41 results on '"Enhancer of Zeste Homolog 2 Protein metabolism"'
Search Results
2. Unveiling the non-canonical functions of EZH2 in prostate cancer.
- Author
-
Yi Y, Li Y, Chen K, and Cao Q
- Subjects
- Male, Humans, Transcription Factors genetics, Enhancer of Zeste Homolog 2 Protein genetics, Enhancer of Zeste Homolog 2 Protein metabolism, Cell Line, Tumor, Gene Expression Regulation, Neoplastic, Prostatic Neoplasms genetics
- Published
- 2023
- Full Text
- View/download PDF
3. Long non-coding RNA UCA1 promotes gallbladder cancer progression by epigenetically repressing p21 and E-cadherin expression.
- Author
-
Cai Q, Jin L, Wang S, Zhou D, Wang J, Tang Z, and Quan Z
- Subjects
- Adult, Aged, Animals, Cell Line, Tumor, Cell Proliferation, Cell Transformation, Neoplastic, Disease Models, Animal, Disease Progression, Enhancer of Zeste Homolog 2 Protein metabolism, Epithelial-Mesenchymal Transition, Female, Gallbladder Neoplasms mortality, Heterografts, Humans, Lymphatic Metastasis, Male, Mice, Middle Aged, Neoplasm Grading, Neoplasm Staging, Prognosis, Promoter Regions, Genetic, Protein Binding, Transcription, Genetic, Tumor Burden, Cadherins genetics, Cyclin-Dependent Kinase Inhibitor p21 genetics, Epigenesis, Genetic, Gallbladder Neoplasms genetics, Gallbladder Neoplasms pathology, Gene Expression Regulation, Neoplastic, RNA, Long Noncoding genetics
- Abstract
A growing number of studies indicated that long non-coding RNAs (lncRNAs) determine some cellular processes in cancer, such as proliferation, metastasis and differentiation. Urothelial carcinoma associated 1 (UCA1), an lncRNA, had been reported for its overexpression and oncogenic effect on various human cancers. In this study, we found that UCA1 was significantly overexpressed in gallbladder cancer (GBC) and positively correlated with tumor size, lymph node metastasis, TNM stage and short survival time. Moreover, UCA1 promoted GBC cell proliferation and metastasis in vitro and tumor growth in vivo. Mechanically, we identified that UCA1 promoted GBC progression through recruiting enhancer of zeste homolog 2 (EZH2) to the promoter of p21 and E-cadherin, and epigenetically suppressing their transcript.
- Published
- 2017
- Full Text
- View/download PDF
4. Oncogenic histone methyltransferase EZH2: A novel prognostic marker with therapeutic potential in endometrial cancer.
- Author
-
Oki S, Sone K, Oda K, Hamamoto R, Ikemura M, Maeda D, Takeuchi M, Tanikawa M, Mori-Uchino M, Nagasaka K, Miyasaka A, Kashiyama T, Ikeda Y, Arimoto T, Kuramoto H, Wada-Hiraike O, Kawana K, Fukayama M, Osuga Y, and Fujii T
- Subjects
- Apoptosis drug effects, Apoptosis genetics, Biomarkers, Tumor genetics, Carcinogenesis genetics, Cell Line, Tumor, Cell Proliferation drug effects, Cell Proliferation genetics, Cisplatin pharmacology, Doxorubicin pharmacology, Endometrial Neoplasms genetics, Endometrial Neoplasms mortality, Enhancer of Zeste Homolog 2 Protein metabolism, Female, Genetic Markers genetics, Histones metabolism, Humans, Prognosis, RNA Interference, RNA, Small Interfering genetics, Antineoplastic Agents pharmacology, Antineoplastic Combined Chemotherapy Protocols pharmacology, Carcinogenesis drug effects, Endometrial Neoplasms drug therapy, Enhancer of Zeste Homolog 2 Protein antagonists & inhibitors, Enhancer of Zeste Homolog 2 Protein genetics, Indoles pharmacology, Pyridones pharmacology
- Abstract
The histone methyltransferase EZH2, a key epigenetic modifier, is known to be associated with human tumorigenesis. However, the physiological importance of EZH2 and its clinical relevance in endometrial cancer remain unclear. Hence, in the present study, we investigated the expression and function of EZH2 in endometrial cancer. In a quantitative real-time PCR analysis of 11 endometrial cancer cell lines and 52 clinical endometrial cancer specimens, EZH2 was significantly overexpressed in cancer cells and tissues compared to that in corresponding normal control cells and tissues. Kaplan-Meier survival analysis using data of the TCGA RNA-seq database and tissue microarrays (TMAs) indicated that EZH2 overexpression is associated with endometrial cancer prognosis. In addition, knockdown of EZH2 using specific siRNAs resulted in growth suppression and apoptosis induction of endometrial cancer cells, accompanied by attenuation of H3K27 trimethylation. Consistent with these results, treatment with GSK126, a specific EZH2 inhibitor, suppressed endometrial cancer cell growth and decreased the number of cancer cell colonies. Furthermore, GSK126 showed additive effects with doxorubicin or cisplatin, which are conventional drugs for treatment of endometrial cancer. Further studies should explore the therapeutic potential of inhibiting EZH2 in patients with endometrial cancer.
- Published
- 2017
- Full Text
- View/download PDF
5. Role of EZH2 in cancer stem cells: from biological insight to a therapeutic target.
- Author
-
Wen Y, Cai J, Hou Y, Huang Z, and Wang Z
- Subjects
- Enhancer of Zeste Homolog 2 Protein metabolism, Humans, Neoplastic Stem Cells pathology, Enhancer of Zeste Homolog 2 Protein genetics, Epigenesis, Genetic genetics, Neoplastic Stem Cells metabolism, Polycomb Repressive Complex 2 genetics
- Abstract
Epigenetic modifications in cancer stem cells largely result in phenotypic and functional heterogeneity in many solid tumors. Increasing evidence indicates that enhancer of zeste homolog 2 (EZH2), the catalytic subunit of Polycomb repressor complex 2, is highly expressed in cancer stem cells of numerous malignant tumors and has a critical function in cancer stem cell expansion and maintenance. Here, we review up-to-date information regarding EZH2 expression patterns, functions, and molecular mechanisms in cancer stem cells in various malignant tumors and discuss the therapeutic potential of targeting EZH2 in tumors.
- Published
- 2017
- Full Text
- View/download PDF
6. The HDAC inhibitor valproate induces a bivalent status of the CD20 promoter in CLL patients suggesting distinct epigenetic regulation of CD20 expression in CLL in vivo.
- Author
-
Scialdone A, Hasni MS, Damm JK, Lennartsson A, Gullberg U, and Drott K
- Subjects
- Aged, Antigens, CD20 metabolism, Cell Line, Tumor, Enhancer of Zeste Homolog 2 Protein genetics, Enhancer of Zeste Homolog 2 Protein metabolism, Epigenesis, Genetic, Female, Gene Expression Regulation, Neoplastic, Histones metabolism, Humans, Leukemia, Lymphocytic, Chronic, B-Cell immunology, Male, Promoter Regions, Genetic genetics, Rituximab therapeutic use, Sequence Deletion genetics, Antigens, CD20 genetics, B-Lymphocytes immunology, Histone Deacetylase Inhibitors therapeutic use, Leukemia, Lymphocytic, Chronic, B-Cell genetics, Valproic Acid therapeutic use
- Abstract
Treatment with anti-CD20 antibodies is only moderately efficient in chronic lymphocytic leukemia (CLL), a feature which has been explained by the inherently low CD20 expression in CLL. It has been shown that CD20 is epigenetically regulated and that histone deacetylase inhibitors (HDACis) can increase CD20 expression in vitro in CLL. To assess whether HDACis can upregulate CD20 also in vivo in CLL, the HDACi valproate was given to three del13q/NOTCH1wt CLL patients and CD20 levels were analysed (the PREVAIL study). Valproate treatment resulted in expected global activating histone modifications suggesting HDAC inhibitory effects. However, although valproate induced expression of CD20 mRNA and protein in the del13q/NOTCH1wt I83-E95 CLL cell line, no such effects were observed in the patients studied. In contrast to the cell line, in patients valproate treatment resulted in transient recruitment of the transcriptional repressor EZH2 to the CD20 promoter, correlating to an increase of the repressive histone mark H3K27me3. This suggests that valproate-mediated induction of CD20 may be hampered by EZH2 mediated H3K27me3 in vivo in CLL. Moreover, valproate treatment resulted in induction of EZH2 and global H3K27me3 in patient cells, suggesting transcriptionally repressive effects of valproate in CLL. Our results suggest new in vivo mechanisms of HDACis which may have implications on the design of future clinical trials in B-cell malignancies.
- Published
- 2017
- Full Text
- View/download PDF
7. Functional and therapeutic significance of EZH2 in urological cancers.
- Author
-
Liu X, Wu Q, and Li L
- Subjects
- Cell Transformation, Neoplastic genetics, Enhancer of Zeste Homolog 2 Protein metabolism, Gene Expression Regulation, Neoplastic, Humans, Male, Enhancer of Zeste Homolog 2 Protein genetics, Urologic Neoplasms genetics
- Abstract
The enhancer of zeste homolog 2 (EZH2) is a core subunit of the polycomb repressor complex 2 (PRC2), which is overexpressed in numerous cancers and mutated in several others. Notably, EZH2 acts not only a critical epigenetic repressor through its role in histone methylation, it is also an activator of gene expression, acting through multiple signaling pathways in distinct cancer types. Increasing evidence suggests that EZH2 is an oncogene and is central to initiation, growth and progression of urological cancers. In this review, we highlight the critical role of EZH2 as a master regulator of tumorigenesis in the prostate, bladder and the kidney through epigenetic control of transcription as well as a modulation of various critical signaling pathways. We also discuss the promise and challenges for EZH2 inhibitors as future anticancer therapeutics, some of which are currently in clinical trials.
- Published
- 2017
- Full Text
- View/download PDF
8. DNA methylation of METTL7A gene body regulates its transcriptional level in thyroid cancer.
- Author
-
Zhou S, Shen Y, Zheng M, Wang L, Che R, Hu W, and Li P
- Subjects
- Cell Line, Tumor, CpG Islands, Down-Regulation, Epigenesis, Genetic, Exons, Gene Expression Regulation, Neoplastic, Humans, Thyroid Neoplasms metabolism, DNA Methylation, Enhancer of Zeste Homolog 2 Protein metabolism, Membrane Proteins genetics, Methyltransferases genetics, Thyroid Neoplasms genetics
- Abstract
DNA methylation is the best-studied epigenetic mechanism for regulating gene transcription and maintaining genome stability. Current research progress of transcriptional regulation by DNA methylation mostly focuses on promoter region where hypomethylated CpG islands are present transcriptional activity, as hypermethylated CpG islands generally result in gene repression. Recently, the DNA methylation patterns across the gene body (intragenic methylation) have increasingly attracted attention towards their role in transcriptional regulation and efficiency, due to the improvement of numerous genome-wide DNA methylation profiling studies. However, the function and mechanism of gene body methylation is still unclear. In this study, we revealed that the methylation level of METTL7A, a seldom studied gene, was downregulated in thyroid cancer compared to normal thyroid cells in vivo and in vitro. Moreover, we determined the methylation level of one CpG site at the exon of the METTL7A gene body impacted the transcriptional activity. Through generating a mutation of this CpG site (CG to CC) of METTL7A exogenous vector artificially in vitro, we observed higher RNA polymerase II recruitment and a declined enrichment of methyl-CpG binding protein-2 in gene body of METTL7A, in papillary thryoid cancer cells (BCPAP) compared to normal thryoid cells. Finally, we revealed that EZH2, a subunit of polycomb repressor complex 2, dominant in thyroid cancer, might be responsible for regulating gene body methylation of METTL7A. Our study depicted the DNA methylation patterns and the transcriptional regulatory mechanism of the gene body in thyroid cancer. Furthermore, this study provides new insight into potential future avenues, for therapies targeting cancer.
