1
|
Liu J, Fan H, Liang X, Chen Y. Polycomb repressor complex: Its function in human cancer and therapeutic target strategy. Biomed Pharmacother 2023; 169:115897. [PMID: 37981459 DOI: 10.1016/j.biopha.2023.115897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/07/2023] [Accepted: 11/13/2023] [Indexed: 11/21/2023] Open
Abstract
The Polycomb Repressor Complex (PRC) plays a pivotal role in gene regulation during development and disease, with dysregulation contributing significantly to various human cancers. The intricate interplay between PRC and cellular signaling pathways sheds light on cancer complexity. PRC presents promising therapeutic opportunities, with inhibitors undergoing rigorous evaluation in preclinical and clinical studies. In this review, we emphasize the critical role of PRC complex in gene regulation, particularly PcG proteins mediated chromatin compaction through phase separation. We also highlight the pathological implications of PRC complex dysregulation in various tumors, elucidating underlying mechanisms driving cancer progression. The burgeoning field of therapeutic strategies targeting PRC complexes, notably EZH2 inhibitors, has advanced significantly. However, we explore the need for combination therapies to enhance PRC targeted treatments efficacy, providing a glimpse into the future of cancer therapeutics.
Collapse
Affiliation(s)
- Jingrong Liu
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China
| | - Hongjie Fan
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China
| | - Xinmiao Liang
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China; CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.
| | - Yang Chen
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China; CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.
| |
Collapse
|
2
|
Hou C, Xiao L, Ren X, Cheng L, Guo B, Zhang M, Yan N. EZH2-mediated H3K27me3 is a predictive biomarker and therapeutic target in uveal melanoma. Front Genet 2022; 13:1013475. [PMID: 36276954 PMCID: PMC9582331 DOI: 10.3389/fgene.2022.1013475] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 09/20/2022] [Indexed: 11/17/2022] Open
Abstract
Although gene mutations and aberrant chromosomes are associated with the pathogenesis and prognosis of uveal melanoma (UM), potential therapeutic targets still need to be explored. We aim to determine the predictive value and potential therapeutic target of EZH2 in uveal melanoma. Eighty-five uveal melanoma samples were recruited in our study, including 19 metastatic and 66 nonmetastatic samples. qRT-PCR, immunohistochemistry staining, and western blotting were applied to detect the expression of EZH2 and H3K27me3. We found that EZH2 (41/85, 48.24%) and H3K27me3 (49/85, 57.65%) were overexpressed in uveal melanoma. The expression of EZH2 was not significantly associated with metastasis. High H3K27me3 expression was correlated with poor patient prognosis. UNC 1999, an EZH2 inhibitor, can downregulate H3K27me3 expression and has the most potency to inhibit OMM1 cell growth by the cell cycle and ferroptosis pathway. These results indicate that H3K27me3 can be a biomarker predicting a poor prognosis of UM. EZH2 is the potential therapeutic target for UM.
Collapse
Affiliation(s)
- Chen Hou
- Research Laboratory of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Lirong Xiao
- Research Laboratory of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiang Ren
- Research Laboratory of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Lin Cheng
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, United States
| | - Bo Guo
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Meixia Zhang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Naihong Yan
- Research Laboratory of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Naihong Yan,
| |
Collapse
|
3
|
Saigusa N, Hirai H, Tada Y, Kawakita D, Nakaguro M, Tsukahara K, Kano S, Ozawa H, Kondo T, Okami K, Togashi T, Sato Y, Urano M, Kajiwara M, Shimura T, Fushimi C, Shimizu A, Okamoto I, Okada T, Suzuki T, Imanishi Y, Watanabe Y, Sakai A, Ebisumoto K, Sato Y, Honma Y, Yamazaki K, Ueki Y, Hanazawa T, Saito Y, Takahashi H, Ando M, Kohsaka S, Matsuki T, Nagao T. The Role of the EZH2 and H3K27me3 Expression as a Predictor of Clinical Outcomes in Salivary Duct Carcinoma Patients: A Large-Series Study With Emphasis on the Relevance to the Combined Androgen Blockade and HER2-Targeted Therapy. Front Oncol 2022; 11:779882. [PMID: 35186711 PMCID: PMC8850643 DOI: 10.3389/fonc.2021.779882] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 12/27/2021] [Indexed: 12/15/2022] Open
Abstract
ObjectiveSalivary duct carcinoma (SDC) is a highly aggressive and uncommon tumor arising not only de novo but also in pleomorphic adenoma. Androgen receptor (AR)- and HER2-targeted therapy have recently been introduced for SDC as promising treatment options; however, no predictive biomarkers have yet been established. EZH2 and H3K27me3 are closely linked to the development and progression of various cancers, and EZH2 is also expected to be a desirable therapeutic target. We therefore explored the clinicopathological and prognostic implications of EZH2 and H3K27me3 in a large cohort of SDC patients, focusing on their impact on the therapeutic efficacy of AR- or HER2-targeted therapy.Materials and MethodsThe EZH2 and H3K27me3 immunohistochemical expression and EZH2 Y646 gain-of-function mutation status were examined in 226 SDCs, and the relationship with the clinicopathological factors as well as clinical outcomes were evaluated within the three groups depending on the treatment: AR-targeted (combined androgen blockade with leuprorelin acetate and bicalutamide; 89 cases), HER2-targeted (trastuzumab and docetaxel; 42 cases), and conventional therapy (112 cases).ResultsEZH2 and H3K27me3 were variably immunoreactive in most SDCs. A positive correlation was found between the expression of EZH2 and H3K27me3. The EZH2 expression in the SDC component was significantly higher than that in the pre-existing pleomorphic adenoma component. EZH2 Y646 was not identified in any cases. EZH2-high cases more frequently had an advanced clinical stage and aggressive histological features than EZH2-low cases. An EZH2-high status in patients treated with AR-targeted therapy was associated with a significantly shorter progression-free and overall survival as well as a lower objective response rate and clinical benefit rate. In addition, a H3K27me3-high status in patients treated with AR-targeted therapy was related to a shorter overall survival. Conversely, there was no association between the EZH2 and H3K27me3 expression and the clinical outcomes in the conventional or HER2-targeted therapy groups.ConclusionsA high expression of EZH2 and H3K27me3 in SDC might be a predictor of a poor efficacy of AR-targeted therapy. Our data provide new insights into the role of EZH2 and H3K27me3 in therapeutic strategies for SDC.
