MB-231 breast cancer cell line. BCAT1, which encodes a branched-chain amino acid transaminase, was identified as one of the target genes controlled by DOT1L through the regulation of H3K79 methylation. Mechanistic investigation revealed that BCAT1 might be an important regulator responsible for DOT1L-mediated sphere formation and cell migration in breast cancer cells.


Subject(s)
Breast Neoplasms/pathology , Methyltransferases/metabolism , Neoplasm Metastasis , Transaminases/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Gene Expression Regulation , Histone-Lysine N-Methyltransferase , Humans , Methylation
15.
Cell Tissue Res ; 363(3): 723-33, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26334721

ABSTRACT

Genetic studies have shown that aberrant activation of p53 signaling leads to embryonic lethality. Maintenance of a fine balance of the p53 protein level is critical for normal development. Previously, we have reported that Jmjd5, a member of the Jumonji C (JmjC) family, regulates embryonic cell proliferation through the control of Cdkn1a expression. Since Cdkn1a is the representative p53-regulated gene, we have examined whether the expression of other p53 target genes is coincidentally upregulated with Cdkn1a in Jmjd5-deficient embryos. The expression of a subset of p53-regulated genes was increased in both Jmjd5 hypomorphic mouse embryonic fibroblasts (MEFs) and Jmjd5-deficient embryos at embryonic day 8.25 without the induced expression of Trp53. Intercrossing of Jmjd5-deficient mice with Trp53 knockout mice showed that the growth defect of Jmjd5 mutant cells was significantly recovered under a Trp53 null genetic background. Chromatin immunoprecipitation analysis in Jmjd5 hypomorphic MEFs indicated the increased recruitment of p53 at several p53 target gene loci, such as Cdkn1a, Pmaip1, and Mdm2. These results suggest that Jmjd5 is involved in the transcriptional regulation of a subset of p53-regulated genes, possibly through the control of p53 recruitment at the gene loci. In Jmjd5-deficient embryos, the enhanced recruitment of p53 might result in the abnormal activation of p53 signaling leading to embryonic lethality.


Subject(s)
Embryonic Development , Jumonji Domain-Containing Histone Demethylases/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Animals , Cell Proliferation , Embryo, Mammalian/cytology , Embryonic Development/genetics , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Genetic Loci , Humans , Jumonji Domain-Containing Histone Demethylases/deficiency , Jumonji Domain-Containing Histone Demethylases/genetics , Mice , Phenotype , Signal Transduction/genetics , Up-Regulation/genetics
16.
Epigenetics ; 10(9): 861-71, 2015.
Article in English | MEDLINE | ID: mdl-26252449

ABSTRACT

Several studies have described phenotypic changes in the offspring of mice exposed to a variety of environmental factors, including diet, toxins, and stress; however, the molecular pathways involved in these changes remain unclear. Using a high fat diet (HFD)-induced obesity mouse model, we examined liver gene expression in male offspring and analyzed chromatin of paternal spermatozoa. We found that the hepatic mRNA level of 7 genes (out of 20 evaluated) was significantly altered in HFD male offspring compared to control mice, suggesting that phenotypic changes in the offspring depend on parental diet. We examined 7 imprinted loci in spermatozoa DNA from HFD-treated and control fathers by bisulfite sequencing, but did not detect changes in DNA methylation associated with HFD. Using chromatin immunoprecipitation followed by high-throughput sequencing, we found differential histone H3-occupancy at genes involved in the regulation of embryogenesis and differential H3K4me1-enrichment at transcription regulatory genes in HFD fathers vs. control mice. These results suggest that dietary exposure can modulate histone composition at regulatory genes implicated in developmental processes.


Subject(s)
Diet, High-Fat/adverse effects , Gene Expression/drug effects , Histones/metabolism , Liver/metabolism , Spermatozoa/drug effects , Animals , DNA Methylation/drug effects , Disease Models, Animal , Gene Expression Regulation/drug effects , Genomic Imprinting/drug effects , Male , Mice , Sequence Analysis, DNA , Spermatozoa/metabolism
17.
Nucleic Acids Res ; 43(3): 1444-55, 2015 Feb 18.
Article in English | MEDLINE | ID: mdl-25578963

ABSTRACT

Lsh, a chromatin remodeling protein of the SNF2 family, is critical for normal heterochromatin structure. In particular, DNA methylation at repeat elements, a hallmark of heterochromatin, is greatly reduced in Lsh(-/-) (KO) cells. Here, we examined the presumed nucleosome remodeling activity of Lsh on chromatin in the context of DNA methylation. We found that dynamic CG methylation was dependent on Lsh in embryonic stem cells. Moreover, we demonstrate that ATP function is critical for de novo methylation at repeat sequences. The ATP binding site of Lsh is in part required to promote stable association of the DNA methyltransferase 3b with the repeat locus. By performing nucleosome occupancy assays, we found distinct nucleosome occupancy in KO ES cells compared to WT ES cells after differentiation. Nucleosome density was restored to wild-type level by re-expressing wild-type Lsh but not the ATP mutant in KO ES cells. Our results suggest that ATP-dependent nucleosome remodeling is the primary molecular function of Lsh, which may promote de novo methylation in differentiating ES cells.