- Published
- 2017
- Full Text
- View/download PDF
9. Long intergenic noncoding RNA 00673 promotes non-small-cell lung cancer metastasis by binding with EZH2 and causing epigenetic silencing of HOXA5.
- Author
-
Ma C, Wu G, Zhu Q, Liu H, Yao Y, Yuan D, Liu Y, Lv T, and Song Y
- Subjects
- A549 Cells, Animals, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung metabolism, Cell Line, Tumor, Cell Movement, Cell Proliferation, Enhancer of Zeste Homolog 2 Protein metabolism, Epigenesis, Genetic, Gene Expression Regulation, Neoplastic, Homeodomain Proteins metabolism, Humans, Lung Neoplasms genetics, Lung Neoplasms metabolism, Lymphatic Metastasis, Mice, Neoplasm Staging, Neoplasm Transplantation, Prognosis, Promoter Regions, Genetic, Protein Binding, Carcinoma, Non-Small-Cell Lung pathology, Enhancer of Zeste Homolog 2 Protein genetics, Homeodomain Proteins genetics, Lung Neoplasms pathology, RNA, Long Noncoding genetics
- Abstract
Metastasis of cancer cells is a key impediment to favorable outcomes of cancer treatment. Functional roles of long noncoding RNAs in several biological processes, including metastasis, have recently been discovered. In our previous work, we reported a positive correlation of increased expression of linc00673 in NSCLC tissues with tumor size, lymph node metastasis, TNM stage, and increased proliferation of NSCLC cells, both, in vitro and in vivo. In this study, we demonstrate that ectopic expression of linc00673 promotes migration and invasion of NSCLC cells. Furthermore, our results indicate that linc00673 could silence HOXA5 expression by recruiting epigenetic repressor, EZH2, at its promoter regions. HOXA5 was identified as a tumor suppressor gene, which inhibited NSCLC cell metastasis by regulating cytoskeletal remodeling. To summarize, we for the first time identified the role of lin00673 in promoting invasion and migration of NSCLC cells. Insights from this study may help to identify novel therapeutic targets for NSCLC.
- Published
- 2017
- Full Text
- View/download PDF
10. Long non-coding RNA LUCAT1 is associated with poor prognosis in human non-small lung cancer and regulates cell proliferation via epigenetically repressing p21 and p57 expression.
- Author
-
Sun Y, Jin SD, Zhu Q, Han L, Feng J, Lu XY, Wang W, Wang F, and Guo RH
- Subjects
- Adult, Aged, Apoptosis genetics, Carcinoma, Non-Small-Cell Lung mortality, Carcinoma, Non-Small-Cell Lung pathology, Cell Line, Tumor, Cell Proliferation, Computational Biology methods, Enhancer of Zeste Homolog 2 Protein metabolism, Female, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Gene Silencing, Humans, Lung Neoplasms mortality, Lung Neoplasms pathology, Male, Middle Aged, Models, Biological, Neoplasm Grading, Neoplasm Staging, Prognosis, Carcinoma, Non-Small-Cell Lung genetics, Cyclin-Dependent Kinase Inhibitor p21 genetics, Cyclin-Dependent Kinase Inhibitor p57 genetics, Epigenesis, Genetic, Lung Neoplasms genetics, RNA Interference, RNA, Long Noncoding genetics
- Abstract
Recently, long non-coding RNAs (lncRNAs) have been recognized as playing key roles in regulating cellular processes, such as proliferation, invasion, and metastasis. These lncRNAs have been shown to be abnormally expressed in tumorigenic processes. However, the role and clinical relevance of LUCAT1 in non-small-cell lung cancer (NSCLC) remain unclear. In this study, we found that the expression of LUCAT1 was significantly up-regulated in NSCLC tissues compared to non-tumor tissues, and its expression was associated with tumor size, tumor-node-metastasis (TNM) stage and overall survival (OS). Further experiments showed that LUCAT1 knockdown inhibited cell proliferation both in vitro and in vivo. Mechanistic investigations showed that LUCAT1 plays a key role in G0/G1 arrest. We further demonstrated that LUCAT1 was associated with polycomb repressor complexes (PRC2) and that this association was required for epigenetically repression of p21 and p57, thus contributing to the regulation of NSCLC cell cycle and proliferation. In summary, our results show that LUCAT1 could regulate tumorigenesis of NSCLC and be biomarker for poor prognosis in NSCLC.
- Published
- 2017
- Full Text
- View/download PDF
11. Long noncoding RNA HOTAIR promotes metastasis of renal cell carcinoma by up-regulating histone H3K27 demethylase JMJD3.
- Author
-
Xia M, Yao L, Zhang Q, Wang F, Mei H, Guo X, and Huang W
- Subjects
- Apoptosis genetics, Blotting, Western, Carcinoma, Renal Cell metabolism, Carcinoma, Renal Cell pathology, Cell Line, Tumor, Cell Movement genetics, Enhancer of Zeste Homolog 2 Protein genetics, Enhancer of Zeste Homolog 2 Protein metabolism, Gene Knockdown Techniques, Histones metabolism, Humans, Jumonji Domain-Containing Histone Demethylases metabolism, Kaplan-Meier Estimate, Kidney Neoplasms metabolism, Kidney Neoplasms pathology, Methylation, Neoplasm Metastasis, Reverse Transcriptase Polymerase Chain Reaction, Snail Family Transcription Factors genetics, Snail Family Transcription Factors metabolism, Up-Regulation, Carcinoma, Renal Cell genetics, Gene Expression Regulation, Neoplastic, Jumonji Domain-Containing Histone Demethylases genetics, Kidney Neoplasms genetics, RNA, Long Noncoding genetics
- Abstract
Long Noncoding RNAs (lncRNAs) are a kind of non-protein coding transcripts longer than 200 nucleotides, and play important roles in diverse biological processes, such as embryonic development and apoptosis. Homeobox (HOX) transcript antisense intergenic RNA (HOTAIR) is a negative prognostic factor in a variety of human cancers, such as breast, liver and lung cancers. HOTAIR can promote cancer cell metastasis by reprogramming chromatin organization. In the present study, HOTAIR expression was elevated in tissues of renal cell carcinoma compared to adjacent normal tissues, and positively correlated with metastasis (P<0.05). The cell migration was inhibited in scratch test and transwell assay after HOTAIR knockdown (P<0.05). Further researches revealed that histone demethylase JMJD3 was reduced and its target gene Snai1 expression was down-regulated after HOTAIR suppression (P<0.05). Meanwhile, the level of histone methytransferase EZH2 target gene PCDHB5 was increased (P<0.05). Collectively, these data suggest that HOTAIR is an important promoter in metastasis of renal cell carcinoma and also plays a dual regulatory role in chromatin state by effecting both histone metylation and demethylation at different gene loci.
- Published
- 2017
- Full Text
- View/download PDF
12. EZH2 expression is a prognostic biomarker in patients with colorectal cancer treated with anti-EGFR therapeutics.
- Author
-
Yamamoto I, Nosho K, Kanno S, Igarashi H, Kurihara H, Ishigami K, Ishiguro K, Mitsuhashi K, Maruyama R, Koide H, Okuda H, Hasegawa T, Sukawa Y, Okita K, Takemasa I, Yamamoto H, Shinomura Y, and Nakase H
- Subjects
- Antibodies, Monoclonal therapeutic use, Biomarkers, Tumor biosynthesis, Cetuximab therapeutic use, Colorectal Neoplasms genetics, Colorectal Neoplasms mortality, Disease-Free Survival, Enhancer of Zeste Homolog 2 Protein biosynthesis, Female, GTP Phosphohydrolases genetics, Humans, Male, Membrane Proteins genetics, MicroRNAs antagonists & inhibitors, MicroRNAs genetics, Middle Aged, Panitumumab, Proto-Oncogene Proteins B-raf genetics, Proto-Oncogene Proteins p21(ras) genetics, Treatment Outcome, Antineoplastic Agents therapeutic use, Biomarkers, Tumor metabolism, Colorectal Neoplasms drug therapy, Colorectal Neoplasms pathology, Enhancer of Zeste Homolog 2 Protein metabolism, ErbB Receptors antagonists & inhibitors
- Abstract
The polycomb group protein enhancer of zeste homolog 2 (EZH2) is a methyltransferase that suppresses microRNA-31 (miR-31) in various human malignancies including colorectal cancer. We recently suggested that miR-31 regulates the signaling pathway downstream of epidermal growth factor receptor (EGFR) in colorectal cancer. Therefore, we conducted this study for assessing the relationship between EZH2 expression and clinical outcomes in patients with colorectal cancer treated with anti-EGFR therapeutics. We immunohistochemically evaluated EZH2 expression and assessed miR-31 and gene mutations [KRAS (codon 61/146), NRAS (codon 12/13/61), and BRAF (codon 600)] in 109 patients with colorectal cancer harboring KRAS (codon 12/13) wild-type. We also evaluated the progression-free survival (PFS) and overall survival (OS). In the result, low EZH2 expression was significantly associated with shorter PFS (log-rank test: P = 0.023) and OS (P = 0.036) in patients with colorectal cancer. In the low-miR-31-expression group and the KRAS (codon 61/146), NRAS, and BRAF wild-type groups, a significantly shorter PFS (P = 0.022, P = 0.039, P = 0.021, and P = 0.036, respectively) was observed in the EZH2 low-expression groups than in the high-expression groups. In the multivariate analysis, low EZH2 expression was associated with a shorter PFS (P = 0.046), independent of the mutational status and miR-31. In conclusion, EZH2 expression was associated with survival in patients with colorectal cancer who were treated with anti-EGFR therapeutics. Moreover, low EZH2 expression was independently associated with shorter PFS in patients with cancer, suggesting that EZH2 expression is a useful additional prognostic biomarker for anti-EGFR therapy.