Collapse
Affiliation(s)
- Natsuki Saigusa
- Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan
| | - Hideaki Hirai
- Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan
| | - Yuichiro Tada
- Department of Head and Neck Oncology and Surgery, International University of Health and Welfare, Mita Hospital, Tokyo, Japan
| | - Daisuke Kawakita
- Department of Otorhinolaryngology, Head and Neck Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Masato Nakaguro
- Department of Pathology and Laboratory Medicine, Nagoya University Hospital, Nagoya, Japan
| | - Kiyoaki Tsukahara
- Department of Otorhinolaryngology, Head and Neck Surgery, Tokyo Medical University, Tokyo, Japan
| | - Satoshi Kano
- Department of Otolaryngology Head and Neck Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroyuki Ozawa
- Department of Otorhinolaryngology Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Takahito Kondo
- Department of Otorhinolaryngology, Head and Neck Surgery, Tokyo Medical University Hachioji Medical Center, Tokyo, Japan
| | - Kenji Okami
- Department of Otolaryngology Head and Neck Surgery, Tokai University School of Medicine, Isehara, Japan
| | - Takafumi Togashi
- Department of Head and Neck Surgery, Niigata Cancer Center Hospital, Niigata, Japan
| | - Yukiko Sato
- Department of Pathology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Makoto Urano
- Department of Diagnostic Pathology Bantane Hospital Fujita Health University, School of Medicine, Nagoya, Japan
| | - Manami Kajiwara
- Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan
| | - Tomotaka Shimura
- Department of Otolaryngology, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Chihiro Fushimi
- Department of Head and Neck Oncology and Surgery, International University of Health and Welfare, Mita Hospital, Tokyo, Japan
| | - Akira Shimizu
- Department of Otorhinolaryngology, Head and Neck Surgery, Tokyo Medical University, Tokyo, Japan
| | - Isaku Okamoto
- Department of Otorhinolaryngology, Head and Neck Surgery, Tokyo Medical University, Tokyo, Japan
| | - Takuro Okada
- Department of Otorhinolaryngology, Head and Neck Surgery, Tokyo Medical University, Tokyo, Japan
| | - Takayoshi Suzuki
- Department of Otolaryngology Head and Neck Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yorihisa Imanishi
- Department of Otorhinolaryngology Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Yoshihiro Watanabe
- Department of Otorhinolaryngology Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Akihiro Sakai
- Department of Otolaryngology Head and Neck Surgery, Tokai University School of Medicine, Isehara, Japan
| | - Koji Ebisumoto
- Department of Otolaryngology Head and Neck Surgery, Tokai University School of Medicine, Isehara, Japan
| | - Yuichiro Sato
- Department of Head and Neck Surgery, Niigata Cancer Center Hospital, Niigata, Japan
| | - Yoshitaka Honma
- Department of Head and Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Keisuke Yamazaki
- Department of Otolaryngology Head and Neck Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yushi Ueki
- Department of Otolaryngology Head and Neck Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Toyoyuki Hanazawa
- Department of Otolaryngology, Head and Neck Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Yuki Saito
- Department of Otolaryngology - Head and Neck Surgery, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hideaki Takahashi
- Department of Otorhinolaryngology, Head and Neck Surgery, Yokohama City University, School of Medicine, Yokohama, Japan
| | - Mizuo Ando
- Department of Otolaryngology-Head & Neck Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shinji Kohsaka
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Takashi Matsuki
- Department of Otorhinolaryngology, Head and Neck Surgery, Kitasato University School of Medicine, Kanagawa, Japan
| | - Toshitaka Nagao
- Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan
- *Correspondence: Toshitaka Nagao,
| |
Collapse
|
4
|
Li N, Wang Z. Integrative Analysis of Deregulated miRNAs Reveals Candidate Molecular Mechanisms Linking H. pylori Infected Peptic Ulcer Disease with Periodontitis. DISEASE MARKERS 2022; 2022:1498525. [PMID: 35132337 PMCID: PMC8817886 DOI: 10.1155/2022/1498525] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 01/17/2022] [Indexed: 11/18/2022]
Abstract