Subject(s)
Adenosine Triphosphate/metabolism , Chromatin Assembly and Disassembly , DNA Helicases/metabolism , DNA Methylation , Nucleosomes/metabolism , Repetitive Sequences, Nucleic Acid , Binding Sites , Cells, Cultured , Humans
18.
PLoS One ; 9(12): e115684, 2014.
Article in English | MEDLINE | ID: mdl-25542019

ABSTRACT

Histone methylation plays a crucial role in various biological and pathological processes including cancer development. In this study, we discovered that JARID2, an interacting component of Polycomb repressive complex-2 (PRC2) that catalyzes methylation of lysine 27 of histone H3 (H3K27), was involved in Transforming Growth Factor-beta (TGF-ß)-induced epithelial-mesenchymal transition (EMT) of A549 lung cancer cell line and HT29 colon cancer cell line. The expression of JARID2 was increased during TGF-ß-induced EMT of these cell lines and knockdown of JARID2 inhibited TGF-ß-induced morphological conversion of the cells associated with EMT. JARID2 knockdown itself had no effect in the expression of EMT-related genes but antagonized TGF-ß-dependent expression changes of EMT-related genes such as CDH1, ZEB family and microRNA-200 family. Chromatin immunoprecipitation assays showed that JARID2 was implicated in TGF-ß-induced transcriptional repression of CDH1 and microRNA-200 family genes through the regulation of histone H3 methylation and EZH2 occupancies on their regulatory regions. Our study demonstrated a novel role of JARID2 protein, which may control PRC2 recruitment and histone methylation during TGF-ß-induced EMT of lung and colon cancer cell lines.


Subject(s)
Colonic Neoplasms/metabolism , Epithelial-Mesenchymal Transition , Lung Neoplasms/metabolism , Polycomb Repressive Complex 2/metabolism , Antigens, CD , Cadherins/genetics , Cadherins/metabolism , Enhancer of Zeste Homolog 2 Protein , HT29 Cells , Histones/metabolism , Humans , MicroRNAs/genetics , Polycomb Repressive Complex 2/genetics , Transforming Growth Factor beta/pharmacology
19.
Biochem Biophys Res Commun ; 453(1): 124-30, 2014 Oct 10.
Article in English | MEDLINE | ID: mdl-25264103

ABSTRACT

Histone methylation is involved in various biological and pathological processes including cancer development. In this study, we found that EED, a component of Polycomb repressive complex-2 (PRC2) that catalyzes methylation of lysine 27 of histone H3 (H3K27), was involved in epithelial-mesenchymal transition (EMT) of cancer cells induced by Transforming Growth Factor-beta (TGF-ß). The expression of EED was increased during TGF-ß-induced EMT and knockdown of EED inhibited TGF-ß-induced morphological conversion of the cells associated with EMT. EED knockdown antagonized TGF-ß-dependent expression changes of EMT-related genes such as CDH1, ZEB1, ZEB2 and microRNA-200 (miR-200) family. Chromatin immunoprecipitation assays showed that EED was implicated in TGF-ß-induced transcriptional repression of CDH1 and miR-200 family genes through the regulation of histone H3 methylation and EZH2 occupancies on their regulatory regions. Our study demonstrated a novel role of EED, which regulates PRC2 activity and histone methylation during TGF-ß-induced EMT of cancer cells.


Subject(s)
Epithelial-Mesenchymal Transition/physiology , Neoplasms/pathology , Neoplasms/physiopathology , Polycomb Repressive Complex 2/physiology , Transforming Growth Factor beta/physiology , Antigens, CD , Cadherins/genetics , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HT29 Cells , Histones/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , MicroRNAs/genetics , Neoplasms/genetics , Polycomb Repressive Complex 2/antagonists & inhibitors , Polycomb Repressive Complex 2/genetics , Promoter Regions, Genetic , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Zinc Finger E-box Binding Homeobox 2 , Zinc Finger E-box-Binding Homeobox 1
20.
Genome Res ; 24(10): 1613-23, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25170028

ABSTRACT

Cytosine methylation is critical in mammalian development and plays a role in diverse biologic processes such as genomic imprinting, X chromosome inactivation, and silencing of repeat elements. Several factors regulate DNA methylation in early embryogenesis, but their precise role in the establishment of DNA methylation at a given site remains unclear. We have generated a comprehensive methylation map in fibroblasts derived from the murine DNA methylation mutant Hells(-/-) (helicase, lymphoid specific, also known as LSH). It has been previously shown that HELLS can influence de novo methylation of retroviral sequences and endogenous genes. Here, we describe that HELLS controls cytosine methylation in a nuclear compartment that is in part defined by lamin B1 attachment regions. Despite widespread loss of cytosine methylation at regulatory sequences, including promoter regions of protein-coding genes and noncoding RNA genes, overall relative transcript abundance levels in the absence of HELLS are similar to those in wild-type cells. A subset of promoter regions shows increases of the histone modification H3K27me3, suggesting redundancy of epigenetic silencing mechanisms. Furthermore, HELLS modulates CG methylation at all classes of repeat elements and is critical for repression of a subset of repeat elements. Overall, we provide a detailed analysis of gene expression changes in relation to DNA methylation alterations, which contributes to our understanding of the biological role of cytosine methylation.


Subject(s)
Cytosine/metabolism , DNA Helicases/genetics , DNA Methylation , DNA/metabolism , Gene Expression Regulation, Developmental , Animals , Cell Nucleus/genetics , Embryonic Stem Cells , Epigenesis, Genetic , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Knockout Techniques , Histones/metabolism , Lamin Type B/metabolism , Mice , Molecular Sequence Data , Mutation , Regulatory Sequences, Nucleic Acid , Repetitive Sequences, Nucleic Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...