- Published
- 2017
- Full Text
- View/download PDF
13. EZH2 inhibition suppresses endometrial cancer progression via miR-361/Twist axis.
- Author
-
Ihira K, Dong P, Xiong Y, Watari H, Konno Y, Hanley SJ, Noguchi M, Hirata N, Suizu F, Yamada T, Kudo M, and Sakuragi N
- Subjects
- Animals, Cell Line, Tumor, Disease Progression, Endometrial Neoplasms genetics, Endometrial Neoplasms metabolism, Endometrial Neoplasms pathology, Enhancer of Zeste Homolog 2 Protein genetics, Enhancer of Zeste Homolog 2 Protein metabolism, Female, Humans, Mice, Mice, Inbred BALB C, Transfection, Xenograft Model Antitumor Assays, Endometrial Neoplasms drug therapy, Enhancer of Zeste Homolog 2 Protein antagonists & inhibitors, Enzyme Inhibitors pharmacology, Indazoles pharmacology, MicroRNAs metabolism, Nuclear Proteins metabolism, Pyridones pharmacology, Twist-Related Protein 1 metabolism
- Abstract
EZH2 inhibition and reactivation of tumor suppressor microRNAs (miRNAs) represent attractive anti-cancer therapeutic strategies. We found that EZH2-suppressed let 7b and miR-361, two likely tumor suppressors, inhibited endometrial cancer (EC) cell proliferation and invasion, and abrogated cancer stem cell-like properties. In EC cells, EZH2 induced and functioned together with YY1 to epigenetically suppress miR-361, which upregulated Twist, a direct target of miR-361. Treating EC cells with GSK343, a specific EZH2 inhibitor, mimicked the effects of siRNA-mediated EZH2 knockdown, upregulating miR-361 and downregulating Twist expression. Combining GSK343 with 5 AZA-2'-deoxycytidine synergistically suppressed cell proliferation and invasion in vitro, and decreased tumor size and weight in EC cell xenografted mice. Quantitative real-time PCR analysis of 24 primary EC tissues showed that lower let-7b and miR-361 levels were associated with worse patient outcomes. These results were validated in a larger EC patient dataset from The Cancer Genome Atlas. Our findings suggest that EZH2 drives EC progression by regulating miR-361/Twist signaling, and support EZH2 inhibition as a promising anti-EC therapeutic strategy.
- Published
- 2017
- Full Text
- View/download PDF
14. EZH2 inhibition in multiple myeloma downregulates myeloma associated oncogenes and upregulates microRNAs with potential tumor suppressor functions.
- Author
-
Alzrigat M, Párraga AA, Agarwal P, Zureigat H, Österborg A, Nahi H, Ma A, Jin J, Nilsson K, Öberg F, Kalushkova A, and Jernberg-Wiklund H
- Subjects
- Cell Line, Tumor, Enhancer of Zeste Homolog 2 Protein genetics, Enhancer of Zeste Homolog 2 Protein metabolism, Enzyme Inhibitors pharmacology, Gene Expression Regulation, Neoplastic, Humans, Interferon Regulatory Factors genetics, Interferon Regulatory Factors metabolism, MicroRNAs metabolism, Multiple Myeloma enzymology, Multiple Myeloma genetics, Multiple Myeloma pathology, Positive Regulatory Domain I-Binding Factor 1 genetics, Positive Regulatory Domain I-Binding Factor 1 metabolism, Proto-Oncogene Proteins c-myc genetics, Proto-Oncogene Proteins c-myc metabolism, Signal Transduction drug effects, Transcriptome, Tumor Cells, Cultured, X-Box Binding Protein 1 genetics, X-Box Binding Protein 1 metabolism, Antineoplastic Agents pharmacology, Enhancer of Zeste Homolog 2 Protein antagonists & inhibitors, Genes, Tumor Suppressor, MicroRNAs genetics, Multiple Myeloma drug therapy, Oncogenes, Pyridones pharmacology
- Abstract
Multiple Myeloma (MM) is a plasma cell tumor localized to the bone marrow (BM). Despite the fact that current treatment strategies have improved patients' median survival time, MM remains incurable. Epigenetic aberrations are emerging as important players in tumorigenesis making them attractive targets for therapy in cancer including MM. Recently, we suggested the polycomb repressive complex 2 (PRC2) as a common denominator of gene silencing in MM and presented the PRC2 enzymatic subunit enhancer of zeste homolog 2 (EZH2) as a potential therapeutic target in MM. Here we further dissect the anti-myeloma mechanisms mediated by EZH2 inhibition and show that pharmacological inhibition of EZH2 reduces the expression of MM-associated oncogenes; IRF-4, XBP-1, PRDM1/BLIMP-1 and c-MYC. We show that EZH2 inhibition reactivates the expression of microRNAs with tumor suppressor functions predicted to target MM-associated oncogenes; primarily miR-125a-3p and miR-320c. ChIP analysis reveals that miR-125a-3p and miR-320c are targets of EZH2 and H3K27me3 in MM cell lines and primary cells. Our results further highlight that polycomb-mediated silencing in MM includes microRNAs with tumor suppressor activity. This novel role strengthens the oncogenic features of EZH2 and its potential as a therapeutic target in MM.
- Published
- 2017
- Full Text
- View/download PDF
15. Opposing roles of PIK3CA gene alterations to EZH2 signaling in non-muscle invasive bladder cancer.
- Author
-
Segovia C, Martínez-Fernández M, Dueñas M, Rubio C, López-Calderón FF, Costa C, Saiz-Ladera C, Fernández-Grajera M, Duarte J, Muñoz HG, de la Rosa F, Villacampa F, Castellano D, and Paramio JM
- Subjects
- Biomarkers, Tumor metabolism, Cell Line, Tumor, Class I Phosphatidylinositol 3-Kinases metabolism, Disease Progression, Enhancer of Zeste Homolog 2 Protein metabolism, Gene Expression Regulation, Neoplastic, Histones metabolism, Humans, Kaplan-Meier Estimate, Methylation, MicroRNAs genetics, MicroRNAs metabolism, Neoplasm Invasiveness, Neoplasm Recurrence, Local, Phosphorylation, Proto-Oncogene Proteins c-akt metabolism, Time Factors, Transcription, Genetic, Urinary Bladder Neoplasms enzymology, Urinary Bladder Neoplasms pathology, Urinary Bladder Neoplasms therapy, Biomarkers, Tumor genetics, Class I Phosphatidylinositol 3-Kinases genetics, Enhancer of Zeste Homolog 2 Protein genetics, Mutation, Signal Transduction, Urinary Bladder Neoplasms genetics
- Abstract
The high rates of tumor recurrence and progression represent a major clinical problem in non-muscle invasive bladder cancer. Previous data showed that EZH2-dependent signaling mediates these processes, whereas the frequent alterations of PIK3CA gene (copy gains and mutations) are predictive of reduced recurrence. Here we show, using clinical samples and bladder cancer cell lines, a functional interaction between EZH2- and PIK3CA-dependent signaling pathways. PIK3CA alterations mediated, on the one hand, the increased expression of two miRNAs, miR-101 and miR-138, which posttranscriptionally downregulate EZH2 expression. On the other hand, PIK3CA alterations facilitate the activation of Akt which phosphorylates EZH2 on Ser21, precluding the trimethylation of histone H3 in K27. Remarkably the increased expression of miR101 or miR138 and the expression of Ser21-phosphorylated EZH2 are good prognostic factors regarding non-muscle invasive bladder cancer recurrence and progression. Collectively, this study provides molecular evidences indicating that the gene expression rewiring occurring in primary bladder tumors, associated with increased EZH2 expression and activity and mediating the increased recurrence and progression risk, are prevented by PIK3CA-dependent signaling. This molecular process may have deep implications in the management of bladder cancer patients and in the design of novel molecularly targeted therapeutic approaches.
- Published
- 2017
- Full Text
- View/download PDF
16. Blocking EZH2 methylation transferase activity by GSK126 decreases stem cell-like myeloma cells.
- Author
-
Zeng D, Liu M, and Pan J
- Subjects
- Animals, Apoptosis drug effects, Bortezomib pharmacology, Cell Line, Tumor, Cell Proliferation drug effects, Disease Models, Animal, Drug Synergism, Enzyme Activation drug effects, Humans, Male, Mice, Mitochondria drug effects, Mitochondria metabolism, Multiple Myeloma drug therapy, Multiple Myeloma pathology, Myeloid Cell Leukemia Sequence 1 Protein metabolism, Proteolysis, Wnt Signaling Pathway drug effects, Xenograft Model Antitumor Assays, Enhancer of Zeste Homolog 2 Protein antagonists & inhibitors, Enhancer of Zeste Homolog 2 Protein metabolism, Indoles pharmacology, Multiple Myeloma metabolism, Neoplastic Stem Cells drug effects, Neoplastic Stem Cells metabolism, Pyridones pharmacology
- Abstract
EZH2 is a critical epigenetic regulator that is deregulated in various types of cancers including multiple myeloma (MM). In the present study, we hypothesized that targeting EZH2 might induce apoptosis in myeloma cells including stem cell-like cells (CSCs). We investigated the effect of EZH2 inhibition on MM cells using a potent inhibitor (GSK126). The results showed that GSK126 effectively abrogated the methylated histone 3 (H3K27me3) level in MM.1S and LP1 cells, and inhibited the number of live cells and colony formation in soft agar of six MM cell lines. GSK126 induced massive apoptosis in MM.1S, LP1 and RPMI8226 cells. Progressive release of mitochondrial cytochrome c and AIF into the cytosol was detected in GSK126-treated MM cells. GSK126 treatment elicited caspase-3-dependent MCL-1 cleavage with accumulation of proapoptotic truncated MCL-1. These results suggested that GSK126 triggers the intrinsic mitochondrial apoptosis pathway. Enhanced apoptosis was observed in the combination of GSK126 with bortezomib. Using ALDH and side population (SP) assays to characterize CSCs, we found that GSK126 eliminated the stem-like myeloma cells by blocking the Wnt/β-catenin pathway. The in vivo anti-tumor effect of GSK126 was confirmed by using RPMI8226 cells in a xenograft mouse model. In conclusion, our findings suggest that EZH2 inactivation by GSK126 is effective in killing MM cells and CSCs as a single agent or in combination with bortezomib. Clinical trial of GSK126 in patients with MM may be warranted.
- Published
- 2017
- Full Text
- View/download PDF
17. Elevated expression of ZNF217 promotes prostate cancer growth by restraining ferroportin-conducted iron egress.
- Author
-
Jiang X, Zhang C, Qi S, Guo S, Chen Y, Du E, Zhang H, Wang X, Liu R, Qiao B, Yang K, Zhang Z, and Xu Y
- Subjects
- Aged, Animals, Carcinogenesis, Cation Transport Proteins genetics, Cell Growth Processes, Cell Line, Tumor, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Enhancer of Zeste Homolog 2 Protein genetics, Enhancer of Zeste Homolog 2 Protein metabolism, Gene Expression Regulation, Neoplastic, Histones genetics, Histones metabolism, Humans, Ion Transport, Male, Mice, Mice, Inbred BALB C, Mice, Nude, Prostatic Neoplasms genetics, Trans-Activators genetics, Transcription Factors genetics, Transcription Factors metabolism, Up-Regulation, Xenograft Model Antitumor Assays, Ferroportin, Cation Transport Proteins metabolism, Iron metabolism, Prostatic Neoplasms metabolism, Trans-Activators metabolism
- Abstract
Although we and other studies indicated ZNF217 expression was increased in prostate cancer (PCa), the factors mediating its misregulated expression and their oncogenic activity remain largely unexplored. Recent evidence demonstrated that ferroportin (FPN) reduction lead to decreased iron export and increased intercellular iron that consequently aggravates the oncogenic effects of iron. In the present study, ZNF217 was identified as a transcriptional repressor that inhibits FPN expression. Increased of ZNF217 expression led to decreased FPN concentration, coupled with resultant intracellular iron retention, increased iron-related cellular activities and enhanced tumor cell growth. In contrast, decreased of ZNF217 expression restrained tumor cell growth by promoting FPN-driven iron egress. Mechanistic investigation manifested that ZNF217 facilitated the H3K27me3 levels of FPN promoter by interacting with EZH2. Besides, we also found that MAZ increased the transcription level of ZNF217, and subsequently inhibited the FPN expression and their iron-related activities. Strikingly, the expression of MAZ, EZH2 and ZNF217 were concurrently upregulated in PCa, leading to decreased expression of FPN, which induce disordered iron metabolism. Collectively, this study underscored that elevated expression of ZNF217 promotes prostate cancer growth by restraining FPN-conducted iron egress.