OBJECTIVE Periodontitis is a highly prevalent oral infectious disease and has been increasingly associated with H. pylori infection, gastric inflammation, and gastric cancer but little is known about epigenetic machinery underlying this potentially bidirectional association. The present study is aimed at identifying key deregulated miRNA, their associated genes, signaling pathways, and compounds linking periodontitis with H. pylori-associated peptic ulcer disease. METHODS miRNA expression datasets for periodontitis-affected and H. pylori-associated peptic ulcer disease-affected tissues were sought from the GEO database. Differentially expressed miRNA (DEmiRNAs) were identified and the overlapping, shared-DEmiRNA between both datasets were determined. Shared-DEmiRNA-target networks construction and functional analyses were constructed using miRNet 2.0, including shared-DEmiRNA-gene, shared-DEmiRNA-transcription factor (TF), and shared-DEmiRNA-compound networks. Functional enrichment analysis for shared DEmiRNA-gene and shared DEmiRNA-TF networks was performed using the KEGG, Reactome, and Geno Ontology (GO) pathways. RESULTS 11 shared-DEmiRNAs were identified, among which 9 showed similar expression patterns in both diseases, and 7 were overexpressed. miRNA hsa-hsa-mir-155-5p and hsa-mir-29a-3p were top miRNA nodes in both gene and TF networks. The topmost candidate miRNA-deregulated genes were PTEN, CCND1, MDM2, TNRC6A, and SCD while topmost deregulated TFs included STAT3, HIF1A, EZH2, CEBPA, and RUNX1. Curcumin, 5-fluorouracil, and the gallotanin 1,2,6-Tri-O-galloyl-beta-D-glucopyranose emerged as the most relevant linkage compound targets. Functional analyses revealed multiple cancer-associated pathways, PI3K pathways, kinase binding, and transcription factor binding among as enriched by the network-associated genes and TFs. CONCLUSION Integrative analysis of deregulated miRNAs revealed candidate molecular mechanisms comprising of top miRNA, their gene, and TF targets linking H. pylori-infected peptic ulcer disease with periodontitis and highlighted compounds targeting both diseases. These findings provide basis for directing future experimental research.
Collapse
Affiliation(s)
- Ning Li
- Department of Prosthetic Dentistry, The Affiliated Stomatological Hospital of Wenzhou Medical University, Longyao Avenue No. 1288, Yongzhong Street, Longwan District, Wenzhou 325000, Zhejiang Province, China
| | - Zhen Wang
- Department of Stomatology, The Quzhou Affiliated Hospital of Wenzhou Medical University (Quzhou People's Hospital), Kecheng District, Minjiang Avenue No. 100, Quzhou 332400, Zhejiang Province, China
| |
Collapse
|
5
|
Purkait S, Patra S, Mitra S, Behera MM, Panigrahi MK, Kumar P, Kar M, Hallur V, Chandra Samal S. Elevated Expression of DNA Methyltransferases and Enhancer of Zeste Homolog 2 in Helicobacter pylori - Gastritis and Gastric Carcinoma. Dig Dis 2021; 40:156-167. [PMID: 33895728 DOI: 10.1159/000516478] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 04/08/2021] [Indexed: 02/02/2023]
Abstract
AIM The aim of this study was to study the role of key epigenetic regulators pertaining to DNA methylation and histone-modification systems in Helicobacter pylori (HP)-associated gastritis and gastric carcinogenesis. METHODS The expression of DNA methyltransferase (DNMT-1, 3A, and 3B) and the catalytic subunit of polycomb repressive complex-2 (enhancer of zeste homolog 2 [EZH2]) in gastric carcinomas (n = 104), mucosa adjacent to carcinoma (n = 104), HP-associated gastritis (n = 95), and histologically normal mucosa (n = 31) was assessed by immunohistochemistry and qRT-PCR. RESULTS The expression of all 3 DNMTs and EZH2 was significantly higher in HP-associated gastritis and carcinoma cases than in those with adjacent and normal mucosa. The expression of DNMT-1 and 3B was maximum in HP-associated gastritis. DNMT-3A showed higher expression in carcinoma-adjacent mucosa than in normal mucosa. Interestingly, the expression of EZH2 was higher in cases of HP-associated gastritis with metaplasia than in those without metaplasia and also in cases of intestinal type of adenocarcinoma. Significant positive correlation of EZH2 was identified with DNMT-1, DNMT-3A, and DNMT-3B. However, none of these markers was associated with survival outcome. CONCLUSION This study establishes an important role of the key epigenetic regulators in the pathogenesis of both HP-associated gastritis and gastric carcinoma. Higher expression of all the epigenetic markers in the gastritis and their persistence in the carcinoma point toward their implications in HP-driven gastric carcinogenesis. Further, an inter-relation between the 2 arms of epigenetics, namely, DNA methylation and histone-modification in the pathogenesis of gastric carcinoma, is also documented. Given the reversibility of epigenetic phenomenon, these molecules may be of important therapeutic use.
