1
|
Wang N, Miao X, Lu W, Ji Y, Zheng Y, Meng D, Liu H, Xiang C. RUNX3 exerts tumor-suppressive role through inhibiting EXOSC4 expression. Funct Integr Genomics 2024; 24:103. [PMID: 38913281 DOI: 10.1007/s10142-024-01363-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 04/23/2024] [Accepted: 05/01/2024] [Indexed: 06/25/2024]
Abstract
Breast cancer severely affects women health. 70% of breast cancer are estrogen receptor positive. Breast cancer stem cells are a group of tumor with plasticity, causing tumor relapse and metastasis. RUNX3 is a tumor suppressor frequently inactivated in estrogen receptor positive breast cancer. However, the mechanism of how RUNX3 is involved in the regualation of cancer stem cell traits in estrogen receptor positive breast cancer remains elusive. In this study, we utilized cut-tag assay to investigate the binding profile RUNX3 in BT474 and T47D cell, and confirmed EXOSC4 as the bona-fide target of RUNX3; RUNX3 could bind to the promoter are of EXOSC4 to suppress its expression. Furthermore, EXOSC4 could increase the colony formation, cell invasion and mammosphere formation ability of breast cancer cells and upregulate the the expression of SOX2 and ALDH1. Consistent with these findings, EXOSC4 was associated with poorer survival for Luminal B/Her2 breast cancer patiens. At last, we confirmed that EXOSC4 mediated the tumor suppressive role of RUNX3 in breast cancer cells. In conclusion, we demonstrate that RUNX3 directly binds to the promoter region of EXOSC4, leading to the suppression of EXOSC4 expression and exerting a tumor-suppressive effect in estrogen receptor postivive breast cancer cells.
Collapse
Affiliation(s)
- Nan Wang
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| | - Xiaodan Miao
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| | - Wenxin Lu
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| | - Yang Ji
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| | - Yuxin Zheng
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| | - Di Meng
- Department of Pathology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Hui Liu
- Department of Pathology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| | - Chenxi Xiang
- Department of Pathology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.
- Department of Pathology, Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
2
|
Park SA, Masunaga N, Kagara N, Ohi Y, Gondo N, Abe K, Yoshinami T, Sota Y, Miyake T, Tanei T, Shimoda M, Sagara Y, Shimazu K. Evaluation of RASSF1A methylation in the lysate of sentinel lymph nodes for detecting breast cancer metastasis: A diagnostic accuracy study. Oncol Lett 2023; 26:475. [PMID: 37809046 PMCID: PMC10551867 DOI: 10.3892/ol.2023.14063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 08/15/2023] [Indexed: 10/10/2023] Open
Abstract
The restriction enzyme-based digital methylation-specific polymerase chain reaction (RE-dMSP) assay is useful for diagnosing sentinel lymph node (SN) metastasis in patients with breast cancer, by detecting tumor-derived methylated Ras association domain-containing protein 1 (RASSF1A). In addition, this assay has high concordance (95.0%) with one-step nucleic acid amplification (OSNA). The present study aimed to perform RE-dMSP using OSNA lysate from more patients and to re-evaluate its clinical usage. Overall, 418 SNs from 347 patients were evaluated using both OSNA and RE-dMSP. The concordance rate was 83.3% (348/418). RASSF1A methylation of the primary tumors was negative in 36 patients. When these patients were excluded, the concordance rate improved to 88.2% (330/374). Of the 79 OSNA-negative cases, 19 were RE-dMSP-positive, although all were positive for cytokeratin 19 expression in the primary tumor, suggesting that RE-dMSP can detect tumor-derived DNA with a higher sensitivity. The percent of methylated reference of the breast tumors showed a wide variety in the 16 OSNA-positive/RE-dMSP-negative cases, and such variability of methylation could have affected the results in these patients. In conclusion, although RE-dMSP can diagnose SN metastasis with high sensitivity and accuracy, and can be a supplementary tool to OSNA in breast cancer, RE-dMSP showed certain discordance with OSNA and critically depended on the absence or heterogeneity of DNA methylation in breast tumors. Further research is expected to develop an assay targeting other DNA alterations, such as mutations.