- Published
- 2016
- Full Text
- View/download PDF
18. Epigenetic regulation of cancer biology and anti-tumor immunity by EZH2.
- Author
-
Christofides A, Karantanos T, Bardhan K, and Boussiotis VA
- Subjects
- Animals, Antineoplastic Agents therapeutic use, Chromatin Assembly and Disassembly, Enhancer of Zeste Homolog 2 Protein antagonists & inhibitors, Enhancer of Zeste Homolog 2 Protein immunology, Enhancer of Zeste Homolog 2 Protein metabolism, Gene Expression Regulation, Enzymologic, Gene Expression Regulation, Neoplastic, Humans, Molecular Targeted Therapy, Neoplasms drug therapy, Neoplasms enzymology, Neoplasms immunology, Signal Transduction, DNA Methylation drug effects, Enhancer of Zeste Homolog 2 Protein genetics, Epigenesis, Genetic drug effects, Neoplasms genetics, Tumor Escape drug effects
- Abstract
Polycomb group proteins regulate chromatin structure and have an important regulatory role on gene expression in various cell types. Two polycomb group complexes (Polycomb repressive complex 1 (PRC1) and 2 (PRC2)) have been identified in mammalian cells. Both PRC1 and PRC2 compact chromatin, and also catalyze histone modifications. PRC1 mediates monoubiquitination of histone H2A, whereas PRC2 catalyzes methylation of histone H3 on lysine 27. These alterations of histones can lead to altered gene expression patterns by regulating chromatin structure. Numerous studies have highlighted the role of the PRC2 catalytic component enhancer of zeste homolog 2 (EZH2) in neoplastic development and progression, and EZH2 mutations have been identified in various malignancies. Through modulating the expression of critical genes, EZH2 is actively involved in fundamental cellular processes such as cell cycle progression, cell proliferation, differentiation and apoptosis. In addition to cancer cells, EZH2 also has a decisive role in the differentiation and function of T effector and T regulatory cells. In this review we summarize the recent progress regarding the role of EZH2 in human malignancies, highlight the molecular mechanisms by which EZH2 aberrations promote the pathogenesis of cancer, and discuss the anti-tumor effects of EZH2 targeting via activating direct anti-cancer mechanisms and anti-tumor immunity.
- Published
- 2016
- Full Text
- View/download PDF
19. EZH2 inhibition promotes epithelial-to-mesenchymal transition in ovarian cancer cells.
- Author
-
Cardenas H, Zhao J, Vieth E, Nephew KP, and Matei D
- Subjects
- Cell Line, Tumor, Enhancer of Zeste Homolog 2 Protein antagonists & inhibitors, Enhancer of Zeste Homolog 2 Protein metabolism, Epithelial-Mesenchymal Transition drug effects, Female, Histones metabolism, Humans, Indoles pharmacology, Methylation drug effects, Ovarian Neoplasms genetics, Ovarian Neoplasms metabolism, Ovarian Neoplasms pathology, Promoter Regions, Genetic genetics, Protein Binding, Pyridones pharmacology, RNA Interference, Transforming Growth Factor beta pharmacology, Zinc Finger E-box Binding Homeobox 2 genetics, Zinc Finger E-box Binding Homeobox 2 metabolism, Enhancer of Zeste Homolog 2 Protein genetics, Epithelial-Mesenchymal Transition genetics, Gene Expression Regulation, Neoplastic
- Abstract
Cancer cells acquire essential characteristics for metastatic dissemination through the process of epithelial-to-mesenchymal transition (EMT), which is regulated by gene expression and chromatin remodeling changes. The enhancer of zeste homolog 2 (EZH2), the catalytic subunit of the polycomb repressive complex 2 (PRC2), catalyzes trimethylation of lysine 27 of histone H3 (H3K27me3) to repress gene transcription. Here we report the functional roles of EZH2-catalyzed H3K27me3 during EMT in ovarian cancer (OC) cells. TGF-β-induced EMT in SKOV3 OC cells was associated with decreased levels of EZH2 and H3K27me3 (P<0.05). These effects were delayed (~72 h relative to EMT initiation) and coincided with increased (>15-fold) expression of EMT-associated transcription factors ZEB2 and SNAI2. EZH2 knockdown (using siRNA) or enzymatic inhibition (by GSK126) induced EMT-like changes in OC cells. The EMT regulator ZEB2 was upregulated in cells treated with either approach. Furthermore, TGF-β enhanced expression of ZEB2 in EZH2 siRNA- or GSK126-treated cells (P<0.01), suggesting that H3K27me3 plays a role in TGF-β-stimulated ZEB2 induction. Chromatin immunoprecipitation assays confirmed that TGF-β treatment decreased binding of EZH2 and H3K27me3 to the ZEB2 promoter (P<0.05). In all, these results demonstrate that EZH2, by repressing ZEB2, is required for the maintenance of an epithelial phenotype in OC cells.
- Published
- 2016
- Full Text
- View/download PDF
20. LncRNA H19 confers chemoresistance in ERα-positive breast cancer through epigenetic silencing of the pro-apoptotic gene BIK.
- Author
-
Si X, Zang R, Zhang E, Liu Y, Shi X, Zhang E, Shao L, Li A, Yang N, Han X, Pan B, Zhang Z, Sun L, and Sun Y
- Subjects
- Apoptosis Regulatory Proteins metabolism, Breast Neoplasms metabolism, Breast Neoplasms pathology, Dose-Response Relationship, Drug, Enhancer of Zeste Homolog 2 Protein genetics, Enhancer of Zeste Homolog 2 Protein metabolism, Estrogen Receptor alpha drug effects, Female, Gene Expression Regulation, Neoplastic, Humans, MCF-7 Cells, Membrane Proteins metabolism, Mitochondrial Proteins, Proto-Oncogene Proteins c-bcl-2 genetics, Proto-Oncogene Proteins c-bcl-2 metabolism, RNA Interference, RNA, Long Noncoding metabolism, Signal Transduction, Transcription, Genetic, Transfection, Antineoplastic Agents pharmacology, Apoptosis drug effects, Apoptosis Regulatory Proteins genetics, Breast Neoplasms drug therapy, Breast Neoplasms genetics, DNA Methylation, Drug Resistance, Neoplasm genetics, Epigenesis, Genetic, Estrogen Receptor alpha metabolism, Membrane Proteins genetics, Paclitaxel pharmacology, RNA, Long Noncoding genetics
- Abstract
Breast cancer is a common malignancy in women. Acquisition of drug resistance is one of the main obstacles encountered in breast cancer therapy. Long non-coding RNA (lncRNA) has been demonstrated to play vital roles in both development and tumorigenesis. However, the relationship between lncRNAs and the development of chemoresistance is not well established. In the present study, the high expression of lncRNA H19 was identified as a powerful factor associated with paclitaxel (PTX) resistance in ERα-positive breast cancer cells, but not in ERα-negative breast cancer cells. LncRNA H19 attenuated cell apoptosis in response to PTX treatment by inhibiting transcription of pro-apoptotic genes BIK and NOXA. H19 was further confirmed to suppress the promoter activity of BIK by recruiting EZH2 and by trimethylating the histone H3 at lysine 27. Interestingly, our data showed that lncRNA H19 was one of the downstream target molecules of ERα. Altered ERα expression may therefore change H19 levels to modulate the apoptosis response to chemotherapy in breast cancer cells. Our data suggest that the ERα-H19-BIK signaling axis plays an important role in promoting chemoresistance.
- Published
- 2016
- Full Text
- View/download PDF
21. Integrated bioinformatics analysis of chromatin regulator EZH2 in regulating mRNA and lncRNA expression by ChIP sequencing and RNA sequencing.
- Author
-
Li Y, Luo M, Shi X, Lu Z, Sun S, Huang J, Chen Z, and He J
- Subjects
- Binding Sites, Cell Line, Tumor, Databases, Genetic, Enhancer of Zeste Homolog 2 Protein metabolism, Gene Regulatory Networks, Humans, Predictive Value of Tests, Promoter Regions, Genetic, RNA, Long Noncoding metabolism, RNA, Messenger metabolism, Signal Transduction, Transcription, Genetic, Chromatin Assembly and Disassembly, Chromatin Immunoprecipitation, Computational Biology methods, Enhancer of Zeste Homolog 2 Protein genetics, Gene Expression Regulation, Neoplastic, RNA, Long Noncoding genetics, RNA, Messenger genetics, Sequence Analysis, RNA
- Abstract
Enhancer of zeste homolog 2 (EZH2), a dynamic chromatin regulator in cancer, represents a potential therapeutic target showing early signs of promise in clinical trials. EZH2 ChIP sequencing data in 19 cell lines and RNA sequencing data in ten cancer types were downloaded from GEO and TCGA, respectively. Integrated ChIP sequencing analysis and co-expressing analysis were conducted and both mRNA and long noncoding RNA (lncRNA) targets were detected. We detected a median of 4,672 mRNA targets and 4,024 lncRNA targets regulated by EZH2 in 19 cell lines. 20 mRNA targets and 27 lncRNA targets were found in all 19 cell lines. These mRNA targets were enriched in pathways in cancer, Hippo, Wnt, MAPK and PI3K-Akt pathways. Co-expression analysis confirmed numerous targets, mRNA genes (RRAS, TGFBR2, NUF2 and PRC1) and lncRNA genes (lncRNA LINC00261, DIO3OS, RP11-307C12.11 and RP11-98D18.9) were potential targets and were significantly correlated with EZH2. We predicted genome-wide potential targets and the role of EZH2 in regulating as a transcriptional suppressor or activator which could pave the way for mechanism studies and the targeted therapy of EZH2 in cancer.
- Published
- 2016
- Full Text
- View/download PDF
22. Targeting EZH1 and EZH2 contributes to the suppression of fibrosis-associated genes by miR-214-3p in cardiac myofibroblasts.