Collapse
Affiliation(s)
- Suvendu Purkait
- Department of Pathology and Lab Medicine, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Susama Patra
- Department of Pathology and Lab Medicine, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Suvradeep Mitra
- Department of Pathology and Lab Medicine, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Minakshi M Behera
- Department of Pathology and Lab Medicine, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Manas Kumar Panigrahi
- Department of Gastroenterology, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Pankaj Kumar
- Department of General Surgery, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Madhabananda Kar
- Department of Surgical Oncology, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Vinaykumar Hallur
- Department of Microbiology, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Subash Chandra Samal
- Department of Gastroenterology, All India Institute of Medical Sciences, Bhubaneswar, India
| |
Collapse
|
6
|
Xiong J, Tu Y, Feng Z, Li D, Yang Z, Huang Q, Li Z, Cao Y, Jie Z. Epigenetics mechanisms mediate the miR-125a/BRMS1 axis to regulate invasion and metastasis in gastric cancer. Onco Targets Ther 2019; 12:7513-7525. [PMID: 31571904 PMCID: PMC6753057 DOI: 10.2147/ott.s210376] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 08/17/2019] [Indexed: 12/24/2022] Open
Abstract
Purpose Altered expression of breast cancer metastasis suppressor 1 (BRMS1), is a tumor suppressor, which is found in many types of cancers, including gastric cancer (GC), but the mechanism by which BRMS1 inhibits invasion and metastasis in GC is unknown. The aim of the study was to investigate the molecular mechanisms of miR-125a/BRMS1 in GC. Materials and methods The expression of BRMS1 and miR-125a were detected by quantitative real-time PCR (qRT-PCR) and analyzed by bioinformatics. BSP and MSP were used to detecte the methylation status of miR-125a and BRMS1 which was treated by 5-Aza or not. Western Blot and qRT-PCR were used to analyze the expression of BRMS1 and EZH2. Transwell was performed to explore the invasion and metastasis ability of GC cells. The nude mice were used for the tumor formation assay. Results BRMS1 may be regulated by copy number variation (CNV), methylation and miR-125a-5p. As one of the essential components of PRC2, EZH2 is an important regulatory factor resulting in the low expression of miR-125a. An epigenetic mechanism mediates the miR-125a/BRMS1 axis to inhibit the invasion and metastasis of GC cells. In vivo experiments, it is also showed that BRMS1 is involved in invasion and metastasis but not the proliferation in GC. Conclusion These studies shed light on the mechanism of BRMS1 inhibition of GC invasion and metastasis and the development of new drugs targeting the miR-125a/BRMS1 axis, which will be a promising therapeutic strategy for GC and other human cancers.
Collapse
Affiliation(s)
- Jianbo Xiong
- Department of Gastrointestinal Surgery, First Affiliated Hospital, Nanchang University, Nanchang 330006, Jiangxi Province, People's Republic of China
| | - Yi Tu
- Department of Pathology, First Affiliated Hospital, Nanchang University, Nanchang 330006, Jiangxi Province, People's Republic of China
| | - Zongfeng Feng
- Department of Gastrointestinal Surgery, First Affiliated Hospital, Nanchang University, Nanchang 330006, Jiangxi Province, People's Republic of China
| | - Daojiang Li
- Department of General Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, People's Republic of China
| | - Zhouwen Yang
- Department of Gastrointestinal Surgery, First Affiliated Hospital, Nanchang University, Nanchang 330006, Jiangxi Province, People's Republic of China
| | - Qiuxia Huang
- Department of Nursing, First Affiliated Hospital, Nanchang University, Nanchang 330006, Jiangxi Province, People's Republic of China
| | - Zhengrong Li
- Department of Gastrointestinal Surgery, First Affiliated Hospital, Nanchang University, Nanchang 330006, Jiangxi Province, People's Republic of China
| | - Yi Cao
- Department of Gastrointestinal Surgery, First Affiliated Hospital, Nanchang University, Nanchang 330006, Jiangxi Province, People's Republic of China
| | - Zhigang Jie
- Department of Gastrointestinal Surgery, First Affiliated Hospital, Nanchang University, Nanchang 330006, Jiangxi Province, People's Republic of China
| |
Collapse
|
7
|
Liu J, Liang L, Huang S, Nong L, Li D, Zhang B, Li T. Aberrant differential expression of EZH2 and H3K27me3 in extranodal NK/T-cell lymphoma, nasal type, is associated with disease progression and prognosis. Hum Pathol 2019; 83:166-176. [DOI: 10.1016/j.humpath.2018.08.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 08/22/2018] [Accepted: 08/29/2018] [Indexed: 12/21/2022]
|
8
|