Collapse
Affiliation(s)
- Sung Ae Park
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Nanae Masunaga
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Naofumi Kagara
- Department of Breast Surgery, Osaka General Medical Center, Osaka 558-8558, Japan
| | - Yasuyo Ohi
- Department of Breast Surgery, Hakuaikai Sagara Hospital, Kagoshima 892-0833, Japan
| | - Naomi Gondo
- Department of Breast Surgery, Hakuaikai Sagara Hospital, Kagoshima 892-0833, Japan
| | - Kaori Abe
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Tetsuhiro Yoshinami
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Yoshiaki Sota
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Tomohiro Miyake
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Tomonori Tanei
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Masafumi Shimoda
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Yasuaki Sagara
- Department of Breast Surgery, Hakuaikai Sagara Hospital, Kagoshima 892-0833, Japan
| | - Kenzo Shimazu
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| |
Collapse
|
3
|
Shu Y, Lan J, Hu Z, Liu W, Song R. Epigenetic regulation of RARB overcomes the radio-resistance of colorectal carcinoma cells via cancer stem cells. JOURNAL OF RADIATION RESEARCH 2023; 64:11-23. [PMID: 36214504 PMCID: PMC9855330 DOI: 10.1093/jrr/rrac060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/19/2022] [Indexed: 06/16/2023]
Abstract
Cancer stem cells (CSCs) are able to survive after cancer therapies, leading to cancer progression and recurrence in colorectal carcinoma (CRC). Therapies targeting CSCs are believed to be promising strategies for efficiently eradicating cancers. This study was to investigate that how retinoic acid receptor beta (RARB) affected the biological characteristics of CSCs and radio-resistance in CRC and the epigenetic mechanism. The sensitivity of CSCs isolated from HCT116 cells to radiotherapy was reduced compared with the parental cells. Using database querying, we found that RARB was one of the most significantly downregulated gene in radio-resistant cells in CRC. Also, RARB was poorly expressed in our isolated CSCs, and overexpression of RARB inhibited the properties of CSCs and enhanced radiotherapy sensitivity. Mechanistically, the methylation of RARB was higher in CSCs compared with HCT116 cells, which was significantly reduced after the application of DNA methylation inhibitor 5-azacytidine (5-azaC). DNA methyltransferases (DNMT1) was found to be recruited into the RARB promoter. 5-AzaC treatment inhibited DNMT1 activity and improved radiotherapy sensitivity by promoting RARB expression. Our results imply that inhibition of DNMT1 can display a new mechanism for the epigenetic mediation of RARB in radio-resistant CRC.
Collapse
Affiliation(s)
- Yuxian Shu
- Department of Comprehensive Radiotherapy, Jiangxi Cancer Hospital, Nanchang 330029, Jiangxi, P.R. China
| | - Jun Lan
- Department 1 of General Surgery, Jiangxi Gao’an People’s Hospital, Gao’an 330800, Jiangxi, P.R. China
| | - Zhaobing Hu
- Department of Oncology, Jingdezhen Second People’s Hospital, Jingdezhen 333000, Jiangxi, P.R. China
| | - Weiguo Liu
- Department of Gastroenterology, Jiangxi Cancer Hospital, Nanchang 330029, Jiangxi, P.R. China
| | - Rongfeng Song
- Corresponding author. Department of Gastroenterology, Jiangxi Cancer Hospital, No. 519, Beijing East Road, Qingshanhu District, Nanchang 330029, Jiangxi, P.R. China. ; Tel/Fax: 13879172671
| |
Collapse
|
4
|
Barrón-Gallardo CA, Garcia-Chagollán M, Morán-Mendoza AJ, Delgadillo-Cristerna R, Martínez-Silva MG, Villaseñor-García MM, Aguilar-Lemarroy A, Jave-Suárez LF. A gene expression signature in HER2+ breast cancer patients related to neoadjuvant chemotherapy resistance, overall survival, and disease-free survival. Front Genet 2022; 13:991706. [PMID: 36338974 PMCID: PMC9634254 DOI: 10.3389/fgene.2022.991706] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 10/11/2022] [Indexed: 11/27/2022] Open
Abstract
Breast cancer ranks first in terms of mortality and incidence rates worldwide among women. The HER2+ molecular subtype is one of the most aggressive subtypes; its treatment includes neoadjuvant chemotherapy and the use of a HER2 antibody. Some patients develop resistance despite positive results obtained using this therapeutic strategy. Objective. To identify prognostic markers for treatment and survival in HER2+ patients. Methods. Patients treated with neoadjuvant chemotherapy were assigned to sensitive and resistant groups based on their treatment response. Differentially expressed genes (DEGs) were identified using RNA-seq analysis. KEGG pathway, gene ontology, and interactome analyses were performed for all DEGs. An enrichment analysis Gene set enrichment analysis was performed. All DEGs were analyzed for overall (OS) and disease-free survival (DFS). Results. A total of 94 DEGs were related to treatment resistance. Survival analysis showed that 12 genes (ATF6B, DHRS13, DIRAS1, ERAL1, GRIN2B, L1CAM, IRX3, PRTFDC1, PBX2, S100B, SLC9A3R2, and TNXB) were good predictors of disease-free survival, and eight genes (GNG4, IL22RA2, MICA, S100B, SERPINF2, HLA-A, DIRAS1, and TNXB) were good predictors of overall survival (OS). Conclusion: We highlighted a molecular expression signature that can differentiate the treatment response, overall survival, and DFS of patients with HER2+ breast cancer.