- Author
-
Zhu WS, Tang CM, Xiao Z, Zhu JN, Lin QX, Fu YH, Hu ZQ, Zhang Z, Yang M, Zheng XL, Wu SL, and Shan ZX
- Subjects
- 3' Untranslated Regions, Angiotensin II, Animals, Binding Sites, Cardiomyopathies chemically induced, Cardiomyopathies genetics, Cardiomyopathies metabolism, Cells, Cultured, Collagen Type I genetics, Collagen Type I metabolism, Collagen Type I, alpha 1 Chain, Collagen Type III genetics, Collagen Type III metabolism, Disease Models, Animal, Enhancer of Zeste Homolog 2 Protein genetics, Fibrosis, Gene Expression Regulation, Male, Mice, Inbred C57BL, MicroRNAs genetics, Myocardium pathology, Myofibroblasts pathology, NF-kappa B metabolism, Oligonucleotides genetics, Oligonucleotides metabolism, PPAR gamma genetics, PPAR gamma metabolism, Polycomb Repressive Complex 2 genetics, RNA Interference, Signal Transduction, Transfection, Cardiomyopathies prevention & control, Enhancer of Zeste Homolog 2 Protein metabolism, MicroRNAs metabolism, Myocardium metabolism, Myofibroblasts metabolism, Polycomb Repressive Complex 2 metabolism
- Abstract
The role of microRNA-214-3p (miR-214-3p) in cardiac fibrosis was not well illustrated. The present study aimed to investigate the expression and potential target of miR-214-3p in angiotensin II (Ang-II)-induced cardiac fibrosis. MiR-214-3p was markedly decreased in the fibrotic myocardium of a mouse Ang-II infusion model, but was upregulated in Ang-II-treated mouse myofibroblasts. Cardiac fibrosis was shown attenuated in Ang-II-infused mice received tail vein injection of miR-214-3p agomir. Consistently, miR-214-3p inhibited the expression of Col1a1 and Col3a1 in mouse myofibroblasts in vitro. MiR-214-3p could bind the 3'-UTRs of enhancer of zeste homolog 1 (EZH1) and -2, and suppressed EZH1 and -2 expressions at the transcriptional level. Functionally, miR-214-3p mimic, in parallel to EZH1 siRNA and EZH2 siRNA, could enhance peroxisome proliferator-activated receptor-γ (PPAR-γ) expression and inhibited the expression of Col1a1 and Col3a1 in myofibroblasts. In addition, enforced expression of EZH1 and -2, and knockdown of PPAR-γ resulted in the increase of Col1a1 and Col3a1 in myofibroblasts. Moreover, the NF-κB signal pathway was verified to mediate Ang-II-induced miR-214-3p expression in myofibroblasts. Taken together, our results revealed that EZH1 and -2 were novel targets of miR-214-3p, and miR-214-3p might be one potential miRNA for the prevention of cardiac fibrosis.
- Published
- 2016
- Full Text
- View/download PDF
23. Loss of long noncoding RNA FOXF1-AS1 regulates epithelial-mesenchymal transition, stemness and metastasis of non-small cell lung cancer cells.
- Author
-
Miao L, Huang Z, Zengli Z, Li H, Chen Q, Yao C, Cai H, Xiao Y, Xia H, and Wang Y
- Subjects
- Carcinoma, Non-Small-Cell Lung metabolism, Carcinoma, Non-Small-Cell Lung pathology, Cell Line, Tumor, Cell Movement genetics, Down-Regulation, Enhancer of Zeste Homolog 2 Protein metabolism, Forkhead Transcription Factors genetics, Forkhead Transcription Factors metabolism, Gene Expression Profiling methods, Humans, Lung Neoplasms metabolism, Lung Neoplasms pathology, Neoplasm Metastasis, Carcinoma, Non-Small-Cell Lung genetics, Cell Self Renewal, Enhancer of Zeste Homolog 2 Protein genetics, Epithelial-Mesenchymal Transition genetics, Gene Expression Regulation, Neoplastic, Lung Neoplasms genetics, RNA, Long Noncoding genetics
- Abstract
Although recent evidence shows that long noncoding RNAs (lncRNAs) are involved in the regulation of gene expression and cancer progression, the understanding of the role of lncRNAs in lung cancer metastasis is still limited. To identify novel lncRNAs in non-small cell lung cancer (NSCLC), we profile NSCLC tumor and matched normal samples using GeneChip® Human Gene 2.0 ST Array, which provides the most accurate, sensitive, and comprehensive measurement of protein coding and lncRNA transcripts. We identified a panel of key factors dysregulated in lung cancer. Among them, the expression of FOXF1-AS1 was significantly downregulated in lung cancer. Stable overexpression of FOXF1-AS1 inhibits lung cancer cell migration and invasion by regulating EMT. Meanwhile, loss of FOXF1-AS1 mediates stem-like properties of lung cancer cells. Interestingly, we found that FOXF1-AS1 physically associates with PRC2 components EZH2 and loss of FOXF1-AS1 mediates cell migration and stem-like properties require EZH2. Loss of FOXF1-AS1 is also correlated with downregulation of FOXF1 in lung cancer. These results suggested that FOXF1-AS1 might regulate EMT, stemness and metastasis of NSCLC cells via EZH2, indicating it as a therapeutic target for future treatment of NSCLC.
- Published
- 2016
- Full Text
- View/download PDF
24. Long non-coding RNA HoxA-AS3 interacts with EZH2 to regulate lineage commitment of mesenchymal stem cells.
- Author
-
Zhu XX, Yan YW, Chen D, Ai CZ, Lu X, Xu SS, Jiang S, Zhong GS, Chen DB, and Jiang YZ
- Subjects
- Animals, Cell Differentiation, Cell Proliferation, Cells, Cultured, Enhancer of Zeste Homolog 2 Protein genetics, Epigenesis, Genetic, Gene Expression Regulation, Neoplastic, Gene Silencing, Humans, Mesenchymal Stem Cells metabolism, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Nude, Osteoblasts cytology, Osteoblasts metabolism, RNA, Long Noncoding genetics, Adipogenesis genetics, Cell Lineage genetics, Enhancer of Zeste Homolog 2 Protein metabolism, Mesenchymal Stem Cells cytology, Osteogenesis genetics, RNA, Long Noncoding metabolism
- Abstract
Long non-coding RNAs (lncRNAs) play an important role in gene regulation and are involving in diverse cellular processes. However, their roles in reprogramming of gene expression profiles during lineage commitment and maturation of mesenchymal stem cells (MSCs) remain poorly understood. In the current study, we characterize the expression of a lncRNA, HoxA-AS3, during the differentiation of MSCs. We showed that HoxA-AS3 is increased upon adipogenic induction of MSCs, while HoxA-AS3 remains unaltered during osteogenic induction. Silencing of HoxA-AS3 in MSCs resulted in decreased adipogenesis and expression of adipogenic markers, PPARG, CEBPA, FABP4 and ADIPOQ. Conversely, knockdown of HoxA-AS3 expression in MSCs exhibited an enhanced osteogenesis and osteogenic markers expression, including RUNX2, SP7, COL1A1, IBSP, BGLAP and SPP1. Mechanistically, HoxA-AS3 interacts with Enhancer Of Zeste 2 (EZH2) and is required for H3 lysine-27 trimethylation (H3K27me3) of key osteogenic transcription factor Runx2. Our data reveal that HoxA-AS3 acts as an epigenetic switch that determines the lineage specification of MSC.
- Published
- 2016
- Full Text
- View/download PDF
25. A polycomb-mediated epigenetic field defect precedes invasive cervical carcinoma.
- Author
-
Wijetunga NA, Ben-Dayan M, Tozour J, Burk RD, Schlecht NF, Einstein MH, and Greally JM
- Subjects
- Biopsy, Carcinogenesis, Carcinoma metabolism, Carcinoma virology, Cluster Analysis, Cohort Studies, DNA Methylation, DNA, Viral genetics, Disease Progression, Enhancer of Zeste Homolog 2 Protein metabolism, Female, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Humans, Papillomaviridae, Papillomavirus Infections virology, Polycomb-Group Proteins metabolism, Transcription, Genetic, Uterine Cervical Neoplasms metabolism, Uterine Cervical Neoplasms virology, Uterine Cervical Dysplasia pathology, Uterine Cervical Dysplasia virology, Carcinoma genetics, Enhancer of Zeste Homolog 2 Protein genetics, Epigenesis, Genetic, Uterine Cervical Neoplasms genetics
- Abstract
Human papillomavirus (HPV)-associated cervical carcinoma is preceded by stages of cervical intra-epithelial neoplasia (CIN) that can variably progress to malignancy. Understanding the different molecular processes involved in the progression of pre-malignant CIN is critical to the development of improved predictive and interventional capabilities. We tested the role of regulators of transcription in both the development and the progression of HPV-associated CIN, performing the most comprehensive genomic survey to date of DNA methylation in HPV-associated cervical neoplasia, testing ~2 million loci throughout the human genome in biopsies from 78 HPV+ women, identifying changes starting in early CIN and maintained through carcinogenesis. We identified loci at which DNA methylation is consistently altered, beginning early in the course of neoplastic disease and progressing with disease advancement. While the loss of DNA methylation occurs mostly at intergenic regions, acquisition of DNA methylation is at sites involved in transcriptional regulation, with strong enrichment for targets of polycomb repression. Using an independent cohort from The Cancer Genome Atlas, we validated the loci with increased DNA methylation and found that these regulatory changes were associated with locally decreased gene expression. Secondary validation using immunohistochemistry showed that the progression of neoplasia was associated with increasing polycomb protein expression specifically in the cervical epithelium. We find that perturbations of genomic regulatory processes occur early and persist in cervical carcinoma. The results indicate a polycomb-mediated epigenetic field defect in cervical neoplasia that may represent a target for early, topical interventions using polycomb inhibitors., Competing Interests: The authors declare no potential conflicts of interest.
- Published
- 2016
- Full Text
- View/download PDF
26. EZH2-mediated Puma gene repression regulates non-small cell lung cancer cell proliferation and cisplatin-induced apoptosis.
- Author
-
Liu H, Li W, Yu X, Gao F, Duan Z, Ma X, Tan S, Yuan Y, Liu L, Wang J, Zhou X, and Yang Y
- Subjects
- Animals, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Apoptosis drug effects, Apoptosis Regulatory Proteins metabolism, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Non-Small-Cell Lung mortality, Carcinoma, Non-Small-Cell Lung pathology, Cell Line, Tumor, Cell Proliferation, Cisplatin pharmacology, Cisplatin therapeutic use, Down-Regulation, Enhancer of Zeste Homolog 2 Protein genetics, Epigenetic Repression, Gene Knockdown Techniques, HEK293 Cells, Humans, Lung pathology, Lung Neoplasms drug therapy, Lung Neoplasms mortality, Lung Neoplasms pathology, Male, Mice, Mice, Nude, Middle Aged, Promoter Regions, Genetic, Proto-Oncogene Proteins metabolism, RNA Interference, RNA, Small Interfering metabolism, Survival Rate, Up-Regulation, Xenograft Model Antitumor Assays, Apoptosis genetics, Apoptosis Regulatory Proteins genetics, Carcinoma, Non-Small-Cell Lung genetics, Enhancer of Zeste Homolog 2 Protein metabolism, Gene Expression Regulation, Neoplastic, Lung Neoplasms genetics, Proto-Oncogene Proteins genetics
- Abstract
Polycomb group (PcG) proteins are highly conserved epigenetic effectors that maintain the silenced state of genes. EZH2 is the catalytic core and one of the most important components of the polycomb repressive complex 2 (PRC2). In non-small cell lung cancer (NSCLC) cells and primary lung tumors, we found that PRC2 components, including EZH2, are overexpressed. High levels of EZH2 protein were associated with worse overall survival rate in NSCLC patients. RNA interference mediated attenuation of EZH2 expression blunted the malignant phenotype in this setting, exerting inhibitory effects on cell proliferation, anchorage-independent growth, and tumor development in a xenograft mouse model. Unexpectedly, we discovered that, in the suppression of EZH2, p53 upregulated modulator of apoptosis (PUMA) expression was concomitantly induced. This is achieved through EZH2 directly binds to the Puma promoter thus epigenetic repression of PUMA expression. Furthermore, cisplatin-induced apoptosis of EZH2-knocking down NSCLC cells was elevated as a consequence of increased PUMA expression. Our work reveals a novel epigenetic regulatory mechanism controlling PUMA expression and suggests that EZH2 offers a candidate molecular target for NSCLC therapy and EZH2-regulated PUMA induction would synergistically increase the sensitivity to platinum agents in non-small cell lung cancers., Competing Interests: The authors declare that they have no conflicts of interest.