Gan L, Xu M, Hua R, Tan C, Zhang J, Gong Y, Wu Z, Weng W, Sheng W, Guo W. The polycomb group protein EZH2 induces epithelial-mesenchymal transition and pluripotent phenotype of gastric cancer cells by binding to PTEN promoter. J Hematol Oncol 2018; 11:9. [PMID: 29335012 PMCID: PMC5769437 DOI: 10.1186/s13045-017-0547-3] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 12/26/2017] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The influences of oncogenic Ezh2 on the progression and prognosis of gastric cancer (GC) and the underlying mechanisms are still poorly understood. Here, we aimed at investigating clinicopathological significance of Ezh2 in GC and the mechanisms underlying its function in GC development. METHODS The expression level of Ezh2 was determined by qRT-PCR, immunoblot, and immunohistochemistry analysis in 156 pairs of GC tissues and adjacent normal gastric mucosa tissues. The biological functions of Ezh2 were assessed by in vitro and in vivo functional experiments. Chromatin immunoprecipitation (ChIP), luciferase, and Western blotting analyses were utilized to identify the relationship between Ezh2 and the PTEN/Akt signaling. RESULTS The expression of Ezh2 was higher in gastric cancer tissues in comparison with para-nontumorous epithelium. High expression of Ezh2 was associated with more aggressive biological behavior and poor prognosis in GC. In vitro studies indicated that Ezh2 promoted GC cells' proliferation and clonogenicity. Besides, Ezh2 led to the acquisition of epithelial-mesenchymal transition (EMT) phenotype of GC cells and enhanced GC cell migration and invasion capacity. In particular, Ezh2 strengthened sphere-forming capacity of GC cells, indicating its role in the enrichment of GC stem cells. Furthermore, we found that PTEN/Akt signaling contributed to the effects of Ezh2 on cancer stem cells (CSC) and EMT phenotype in GC cells, and blocking PTEN signaling significantly rescued the effects of Ezh2. CONCLUSIONS Taken together, Ezh2 has a central role in regulating diverse aspects of the pathogenesis of GC in part by involving PTEN/Akt signaling, indicating that it could be an independent prognostic factor and potential therapeutic target.
Collapse
Affiliation(s)
- Lu Gan
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Midie Xu
- Department of Pathology and tissue bank, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Ruixi Hua
- Department of Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510000, China
| | - Cong Tan
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Jieyun Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yiwei Gong
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhenhua Wu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Weiwei Weng
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Weiqi Sheng
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Weijian Guo
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
9
|
Pan YM, Wang CG, Zhu M, Xing R, Cui JT, Li WM, Yu DD, Wang SB, Zhu W, Ye YJ, Wu Y, Wang S, Lu YY. STAT3 signaling drives EZH2 transcriptional activation and mediates poor prognosis in gastric cancer. Mol Cancer 2016; 15:79. [PMID: 27938379 PMCID: PMC5148878 DOI: 10.1186/s12943-016-0561-z] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 11/23/2016] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND STAT3 signaling plays the pivotal role in tumorigenesis through EZH2 epigenetic modification, which enhanced STAT3 activity by increased tyrosine phosphorylation of STAT3. Here, another possible feedback mechanism and clinical significance of EZH2 and STAT3 were investigated in gastric cancer (GC). METHODS STAT3, p-STAT3 (Tyr 705) and EZH2 expression were examined in 63 GC specimens with matched normal tissues by IHC staining. EZH2 and STAT3 were also identified in five GC cell lines using RT-PCR and western blot analyses. p-STAT3 protein was detected by western blotting. In order to investigate whether EZH2 expression was directly regulated by STAT3, EZH2 expression was further detected using siRNA for STAT3 or IL-6 stimulation, with dual luciferase reporter analyses, electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation (ChIP) assays. The clinical significance of STAT3, p-STAT3 and EZH2 expression was evaluated by multi-factor COX regression and Kaplan-Meier analyses. RESULTS Hyper-activation of STAT3, p-STAT3 and EZH2 expression were observed in GC cells and tissues. STAT3 signaling was correlated with EZH2 expression in GC (R = 0.373, P = 0.003), which was consistent with our data showing that STAT3 as the transcriptional factor enhanced EZH2 transcriptional activity by binding the relative promoter region (-214 ~ -206). STAT3 was an independent signature for poor survival (P = 0.002). Patients with STAT3+/EZH2+ or p-STAT3+/EZH2+ had a worse outcome than others (P < 0.001); Besides, high levels of STAT3 and EZH2 was associated with advanced TNM staging (P = 0.017). Moreover, treatment with a combination of siSTAT3 and EZH2-specific inhibitor, 3-deazaneplanocin A (DZNEP), increased the apoptotic ratio of cells. It is benefit for targeting STAT3-EZH2 interplay in GC treatment. CONCLUSIONS Our results indicate that STAT3 status mediated EZH2 upregulation, associated with advanced TNM stage and poor prognosis, suggesting that combination with knockdown of STAT3 and EZH2 inhibitor might be a novel therapy in GC treatment. Collectively, STAT3, p-STAT3 and EZH2 expression were provided for the precision medicine in GC patients.