Collapse
Affiliation(s)
- Carlos A. Barrón-Gallardo
- Programa de Doctorado en Ciencias Biomédicas, Centro Universitario de Ciencias de La Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - Mariel Garcia-Chagollán
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de La Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - Andres J. Morán-Mendoza
- Hospital de Ginecología, Centro Médico Nacional de Occidente, Instituto Mexicano Del Seguro Social (IMSS), Guadalajara, Mexico
| | - Raul Delgadillo-Cristerna
- Departamento de Radiología e Imagen, Centro Médico Nacional de Occidente, Instituto Mexicano Del Seguro Social (IMSS), Guadalajara, Mexico
| | - María G. Martínez-Silva
- Departamento de Anatomía Patológica, Centro Médico Nacional de Occidente, Instituto Mexicano Del Seguro Social (IMSS), Guadalajara, Mexico
| | - María M. Villaseñor-García
- División de Inmunología, Centro de Investigación Biomédica de Occidente, Instituto Mexicano Del Seguro Social (IMSS), Guadalajara, Mexico
| | - Adriana Aguilar-Lemarroy
- División de Inmunología, Centro de Investigación Biomédica de Occidente, Instituto Mexicano Del Seguro Social (IMSS), Guadalajara, Mexico
- *Correspondence: Adriana Aguilar-Lemarroy, ; Luis F. Jave-Suárez,
| | - Luis F. Jave-Suárez
- División de Inmunología, Centro de Investigación Biomédica de Occidente, Instituto Mexicano Del Seguro Social (IMSS), Guadalajara, Mexico
- *Correspondence: Adriana Aguilar-Lemarroy, ; Luis F. Jave-Suárez,
| |
Collapse
|
5
|
Belpaire M, Taminiau A, Geerts D, Rezsohazy R. HOXA1, a breast cancer oncogene. Biochim Biophys Acta Rev Cancer 2022; 1877:188747. [PMID: 35675857 DOI: 10.1016/j.bbcan.2022.188747] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/27/2022] [Accepted: 06/01/2022] [Indexed: 12/24/2022]
Abstract
More than 25 years ago, the first literature records mentioned HOXA1 expression in human breast cancer. A few years later, HOXA1 was confirmed as a proper oncogene in mammary tissue. In the following two decades, molecular data about the mode of action of the HOXA1 protein, the factors contributing to activate and maintain HOXA1 gene expression and the identity of its target genes have accumulated and provide a wider view on the association of this transcription factor to breast oncogenesis. Large-scale transcriptomic data gathered from wide cohorts of patients further allowed refining the relationship between breast cancer type and HOXA1 expression. Several recent reports have reviewed the connection between cancer hallmarks and the biology of HOX genes in general. Here we take HOXA1 as a paradigm and propose an extensive overview of the molecular data centered on this oncoprotein, from what its expression modulators, to the interactors contributing to its oncogenic activities, and to the pathways and genes it controls. The data converge to an intricate picture that answers questions on the multi-modality of its oncogene activities, point towards better understanding of breast cancer aetiology and thereby provides an appraisal for treatment opportunities.