- Published
- 2016
- Full Text
- View/download PDF
27. GSK3β inactivation promotes the oncogenic functions of EZH2 and enhances methylation of H3K27 in human breast cancers.
- Author
-
Ko HW, Lee HH, Huo L, Xia W, Yang CC, Hsu JL, Li LY, Lai CC, Chan LC, Cheng CC, Labaff AM, Liao HW, Lim SO, Li CW, Wei Y, Nie L, Yamaguchi H, and Hung MC
- Subjects
- Catalysis, Cell Line, Tumor, Cell Movement, Cytosol metabolism, Enhancer of Zeste Homolog 2 Protein genetics, Female, Gene Expression Profiling, Humans, Mutation, Oncogenes, Phosphorylation, Breast Neoplasms metabolism, DNA Methylation, Enhancer of Zeste Homolog 2 Protein metabolism, Gene Expression Regulation, Neoplastic, Glycogen Synthase Kinase 3 beta metabolism, Histones chemistry
- Abstract
During the process of tumorigenesis, inactivation of tumor suppressors is a critical step. EZH2, a histone methyltransferase, promotes cell growth and migration through catalyzing trimethylation of histone H3 at Lys 27 (H3K27me3) and plays an important role in tumorigenesis. Its expression can be controlled by phosphorylation. However, the regulation of EZH2 activity by tumor suppressor kinase is not well understood. In this study, we show that glycogen synthase kinase 3 beta (GSK3β) negatively regulates H3K27 trimethylation. We also validate that GSKβ physically interacts with EZH2, and their interaction occurs in the cytosol. GSK3β phosphorylates EZH2 at Ser363 and Thr367 in vitro, and activating GSK3β upregulates Thr367 phosphorylationin vivo. Cells expressing GSK3β-non-phosphorylatable mutant EZH2 have higher H3K27 trimethylation and enhanced ability of cell migration and anchorage-independent growth. Inactivation of GSK3β as measured by its phosphorylation at Ser9 is positively correlated with higher level of H3K27 trimethylation in tumor tissues from breast cancer patients. Our study indicated that GSK3β phosphorylates EZH2 at Ser363 and Thr367, resulting in reduced H3K27 trimethylation and biological activity of EZH2 in breast cancer., Competing Interests: The authors have no conflicts of interest to declare.
- Published
- 2016
- Full Text
- View/download PDF
28. Disruption of the EZH2/miRNA/β-catenin signaling suppresses aerobic glycolysis in glioma.
- Author
-
Wang Y, Wang M, Wei W, Han D, Chen X, Hu Q, Yu T, Liu N, You Y, and Zhang J
- Subjects
- 3' Untranslated Regions, Aerobiosis, Animals, Cell Line, Tumor, DNA Methylation, Epigenesis, Genetic, Gene Silencing, Glucose metabolism, Glycolysis, Histones metabolism, Humans, Mice, Mice, Nude, Neoplasm Transplantation, Signal Transduction, Brain Neoplasms metabolism, Enhancer of Zeste Homolog 2 Protein metabolism, Gene Expression Regulation, Neoplastic, Glioma metabolism, MicroRNAs metabolism, beta Catenin metabolism
- Abstract
EZH2 is up-regulated in various cancer types, implicating its role in tumorigenesis. Our recent data have shown that repression of EZH2 inhibited glioma growth by inhibition β-catenin signaling. Here, we identified several miRNAs that were repressed by EZH2, which in turn regulate β-catenin expression by its 3'UTR, such as miR-1224-3p, miR-328 and miR-214. Further, EZH2 silenced miR-328 expression by binding to miR-328 promoter and promoting methylation of miR-328 promoter. Finally, miR-328 largely abrogated EZH2 effects on β-catenin expression and glucose metabolism in glioma cells. Taken together, we propose a model for a coordinated EZH2-β-catenin oncoprotein axis, and epigenetic link between histone modification and DNA methylation, mediated by EZH2-scilenced miRNAs., Competing Interests: The authors declare no conflicts of interest.
- Published
- 2016
- Full Text
- View/download PDF
29. Long noncoding RNA DANCR promotes invasion of prostate cancer through epigenetically silencing expression of TIMP2/3.
- Author
-
Jia J, Li F, Tang XS, Xu S, Gao Y, Shi Q, Guo W, Wang X, He D, and Guo P
- Subjects
- Animals, Benzamides, Cell Line, Tumor, Cell Movement, Enhancer of Zeste Homolog 2 Protein metabolism, Gene Silencing, Humans, Male, Mice, Mice, Nude, Neoplasm Invasiveness, Neoplasm Metastasis, Neoplasm Transplantation, Nitriles, Phenylthiohydantoin analogs & derivatives, Phenylthiohydantoin pharmacology, Promoter Regions, Genetic, RNA, Long Noncoding metabolism, Receptors, Androgen metabolism, Epigenesis, Genetic, Prostatic Neoplasms metabolism, RNA, Long Noncoding genetics, Tissue Inhibitor of Metalloproteinase-2 metabolism, Tissue Inhibitor of Metalloproteinase-3 metabolism
- Abstract
LncRNA DANCR suppresses differentiation of epithelial cells, however, its function in prostate cancer development is still unknown. In the present study, we found the expression of DANCR increases in prostate cancer tissues and cells compared to normal prostate tissues and cells, moreover, DANCR promotes invasion and migration of prostate cancer cells in vitro and metastasis of tumor xenografts in nude mice. Mechanistically, we found that TIMP2/3, which are critical metastasis inhibitor of prostate cancer, were down-regulated by DANCR synergistically with EZH2 through epigenetically silencing their promoter by chromatin immunoprecipitation assay. In addition, we further investigated whether DANCR is regulated by the differentiation-promoting androgen-androgen receptor (AR) pathway and found that DANCR expression is repressed by androgen-AR; furthermore, DANCR impedes the upregulation of TIMP2/3 and the suppression of invasion and migration by androgen-AR. On the other hand, interestingly, we found that in prostate cancer cells DANCR knockdown decreased the promotion of invasion and migration by the treatment of enzalutamide, which is an AR inhibitor. In summary, our results indicate that DANCR promotes prostate cancer invasion and metastasis through repressing the expression of TIMP2/3, and suggest that DANCR could be a potential target for preventing prostate cancer metastasis, and knockdown DANCR may lessen the potential side effect of AR inhibitor., Competing Interests: The authors declare no conflicts of interest.
- Published
- 2016
- Full Text
- View/download PDF
30. Post-translational modification-regulated leukocyte adhesion and migration.
- Author
-
Loh JT and Su IH
- Subjects
- Adaptive Immunity physiology, Carcinogenesis pathology, Dendritic Cells physiology, Dermatitis, Allergic Contact physiopathology, Endothelium, Vascular, Humans, Immunity, Innate physiology, Integrins metabolism, Methylation, Signal Transduction physiology, Cell Adhesion physiology, Cell Movement physiology, Enhancer of Zeste Homolog 2 Protein metabolism, Leukocytes physiology, Protein Processing, Post-Translational, Talin metabolism
- Abstract
Leukocytes undergo frequent phenotypic changes and rapidly infiltrate peripheral and lymphoid tissues in order to carry out immune responses. The recruitment of circulating leukocytes into inflamed tissues depends on integrin-mediated tethering and rolling of these cells on the vascular endothelium, followed by transmigration into the tissues. This dynamic process of migration requires the coordination of large numbers of cytosolic and transmembrane proteins whose functional activities are typically regulated by post-translational modifications (PTMs). Our recent studies have shown that the lysine methyltransferase, Ezh2, critically regulates integrin signalling and governs the adhesion dynamics of leukocytes via direct methylation of talin, a key molecule that controls these processes by linking integrins to the actin cytoskeleton. In this review, we will discuss the various modes of leukocyte migration and examine how PTMs of cytoskeletal/adhesion associated proteins play fundamental roles in the dynamic regulation of leukocyte migration. Furthermore, we will discuss molecular details of the adhesion dynamics controlled by Ezh2-mediated talin methylation and the potential implications of this novel regulatory mechanism for leukocyte migration, immune responses, and pathogenic processes, such as allergic contact dermatitis and tumorigenesis., Competing Interests: There is no conflict of interest.
- Published
- 2016
- Full Text
- View/download PDF
31. The histone methyltransferase EZH2 as a novel prosurvival factor in clinically aggressive chronic lymphocytic leukemia.
- Author
-
Papakonstantinou N, Ntoufa S, Chartomatsidou E, Kotta K, Agathangelidis A, Giassafaki L, Karamanli T, Bele P, Moysiadis T, Baliakas P, Sutton LA, Stavroyianni N, Anagnostopoulos A, Makris AM, Ghia P, Rosenquist R, and Stamatopoulos K
- Subjects
- Aged, Cell Survival genetics, Cells, Cultured, Disease Progression, Enhancer of Zeste Homolog 2 Protein metabolism, Female, Histones metabolism, Humans, Immunoglobulin Heavy Chains genetics, Immunoglobulin Variable Region genetics, Kaplan-Meier Estimate, Leukemia, Lymphocytic, Chronic, B-Cell enzymology, Leukemia, Lymphocytic, Chronic, B-Cell pathology, Male, Methylation, Middle Aged, Mutation, Prognosis, Tumor Suppressor Protein p53 genetics, Enhancer of Zeste Homolog 2 Protein genetics, Gene Expression Profiling, Gene Expression Regulation, Leukemic, Leukemia, Lymphocytic, Chronic, B-Cell genetics
- Abstract
The histone methyltransferase EZH2 induces gene repression through trimethylation of histone H3 at lysine 27 (H3K27me3). EZH2 overexpression has been reported in many types of cancer and associated with poor prognosis. Here we investigated the expression and functionality of EZH2 in chronic lymphocytic leukemia (CLL). Aggressive cases with unmutated IGHV genes (U-CLL) displayed significantly higher EZH2 expression compared to indolent CLL cases with mutated IGHV genes (M-CLL); furthermore, in U-CLL EZH2 expression was upregulated with disease progression. Within U-CLL, EZH2high cases harbored significantly fewer (p = 0.033) TP53 gene abnormalities compared to EZH2low cases. EZH2high cases displayed high H3K27me3 levels and increased viability suggesting that EZH2 is functional and likely confers a survival advantage to CLL cells. This argument was further supported by siRNA-mediated downmodulation of EZH2 which resulted in increased apoptosis. Notably, at the intraclonal level, cell proliferation was significantly associated with EZH2 expression. Treatment of primary CLL cells with EZH2 inhibitors induced downregulation of H3K27me3 levels leading to increased cell apoptosis. In conclusion, EZH2 is overexpressed in adverse-prognosis CLL and associated with increased cell survival and proliferation. Pharmacologic inhibition of EZH2 catalytic activity promotes apoptosis, highlighting EZH2 as a novel potential therapeutic target for specific subgroups of patients with CLL., Competing Interests: KS received research support from Glaxo SmithKline and Janssen Pharmaceuticals.
- Published
- 2016
- Full Text
- View/download PDF
32. EZH2-mediated repression of GSK-3β and TP53 promotes Wnt/β-catenin signaling-dependent cell expansion in cervical carcinoma.