Collapse
Affiliation(s)
- Yuan-Ming Pan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute , 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Cheng-Gang Wang
- Department of Gastroenterology Surgery, Surgical Oncology Laboratory, People's Hospital, Peking University, Beijing, 100044, China.,Department of Cardiology, Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Min Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute , 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Rui Xing
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute , 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Jian-Tao Cui
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute , 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Wen-Mei Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute , 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - De-Dong Yu
- Department of Oncology/Institute for Cancer Research, Baotou Central Hospital, Inner Mongolia, 014040, China
| | - Shu-Bin Wang
- Department of Oncology/Institute for Cancer Research, Baotou Central Hospital, Inner Mongolia, 014040, China
| | - Wei Zhu
- Department of Oncology/Institute for Cancer Research, Baotou Central Hospital, Inner Mongolia, 014040, China
| | - Ying-Jiang Ye
- Department of Gastroenterology Surgery, Surgical Oncology Laboratory, People's Hospital, Peking University, Beijing, 100044, China
| | - Yun Wu
- Department of Oncology/Institute for Cancer Research, Baotou Central Hospital, Inner Mongolia, 014040, China. .,Department of Oncology/Institute for Cancer Research, Baotou Central Hospital, Baotou, 014040, People's Republic of China.
| | - Shan Wang
- Department of Gastroenterology Surgery, Surgical Oncology Laboratory, People's Hospital, Peking University, Beijing, 100044, China. .,Department of Gastroenterological Surgery, Surgical Oncology Laboratory, People's Hospital, Beijing University, No. 11, South Xizhimen Street, Beijing, 100044, People's Republic of China.
| | - You-Yong Lu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute , 52 Fucheng Road, Haidian District, Beijing, 100142, China.
| |
Collapse
|
10
|
Calcagno DQ, de Arruda Cardoso Smith M, Burbano RR. Cancer type-specific epigenetic changes: gastric cancer. Methods Mol Biol 2015; 1238:79-101. [PMID: 25421656 DOI: 10.1007/978-1-4939-1804-1_5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Gastric cancer (GC) remains a major cause of mortality despite declining rate in the world. Epigenetic alterations contribute significantly to the development and progression of gastric tumors. Epigenetic refers to the number of modifications of the chromatin structure that affect gene expression without altering the primary sequence of DNA, and these changes lead to transcriptional activation or silencing of the gene. Over the years, the study of epigenetic processes has increased, and novel therapeutic approaches have emerged. This chapter summarizes the main epigenomic mechanisms described recently involved in gastric carcinogenesis, focusing on the roles that aberrant DNA methylation, histone modifications (histone acetylation and methylation), and miRNAs (oncogenic and tumor suppressor function of miRNA) play in the onset and progression of gastric tumors. Clinical implications of these epigenetic alterations in GC are also discussed.
Collapse
Affiliation(s)
- Danielle Queiroz Calcagno
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Rua dos Mundurucus, 4487, Guamá, CEP 66073-000 Belém, PA, Brazil,
| | | | | |
Collapse
|
11
|
Calcagno DQ, Gigek CO, Chen ES, Burbano RR, Smith MDAC. DNA and histone methylation in gastric carcinogenesis. World J Gastroenterol 2013; 19:1182-92. [PMID: 23482412 PMCID: PMC3587474 DOI: 10.3748/wjg.v19.i8.1182] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Revised: 06/13/2012] [Accepted: 06/28/2012] [Indexed: 02/06/2023] Open
Abstract
Epigenetic alterations contribute significantly to the development and progression of gastric cancer, one of the leading causes of cancer death worldwide. Epigenetics refers to the number of modifications of the chromatin structure that affect gene expression without altering the primary sequence of DNA, and these changes lead to transcriptional activation or silencing of the gene. Over the years, the study of epigenetic processes has increased, and novel therapeutic approaches that target DNA methylation and histone modifications have emerged. A greater understanding of epigenetics and the therapeutic potential of manipulating these processes is necessary for gastric cancer treatment. Here, we review recent research on the effects of aberrant DNA and histone methylation on the onset and progression of gastric tumors and the development of compounds that target enzymes that regulate the epigenome.