Collapse
Affiliation(s)
- Magali Belpaire
- Animal Molecular and Cellular Biology Group (AMCB), Louvain Institute of Biomolecular Science and Technology (LIBST), UCLouvain, Louvain-la-Neuve, Belgium
| | - Arnaud Taminiau
- Animal Molecular and Cellular Biology Group (AMCB), Louvain Institute of Biomolecular Science and Technology (LIBST), UCLouvain, Louvain-la-Neuve, Belgium
| | - Dirk Geerts
- Heart Failure Research Center, Amsterdam University Medical Center (AMC), Universiteit van Amsterdam, Amsterdam, the Netherlands.
| | - René Rezsohazy
- Animal Molecular and Cellular Biology Group (AMCB), Louvain Institute of Biomolecular Science and Technology (LIBST), UCLouvain, Louvain-la-Neuve, Belgium.
| |
Collapse
|
6
|
Zhu C, Zhang S, Liu D, Wang Q, Yang N, Zheng Z, Wu Q, Zhou Y. A Novel Gene Prognostic Signature Based on Differential DNA Methylation in Breast Cancer. Front Genet 2021; 12:742578. [PMID: 34956313 PMCID: PMC8693898 DOI: 10.3389/fgene.2021.742578] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 11/15/2021] [Indexed: 12/11/2022] Open
Abstract
Background: DNA methylation played essential roles in regulating gene expression. The impact of DNA methylation status on the occurrence and development of cancers has been well demonstrated. However, little is known about its prognostic role in breast cancer (BC). Materials: The Illumina Human Methylation450 array (450k array) data of BC was downloaded from the UCSC xena database. Transcriptomic data of BC was downloaded from the Cancer Genome Atlas (TCGA) database. Firstly, we used univariate and multivariate Cox regression analysis to screen out independent prognostic CpGs, and then we identified methylation-associated prognosis subgroups by consensus clustering. Next, a methylation prognostic model was developed using multivariate Cox analysis and was validated with the Illumina Human Methylation27 array (27k array) dataset of BC. We then screened out differentially expressed genes (DEGs) between methylation high-risk and low-risk groups and constructed a methylation-based gene prognostic signature. Further, we validated the gene signature with three subgroups of the TCGA-BRCA dataset and an external dataset GSE146558 from the Gene Expression Omnibus (GEO) database. Results: We established a methylation prognostic signature and a methylation-based gene prognostic signature, and there was a close positive correlation between them. The gene prognostic signature involved six genes: IRF2, KCNJ11, ZDHHC9, LRP11, PCMT1, and TMEM70. We verified their expression in mRNA and protein levels in BC. Both methylation and methylation-based gene prognostic signatures showed good prognostic stratification ability. The AUC values of 3-years, 5-years overall survival (OS) were 0.737, 0.744 in the methylation signature and 0.725, 0.715 in the gene signature, respectively. In the validation groups, high-risk patients were confirmed to have poorer OS. The AUC values of 3 years were 0.757, 0.735, 0.733 in the three subgroups of TCGA dataset and 0.635 in GSE146558 dataset. Conclusion: This study revealed the DNA methylation landscape and established promising methylation and methylation-based gene prognostic signatures that could serve as potential prognostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Chunmei Zhu
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shuyuan Zhang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Di Liu
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Qingqing Wang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ningning Yang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhewen Zheng
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Qiuji Wu
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yunfeng Zhou
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
7
|
Sin RWY, Foo DCC, Iyer DN, Fan MSY, Li X, Lo OSH, Law WL, Ng L. A Pilot Study Investigating the Expression Levels of Pluripotency-Associated Genes in Rectal Swab Samples for Colorectal Polyp and Cancer Diagnosis and Prognosis. Stem Cells Int 2021; 2021:4139528. [PMID: 34335790 PMCID: PMC8324395 DOI: 10.1155/2021/4139528] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 12/11/2020] [Accepted: 06/09/2021] [Indexed: 02/06/2023] Open
Abstract
Change in gene expression is inevitable in cancer development. With more studies demonstrating the contributions of cancer stem cells (CSCs) in colorectal cancer (CRC) development, this study is aimed at investigating whether rectal swab specimen serves as a tool for detection of dysregulation of CSC or stem cell (SC) markers and at evaluating its potential as a new promising screening method for high-risk patients. Expression levels of 15 pluripotency-associated genes were assessed by quantitative PCR in 53 rectal swab specimens referred for endoscopic screening. Dysregulated genes and joint panels based on such genes were examined for their diagnostic potentials for both polyp and CRC. Out of 15 genes, Oct4, CD26, c-MYC, and CXCR4 showed significantly differential expression among normal, polyp, and CRC patients. A panel of Oct4 and CD26 showed an AUC value of 0.80 (p = 0.003) in identifying CRC patients from polyp/normal subjects, with sensitivity and specificity of 84.6% and 69.2%. A panel of c-MYC and CXCR4 achieved CRC/polyp identification with an AUC value of 0.79 (p = 0.002), with a sensitivity of 82.8% and specificity of 80.0%. The sensitivity for polyp and CRC was 80.0% and 85.7%, respectively. Further analysis showed that higher c-MYC and CXCR4 level was detected in normal subjects who developed polyps after 5-6 years, in comparison with subjects with no lesion developed, and the AUC of the c-MYC and CXCR4 panel increased to 0.88 (p < 0.001), with sensitivity and specificity of 84.4% and 92.3%, respectively, when these patients were included in the polyp group. This study suggests that the Oct4 and CD26 panel is a promising biomarker for distinguishing CRC from normal and polyp patients, whereas the c-MYC and CXCR4 panel may identify polyp and CRC from normal individuals.