- Author
-
Chen Q, Zheng PS, and Yang WT
- Subjects
- Animals, CRISPR-Cas Systems, Cell Line, Tumor, Enhancer of Zeste Homolog 2 Protein metabolism, Female, Gene Expression Regulation, Neoplastic, Glycogen Synthase Kinase 3 metabolism, HeLa Cells, Humans, Mice, Inbred BALB C, Mice, Nude, RNA Interference, Transplantation, Heterologous, Tumor Suppressor Protein p53 metabolism, Uterine Cervical Neoplasms metabolism, Uterine Cervical Neoplasms pathology, beta Catenin metabolism, Cell Proliferation genetics, Enhancer of Zeste Homolog 2 Protein genetics, Glycogen Synthase Kinase 3 genetics, Tumor Suppressor Protein p53 genetics, Uterine Cervical Neoplasms genetics, Wnt Signaling Pathway genetics, beta Catenin genetics
- Abstract
Enhancer of zeste homolog 2 (EZH2), a catalytic core component of the Polycomb repressive complex 2 (PRC2), stimulates the silencing of target genes through histone H3 lysine 27 trimethylation (H3K27me3). Recent findings have indicated EZH2 is involved in the development and progression of various human cancers. However, the exact mechanism of EZH2 in the promotion of cervical cancer is largely unknown. Here, we show that EZH2 expression gradually increases during the progression of cervical cancer. We identified a significant positive correlation between EZH2 expression and cell proliferation in vitro and tumor formation in vivo by the up-regulation or down-regulation of EZH2 using CRISPR-Cas9-mediated gene editing technology and shRNA in HeLa and SiHa cells. Further investigation indicated that EZH2 protein significantly accelerated the cell cycle transition from the G0/G1 to S phase. TOP/FOP-Flash reporter assay revealed that EZH2 significantly activated Wnt/β-catenin signaling and the target genes of Wnt/β-catenin pathway were up-regulated, including β-catenin, cyclin D1, and c-myc. Moreover, dual-luciferase reporter and chromatin immunoprecipitation (ChIP) assays confirmed that EZH2 inhibited the expression of glycogen synthase kinase-3β (GSK-3β) and TP53 through physically interacting with motifs in the promoters of the GSK-3β and TP53 genes. Additionally, blockage of the Wnt/β-catenin pathway resulted in significant inhibition of cell proliferation, and activation of the Wnt/β-catenin pathway resulted in significant enhancement of cell proliferation, as induced by EZH2. Taken together, our data demonstrate that EZH2 promotes cell proliferation and tumor formation in cervical cancer through activating the Wnt/β-catenin pathway by epigenetic silencing via GSK-3β and TP53., Competing Interests: Disclose any potential conflicts of interest.
- Published
- 2016
- Full Text
- View/download PDF
33. Long noncoding RNA MALAT1 promotes malignant development of esophageal squamous cell carcinoma by targeting β-catenin via Ezh2.
- Author
-
Wang W, Zhu Y, Li S, Chen X, Jiang G, Shen Z, Qiao Y, Wang L, Zheng P, and Zhang Y
- Subjects
- Adult, Aged, Animals, Carcinoma, Squamous Cell metabolism, Carcinoma, Squamous Cell mortality, Esophageal Neoplasms metabolism, Esophageal Neoplasms mortality, Esophageal Squamous Cell Carcinoma, Female, Heterografts, Humans, Kaplan-Meier Estimate, Male, Mice, Mice, Nude, Middle Aged, Carcinoma, Squamous Cell pathology, Enhancer of Zeste Homolog 2 Protein metabolism, Esophageal Neoplasms pathology, Gene Expression Regulation, Neoplastic physiology, RNA, Long Noncoding metabolism, beta Catenin metabolism
- Abstract
Evidences have shown that lncRNAs involve in the initiation and progression of various cancers including esophageal squamous cell carcinoma (ESCC). The aberrant expression of lncRNA MALAT1 was investigated in 106 paired ESCC tissues and adjacent non-cancerous tissues by qRT-PCR. Down-regulated MALAT1 and Ezh2 over-expression plasmid were constructed respectively to analyze the expression of β-catenin, Lin28 and Ezh2 genes. We found that the MALAT1 expression level was higher in human ESCC tissues (P=0.0011), which was closely correlated with WHO grade (P=0.0395, P=0.0331), lymph node metastasis (P=0.0213) and prognosis (P=0.0294). Silencing of MALAT1 expression inhibited cell proliferation, migration and tumor sphere formation, while increasing cell apoptosis of esophageal cancer in vitro. Down-regulation of MALAT1 decreased the expression of β-catenin, Lin28 and Ezh2 genes, while over-expressed Ezh2 combined with MALAT1 down-regulation completely reversed the si-MALAT1-mediated repression of β-catenin and Lin28 in esophageal cancer cells. Animal experiments showed that knockdown of MALAT1 decreased tumor formation and improved survival. MALAT1 promotes the initiation and progression of ESCC, suggesting that inhibition of MALAT1 might be a potential target for treatment of ESCC., Competing Interests: The authors have declared that no conflict of interest exists.
- Published
- 2016
- Full Text
- View/download PDF
34. MALAT1 long ncRNA promotes gastric cancer metastasis by suppressing PCDH10.
- Author
-
Qi Y, Ooi HS, Wu J, Chen J, Zhang X, Tan S, Yu Q, Li YY, Kang Y, Li H, Xiong Z, Zhu T, Liu B, Shao Z, and Zhao X
- Subjects
- Cell Line, Tumor, Gene Expression Regulation, Neoplastic physiology, Humans, Neoplasm Invasiveness pathology, Protocadherins, Stomach Neoplasms genetics, Cadherins metabolism, Enhancer of Zeste Homolog 2 Protein metabolism, Neoplasm Invasiveness genetics, RNA, Long Noncoding metabolism, Stomach Neoplasms pathology
- Abstract
EZH2, the catalytic component of polycomb repressive complex 2 (PRC2), is frequently overexpressed in human cancers and contributes to tumor initiation and progression, in part through transcriptional silencing of tumor suppressor genes. A number of noncoding RNAs (ncRNAs) recruit EZH2 to specific chromatin loci, where they modulate gene expression. Here, we used RNA immunoprecipitation sequencing (RIP-seq) to profile EZH2-associated transcripts in human gastric cancer cell lines. We identified 8,256 transcripts, including both noncoding and coding transcripts, some of which were derived from cancer-related loci. In particular, we found that long noncoding RNA (lncRNA) MALAT1 binds EZH2, suppresses the tumor suppressor PCDH10, and promotes gastric cellular migration and invasion. Our work thus provides a global view of the EZH2-associated transcriptome and offers new insight into the function of EZH2 in gastric tumorigenesis.
- Published
- 2016
- Full Text
- View/download PDF
35. The relationship between EZH2 expression and microRNA-31 in colorectal cancer and the role in evolution of the serrated pathway.
- Author
-
Kurihara H, Maruyama R, Ishiguro K, Kanno S, Yamamoto I, Ishigami K, Mitsuhashi K, Igarashi H, Ito M, Tanuma T, Sukawa Y, Okita K, Hasegawa T, Imai K, Yamamoto H, Shinomura Y, and Nosho K
- Subjects
- Adenocarcinoma genetics, Adenocarcinoma metabolism, Adenocarcinoma pathology, Adenomatous Polyps genetics, Adenomatous Polyps metabolism, Adenomatous Polyps pathology, Adult, Aged, Colonic Polyps genetics, Colonic Polyps metabolism, Colonic Polyps pathology, Colorectal Neoplasms genetics, Colorectal Neoplasms metabolism, Female, Gene Expression Regulation, Neoplastic physiology, Humans, Kaplan-Meier Estimate, Male, MicroRNAs genetics, Middle Aged, Precancerous Conditions genetics, Precancerous Conditions metabolism, Colorectal Neoplasms pathology, Enhancer of Zeste Homolog 2 Protein metabolism, MicroRNAs metabolism, Precancerous Conditions pathology
- Abstract
Polycomb group protein enhancer of zeste homolog 2 (EZH2) is a methyltransferase that correlates with the regulation of invasion and metastasis and is overexpressed in human cancers such as colorectal cancer. MicroRNA-31 (miR-31) plays an oncogenic role and is associated with BRAF mutation and poor prognosis in colorectal cancer. EZH2 is functionally considered to suppress miR-31 expression in human cancers; however, no study has reported its relationship with colon cancer. We therefore evaluated EZH2 expression using immunohistochemistry and assessed miR-31 and epigenetic alterations using 301 colorectal carcinomas and 207 premalignant lesions. Functional analysis was performed to identify the association between EZH2 and miR-31 using cancer cell lines. In the current study, negative, weak, moderate, and strong EZH2 expressions were observed in 15%, 19%, 25%, and 41% of colorectal cancers, respectively. EZH2 was inversely associated with miR-31 (P < 0.0001), independent of clinicopathological and molecular features. In a multivariate stage-stratified analysis, high EZH2 expression was related to favorable prognosis (P = 0.0022). Regarding premalignant lesions, negative EZH2 expression was frequently detected in sessile serrated adenomas/polyps (SSA/Ps) (76%; P < 0.0001) compared with hyperplastic polyps, traditional serrated adenomas, and non-serrated adenomas (25-36%). Functional analysis demonstrated that the knockdown of EZH2 increased miR-31 expression. In conclusion, an inverse association was identified between EZH2 and miR-31 in colorectal cancers. Our data also showed that upregulation of EZH2 expression may be rare in SSA/Ps. These results suggest that EZH2 suppresses miR-31 in colorectal cancer and may correlate with differentiation and evolution of serrated pathway.
- Published
- 2016
- Full Text
- View/download PDF
36. Long non-coding RNA LINC01133 represses KLF2, P21 and E-cadherin transcription through binding with EZH2, LSD1 in non small cell lung cancer.
- Author
-
Zang C, Nie FQ, Wang Q, Sun M, Li W, He J, Zhang M, and Lu KH
- Subjects
- A549 Cells, Aged, Animals, Antigens, CD, Cadherins biosynthesis, Carcinoma, Non-Small-Cell Lung metabolism, Carcinoma, Non-Small-Cell Lung pathology, Cell Line, Tumor, Cell Proliferation physiology, Cyclin-Dependent Kinase Inhibitor p21 biosynthesis, Heterografts, Humans, Kruppel-Like Transcription Factors biosynthesis, Lung Neoplasms metabolism, Lung Neoplasms pathology, Mice, Mice, Inbred BALB C, Mice, Nude, Prognosis, Transcription, Genetic, Transfection, Cadherins genetics, Carcinoma, Non-Small-Cell Lung genetics, Cyclin-Dependent Kinase Inhibitor p21 genetics, Enhancer of Zeste Homolog 2 Protein metabolism, Histone Demethylases metabolism, Kruppel-Like Transcription Factors genetics, Lung Neoplasms genetics, RNA, Long Noncoding genetics
- Abstract
Long non-coding RNAs are emerging as crucial regulators and prognostic markers in multiple cancers including non small cell lung cancer (NSCLC). In this study, we screened LINCO1133 as a new candidate lncRNA which promotes NSCLC development and progression, in two independent datasets (GSE18842 and GSE19804) from the Gene Expression Omnibus (GEO). LINC01133 is previously found to be over-expressed in lung squamous cell cancer (LSCC) and knockdown its expression inhibits LSCC cells invasion. However, its' molecular mechanism and downstream targets involving in regulation of cancer cells phenotype is not known. Here, we found that LINC01133 expression is up-regulated in NSCLC tissues, and its' over-expression is associated with patients poor prognosis and short survival time. LINC01133 knockdown decreased NSCLC cells proliferation, migration, invasion and induced cell cycle G1/S phase arrest and cell apoptosis. Mechanistic investigations showed that LINC01133 could interact with EZH2, LSD1 and recruit them to KLF2, P21 or E-cadherin promoter regions to repress their transcription. Furthermore, rescue experiments demonstrated that LINC01133 oncogenic function is partly through regulating KLF2. Lastly, we found that there was negative correlation between LINC01133 and KLF2, P21 or E-cadherin in NSCLC. Overall, our findings illuminate how LINC01133 over-expression confers an oncogenic function in NSCLC that may offer a novel therapy target in this disease.