Collapse
|
12
|
Lee H, Yoon SO, Jeong WY, Kim HK, Kim A, Kim BH. Immunohistochemical analysis of polycomb group protein expression in advanced gastric cancer. Hum Pathol 2012; 43:1704-10. [PMID: 22520951 DOI: 10.1016/j.humpath.2011.12.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Revised: 12/22/2011] [Accepted: 12/23/2011] [Indexed: 12/28/2022]
Abstract
The polycomb group proteins have recently captured the attention of cancer biologists. enhancer of zeste homologue 2 (EZH2) and B lymphoma Mo-MLV insertion region 1 homolog (BMI-1) are the best-characterized polycomb group proteins; their deregulation contributes to the development of many malignancies including gastric cancers. H3 trimethylation at lysine 27 and DNA methylase DNA methyltransferase 3B proteins are associated with the recruitment of polycomb group proteins. Overexpression of polycomb group proteins is associated with poor prognoses in some types of cancers but with favorable prognoses in others. In the present study, we investigated the expression of the polycomb group proteins EZH2 and BMI-1 and the associated proteins H3 trimethylation at lysine 27 and DNA methyltransferase 3B in advanced gastric cancers. Based on immunohistochemical detection, we evaluated the clinical relevance of these proteins in 178 cases of advanced gastric cancers that were managed with radical surgery and adjuvant systemic chemotherapy. BMI-1, enhancer of zeste homologue 2, H3 trimethylation at lysine 27, and DNA methyltransferase 3B proteins were overexpressed in the nuclei of gastric carcinoma compared with adjacent nonneoplastic gastric parenchyma. The high-level expression of BMI-1, enhancer of zeste homologue 2, H3 trimethylation at lysine 27, and DNA methyltransferase 3B proteins were frequently noted in advanced gastric cancer tissues (70.8%, 92.1%, 58.4%, and 64.6% of cases, respectively) and well intercorrelated in expression (P < .05). The expression level of BMI-1, enhancer of zeste homologue 2, and DNA methyltransferase 3B showed correlation with sex, gross type, and histologic type of the tumor among clinicopathologic variables. In terms of patient survival, low-level expression of enhancer of zeste homologue 2 was associated with cancer-related death (P = .018) and shorter overall survival (P = .005). Low-level expression of enhancer of zeste homologue 2 may represent a negative prognostic marker (P = .005) and indicate high risk in patients with advanced gastric cancer after surgery and adjuvant chemotherapy.
Collapse
Affiliation(s)
- Hyunjoo Lee
- Department of Pathology, Gangwon National University Hospital, Chuncheon 200-722, Korea
| | | | | | | | | | | |
Collapse
|
13
|
Co-expression of Bmi1 and EZH2 as an independent poor prognostic factor in esophageal squamous cell carcinoma. Pathol Res Pract 2012; 208:462-9. [PMID: 22766604 DOI: 10.1016/j.prp.2012.05.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Revised: 05/07/2012] [Accepted: 05/13/2012] [Indexed: 02/08/2023]
Abstract
Bmi1 polycomb ring finger oncogene (Bmi1) and the enhancer of zeste homolog 2 (EZH2) are members of polycomb repressive complex (PRC) 1 and PRC2, respectively. PRC1 represses tumor suppressor genes such as p16INK4a and p14ARF in a PRC2-dependent manner. There have been few studies on Bmi1 or EZH2 expression in esophageal squamous cell carcinoma (ESCC). We investigated Bmi1 and EZH2 expression in 164 cases of ESCCs using immunohistochemistry, and evaluated the correlation with clinicopathologic features and their prognostic significance. Bmi1 and EZH2 were more highly expressed in tumor than in adjacent normal tissue (p<0.001). High expression of Bmi1 or EZH2 alone was not correlated with any clinicopathologic parameter and did not influence the prognosis. However, the group with high expression of both Bmi1 and EZH2 showed the poorest prognosis in overall survival (p=0.027) and disease-free survival (p=0.007). Also, it was an independent prognostic factor in overall survival (p=0.047). High expression of both Bmi1 and EZH2, not each alone, is an independent poor prognostic factor in ESCCs, supporting the repression of tumor suppressor gene by Bmi1 in an EZH2-dependent manner. This result suggests that both Bmi1 and EZH2, not each alone, could be potent candidates of new target therapy in ESCCs.
Collapse
|
14
|
Cheng LL, Itahana Y, Lei ZD, Chia NY, Wu Y, Yu Y, Zhang SL, Thike AA, Pandey A, Rozen S, Voorhoeve PM, Yu Q, Tan PH, Bay BH, Itahana K, Tan P. TP53 genomic status regulates sensitivity of gastric cancer cells to the histone methylation inhibitor 3-deazaneplanocin A (DZNep). Clin Cancer Res 2012; 18:4201-12. [PMID: 22675170 DOI: 10.1158/1078-0432.ccr-12-0036] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE DZNep (3-deazaneplanocin A) depletes EZH2, a critical component of polycomb repressive complex 2 (PRC2), which is frequently deregulated in cancer. Despite exhibiting promising anticancer activity, the specific genetic determinants underlying DZNep responsiveness in cancer cells remain largely unknown. We sought to determine molecular factors influencing DZNep response in gastric cancer. EXPERIMENTAL DESIGN Phenotypic effects of DZNep were evaluated in a panel of gastric cancer cell lines. Sensitive lines were molecularly interrogated to identify potential predictors of DZNep responsiveness. The functional importance of candidate predictors was evaluated using short hairpin RNA (shRNA) and siRNA technologies. RESULTS DZNep depleted PRC2 pathway components in almost all gastric cancer lines, however, only a subset of lines exhibited growth inhibition upon treatment. TP53 genomic status was significantly associated with DZNep cellular responsiveness, with TP53 wild-type (WT) lines being more sensitive (P < 0.001). In TP53-WT lines, DZNep stabilized p53 by reducing ubiquitin conjugation through USP10 upregulation, resulting in activation of canonical p53 target genes. TP53 knockdown in TP53-WT lines attenuated DZNep sensitivity and p53 target activation, showing the functional importance of an intact p53 pathway in regulating DZNep cellular sensitivity. In primary human gastric cancers, EZH2 expression was negatively correlated with p53 pathway activation, suggesting that higher levels of EZH2 may repress p53 activity. CONCLUSION Our results highlight an important role for TP53 genomic status in influencing DZNep response in gastric cancer. Clinical trials evaluating EZH2-targeting agents such as DZNep should consider stratifying patients with gastric cancer by their TP53 genomic status.
Collapse
Affiliation(s)
- Lai Ling Cheng
- Cancer and Stem Cell Biology Program, Duke-NUS Graduate Medical School, Singapore
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Zhou Y, Du WD, Wu Q, Liu Y, Chen G, Ruan J, Xu S, Yang F, Zhou FS, Tang XF, Tang HY, Zuo XB, Zhang FY, Sun LD, Zhang XJ. EZH2 genetic variants affect risk of gastric cancer in the Chinese Han population. Mol Carcinog 2012; 53:589-97. [PMID: 22228224 DOI: 10.1002/mc.21871] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Revised: 12/09/2011] [Accepted: 12/12/2011] [Indexed: 12/20/2022]
Abstract
Enhancer of zeste 2 (EZH2) gene encodes a histone methyltransferase that constitutes the catalytic component of the polycomb repressive complex-2 (PRC2) to initiate epigenetic silencing of genes. It is reported that the expression level of EZH2 in gastric cancer tissue was highly correlated with tumor progression, however, whether EZH2 genetic variants were associated with the risk of gastric cancer remains yet unknown. In this study, we conducted a genotyping analysis for EZH2 in 311 cases of gastric cancer and 425 controls from the Chinese Han population. We found five single nucleotide polymorphisms (SNP; rs12670401, rs6464926, rs2072407, rs734005, and rs734004) of EZH2 gene were significantly associated with the risk of gastric cancer. Of which, the rs12670401 with the minor allele C and rs6464926 with the minor allele T revealed strong associations with increased gastric cancer risk [P = 0.009, adjusted odds ratio (aOR) = 1.327, 95% CI = 1.075-1.683 and P = 0.012, aOR = 1.310, 95% CI = 1.059-1.619]. The other three SNPs, rs2072407, rs734005, and rs734004 contributed to significantly reduced risk of gastric cancer (P = 0.033, aOR = 0.787, 95% CI = 0.633-0.981, P = 0.045, aOR = 0.799, 95% CI = 0.642-0.995 and P = 0.048, aOR = 0.803, 95% CI = 0.645-0.999), respectively. We further found that rs12670401 and rs6464926 were in a strong LD while rs2072407, rs734005, and rs734004 were in another. Haplotype analysis of the five SNPs showed that haplotype CCTCT reduced the risk of gastric cancer (P = 0.031 and aOR = 0.784), while haplotype GTCTC significantly elevated the risk of gastric cancer (P = 0.011 and aOR = 1.310). We concluded that EZH2 variants were significantly associated with gastric cancer risk. Our results for the first time provided new insight into susceptibility factors of EZH2 gene variants in carcinogenesis of gastric cancer of the Chinese Han population.
Collapse
Affiliation(s)
- Yuan Zhou
- Key Lab of Genome Research of Anhui Province, Anhui Medical University, Hefei, China; Department of Biology, Anhui Medical University, Hefei, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Piunti A, Pasini D. Epigenetic factors in cancer development: polycomb group proteins. Future Oncol 2011; 7:57-75. [PMID: 21174538 DOI: 10.2217/fon.10.157] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The role of chromatin-modifying factors in cancer biology emerged exponentially in the last 10 years, and increased attention has been focused on Polycomb group (PcG) proteins and their enzymatic activities. PcG proteins are repressive chromatin modifiers required for proliferation and development. The frequent deregulation of PcG activities in human tumors has direct oncogenic effects and results, essential for cancer cell proliferation. Here we will review the recent findings regarding PcG proteins in prospective tumor development, focusing on the molecular mechanisms that deregulate PcG expression in different tumors, at the downstream pathways to PcG expression (that contribute to cancer development) and at the mechanisms that regulate PcG recruitment to specific targets. Finally, we will speculate on the benefit of PcG inhibition for cancer treatment, reviewing potential pharmacological strategies.
Collapse
Affiliation(s)
- Andrea Piunti
- Department of Experimental Oncology, European Institute of Oncology, IFOM-IEO Campus, Via Adamello 16, Milan, Italy
| | | |
Collapse
|
17
|
Findeis-Hosey JJ, Huang J, Li F, Yang Q, McMahon LA, Xu H. High-grade neuroendocrine carcinomas of the lung highly express enhancer of zeste homolog 2, but carcinoids do not. Hum Pathol 2011; 42:867-72. [DOI: 10.1016/j.humpath.2010.09.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Revised: 08/31/2010] [Accepted: 09/17/2010] [Indexed: 10/18/2022]
|