- Published
- 2016
- Full Text
- View/download PDF
37. EZH2 promotes cell migration and invasion but not alters cell proliferation by suppressing E-cadherin, partly through association with MALAT-1 in pancreatic cancer.
- Author
-
Han T, Jiao F, Hu H, Yuan C, Wang L, Jin ZL, Song WF, and Wang LW
- Subjects
- Adult, Aged, Antigens, CD, Cell Proliferation, Female, Humans, Kaplan-Meier Estimate, Male, Middle Aged, Neoplasm Invasiveness pathology, Pancreatic Neoplasms metabolism, Pancreatic Neoplasms mortality, Cadherins metabolism, Cell Movement, Enhancer of Zeste Homolog 2 Protein metabolism, Gene Expression Regulation, Neoplastic physiology, Pancreatic Neoplasms pathology, RNA, Long Noncoding metabolism
- Abstract
Enhancer of zeste homolog 2 (EZH2) is an essential component of the polycomb repressive complex 2 (PRC2), which is required for epigenetic silencing of target genes, including those affecting cancer progression. Its role in pancreatic cancer remains to be clarified; therefore, we investigated the effects of aberrantly expressed EZH2 on pancreatic cancer. We found that EZH2 expression is up-regulated in pancreatic cancer tissues and positively correlated with lymph node metastasis and advanced clinical stage in pancreatic cancer patients. EZH2 knockdown in pancreatic cancer cell lines inhibited cell migration and invasion, but did not alter cell proliferation. Silencing of EZH2 also increased E-cadherin expression in vitro, and E-cadherin expression was inversely correlated with EZH2 expression in pancreatic cancer tissue samples. Patients with high EZH2 and low E-cadherin expression had the worst prognosis. RIP and ChIP assays suggest that EZH2 is recruited to the E-cadherin promoter by the long non-coding RNA, MALAT-1 (metastasis associated in lung adenocarcinoma transcript 1), where it represses E-cadherin expression. Our results show that EZH2-based therapies may be an option for the treatment of pancreatic cancer.
- Published
- 2016
- Full Text
- View/download PDF
38. Long intergenic non-coding RNA 00152 promotes tumor cell cycle progression by binding to EZH2 and repressing p15 and p21 in gastric cancer.
- Author
-
Chen WM, Huang MD, Sun DP, Kong R, Xu TP, Xia R, Zhang EB, and Shu YQ
- Subjects
- Cell Line, Tumor, Cell Proliferation genetics, Cyclin-Dependent Kinase Inhibitor p15 genetics, Cyclin-Dependent Kinase Inhibitor p21 genetics, Female, Humans, Lymphatic Metastasis genetics, Male, Middle Aged, Neoplasm Staging, RNA, Long Noncoding genetics, RNA-Binding Proteins metabolism, Stomach Neoplasms genetics, Stomach Neoplasms mortality, Cell Cycle genetics, Cyclin-Dependent Kinase Inhibitor p15 antagonists & inhibitors, Cyclin-Dependent Kinase Inhibitor p21 antagonists & inhibitors, Enhancer of Zeste Homolog 2 Protein metabolism, RNA, Long Noncoding metabolism, Stomach Neoplasms pathology
- Abstract
Long noncoding RNAs (lncRNAs) play important regulatory roles in several human cancers. Integrated analysis revealed that expression of long intergenic non-coding RNA 152 (LINC00152) was significantly upregulated in gastric cancer (GC). Further analysis in a cohort of 97 GC patients revealed that LINC00152 expression was positively correlated with tumor invasion depth, lymph node metastasis, higher TNM stage, and poor survival. Gene set enrichment analysis revealed that cell proliferation and cell cycle progression were increased in patients with high LINC00152 expression. In both GC cell lines and xenograft systems, LINC00152 overexpression facilitated GC cell proliferation by accelerating the cell cycle, whereas LINC00152 knockdown had the opposite effect. Moreover, by binding to enhancer of zeste homolog 2 (EZH2), LINC00152 promotes GC tumor cell cycle progression by silencing the expression of p15 and p21. These findings suggest that LINC00152 may play contribute to the progression of GC and may be an effective therapeutic target.
- Published
- 2016
- Full Text
- View/download PDF
39. Genome-wide profiling of histone H3 lysine 27 and lysine 4 trimethylation in multiple myeloma reveals the importance of Polycomb gene targeting and highlights EZH2 as a potential therapeutic target.
- Author
-
Agarwal P, Alzrigat M, Párraga AA, Enroth S, Singh U, Ungerstedt J, Österborg A, Brown PJ, Ma A, Jin J, Nilsson K, Öberg F, Kalushkova A, and Jernberg-Wiklund H
- Subjects
- Chromatin metabolism, Down-Regulation, Enhancer of Zeste Homolog 2 Protein genetics, Enhancer of Zeste Homolog 2 Protein metabolism, Gene Expression Profiling, Histones genetics, Humans, Lysine metabolism, Methylation, Molecular Targeted Therapy, Multiple Myeloma metabolism, Polycomb-Group Proteins metabolism, Enhancer of Zeste Homolog 2 Protein antagonists & inhibitors, Enzyme Inhibitors pharmacology, Histones metabolism, Multiple Myeloma drug therapy, Multiple Myeloma genetics, Polycomb-Group Proteins genetics
- Abstract
Multiple myeloma (MM) is a malignancy of the antibody-producing plasma cells. MM is a highly heterogeneous disease, which has hampered the identification of a common underlying mechanism for disease establishment as well as the development of targeted therapy. Here we present the first genome-wide profiling of histone H3 lysine 27 and lysine 4 trimethylation in MM patient samples, defining a common set of active H3K4me3-enriched genes and silent genes marked by H3K27me3 (H3K27me3 alone or bivalent) unique to primary MM cells, when compared to normal bone marrow plasma cells. Using this epigenome profile, we found increased silencing of H3K27me3 targets in MM patients at advanced stages of the disease, and the expression pattern of H3K27me3-marked genes correlated with poor patient survival. We also demonstrated that pharmacological inhibition of EZH2 had anti-myeloma effects in both MM cell lines and CD138+ MM patient cells. In addition, EZH2 inhibition decreased the global H3K27 methylation and induced apoptosis. Taken together, these data suggest an important role for the Polycomb repressive complex 2 (PRC2) in MM, and highlights the PRC2 component EZH2 as a potential therapeutic target in MM.
- Published
- 2016
- Full Text
- View/download PDF
40. Inhibition of EZH2 by chemo- and radiotherapy agents and small molecule inhibitors induces cell death in castration-resistant prostate cancer.
- Author
-
Wu C, Jin X, Yang J, Yang Y, He Y, Ding L, Pan Y, Chen S, Jiang J, and Huang H
- Subjects
- Animals, Blotting, Western, Cell Movement drug effects, Cell Movement radiation effects, Cell Proliferation drug effects, Cell Proliferation radiation effects, Enhancer of Zeste Homolog 2 Protein genetics, Enhancer of Zeste Homolog 2 Protein metabolism, Humans, Immunoenzyme Techniques, Male, Mice, Mice, Inbred NOD, Mice, SCID, Prostatic Neoplasms metabolism, Prostatic Neoplasms therapy, RNA, Messenger genetics, RNA, Small Interfering genetics, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Apoptosis drug effects, Apoptosis radiation effects, Chemoradiotherapy, Enhancer of Zeste Homolog 2 Protein antagonists & inhibitors, Indoles pharmacology, Prostatic Neoplasms pathology, Pyridones pharmacology
- Abstract
Androgen deprivation therapy is the mainstay of treatment of advanced prostate cancer (PCa). However, a significant portion of patients experience disease relapse and tumors ultimately evolve into castration resistant prostate cancer (CRPC), for which there is no cure in the clinic. The Polycomb protein enhancer of zeste homolog 2 (EZH2) is frequently overexpressed in CRPC. It is unclear whether EZH2 can be a therapeutic target in CRPC. Here, we demonstrated that chemo- and radiotherapy agents such as camptothecin (CPT) and γ irradiation decrease EZH2 expression in various PCa cell lines. We provided evidence that functional p53 and RB proteins are required for CPT- and irradiation-induced downregulation of EZH2 in CRPC cells. We demonstrated that EZH2-specific small molecule inhibitors mitigate CRPC cell growth. We further showed that the EZH2 inhibitor GSK126 inhibits both Polycomb-dependent and -independent functions of EZH2 in PCa cells. Importantly, we found that inhibition of EZH2 by genetic and pharmacological means sensitizes CRPC cells to CPT-induced apoptotic death and growth inhibition in culture and in mice. Our data suggest that concomitant administration of small molecule inhibitors of EZH2 may significantly increase the anti-tumor efficacy of conventional chemo- and radiotherapies in CRPC.
- Published
- 2016
- Full Text
- View/download PDF
41. EZH2 in normal hematopoiesis and hematological malignancies.
- Author
-
Herviou L, Cavalli G, Cartron G, Klein B, and Moreaux J
- Subjects
- Adenosine analogs & derivatives, Adenosine pharmacology, B-Lymphocytes cytology, Cell Differentiation, Cell Proliferation, Epigenesis, Genetic, Ethylenediamines pharmacology, Gene Expression Regulation, Hematologic Neoplasms drug therapy, Histones metabolism, Humans, Indoles pharmacology, Methylation, Precision Medicine, Pyrazoles pharmacology, Pyridones pharmacology, Enhancer of Zeste Homolog 2 Protein antagonists & inhibitors, Enhancer of Zeste Homolog 2 Protein metabolism, Hematologic Neoplasms pathology, Hematopoiesis physiology, Hematopoietic Stem Cells cytology, Polycomb Repressive Complex 2 metabolism
- Abstract
Enhancer of zeste homolog 2 (EZH2), the catalytic subunit of the Polycomb repressive complex 2, inhibits gene expression through methylation on lysine 27 of histone H3. EZH2 regulates normal hematopoietic stem cell self-renewal and differentiation. EZH2 also controls normal B cell differentiation. EZH2 deregulation has been described in many cancer types including hematological malignancies. Specific small molecules have been recently developed to exploit the oncogenic addiction of tumor cells to EZH2. Their therapeutic potential is currently under evaluation. This review summarizes the roles of EZH2 in normal and pathologic hematological processes and recent advances in the development of EZH2 inhibitors for the personalized treatment of patients with hematological malignancies.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.