Collapse
Affiliation(s)
- Ryan Wai-Yan Sin
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Dominic Chi-Chung Foo
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Deepak Narayanan Iyer
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - May Sau-Yee Fan
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Xue Li
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Oswens Siu-Hung Lo
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Wai-Lun Law
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Lui Ng
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
8
|
Li H, Wang X, Zhang M, Wang M, Zhang J, Ma S. Identification of HOXA1 as a Novel Biomarker in Prognosis of Head and Neck Squamous Cell Carcinoma. Front Mol Biosci 2021; 7:602068. [PMID: 33763449 PMCID: PMC7982851 DOI: 10.3389/fmolb.2020.602068] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/12/2020] [Indexed: 12/24/2022] Open
Abstract
Hox genes, a highly conserved homolog in most animals, play vital functions in cell development and organ formation. In recent years, researchers have discovered that it can act as a tumor regulator, and its members can participate in tumorigenesis by regulating receptor signaling, cell differentiation, apoptosis, migration, EMT, and angiogenesis. Hox genes and which major members play a vital role in the progress of head and neck squamous cell carcinoma (HNSCC) is still unclear. After analyzing the expression differences and prognostic value of all Hox genes through the TCGA-HNSC database, we use histochemistry stains in 52 pairs of HNSCC slices to verify the expression level of the key member-HOXA1. In correlation analysis, we found that high HOXA1 expression is related to poor pathological grade (p = 0.0077), advanced T stage (p = 0.021) and perineural invasion (PNI) (p = 0.0019). Furthermore, we used Cox univariate and multivariate regression analysis to confirm the independent predictive power of HOXA1 expression. To explore the underlying mechanisms behind HOXA1, we ran GSVA and GSEA and found fourteen mutual signaling pathways, including neuroprotein secretion and transport, tumor-associated signaling pathways, cell adhere junction and metabolic reprogramming. Finally, we found that the high expression of HOXA1 is significantly related to the decrease of CD8+ T cell infiltration and the decline of DNA methylation level. Our findings demonstrated that HOXA1, as a notable member of the HOX family, maybe an independent prognostic indicator in HNSCC.
Collapse
Affiliation(s)
- Hui Li
- Department of Otolaryngology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Xiaomin Wang
- Department of Otolaryngology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Mingjie Zhang
- Department of Otolaryngology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Mengjun Wang
- Department of Otolaryngology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Junjie Zhang
- Department of Otolaryngology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Shiyin Ma
- Department of Otolaryngology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| |
Collapse
|
9
|
Phillips-Chavez C, Watson M, Coward J, Schloss J. A systematic literature review assessing if genetic biomarkers are predictors for platinum-based chemotherapy response in ovarian cancer patients. Eur J Clin Pharmacol 2020; 76:1059-1074. [PMID: 32440721 DOI: 10.1007/s00228-020-02874-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 04/06/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Ovarian cancer is the deadliest of gynecologic malignancies with the 5-year overall survival rate remaining at approximately 30%, a rate that has not improved over the last three decades. Standard of care for epithelial ovarian cancer patients consists of a platinum compound with a taxane given intravenously following debulking surgery; however, 80% of cases relapse within 2 years of diagnosis. This review sought to identify key underlying biomarkers related to platinum resistance in ovarian cancer to establish possible prognostic biomarkers of chemoresponse. METHODS A systematic literature review was conducted across three databases PubMed, EMBASE and SCOPUS to summarise the evidence for prognostic biomarkers in platinum-resistant ovarian cancer patients. RESULTS Forty-eight human studies were used in the review encompassing 6719 participants in retrospective and prospective study designs. A total of 68 biomarkers were reported that were significantly correlated with chemoresponse and/or survival reporting a p value less than or equal to 0.05. CONCLUSION This review accentuates the pleiotropic phenotypic complexities related to the response to platinum therapy in ovarian cancer. A one-size-fits-all approach may be ineffective in a large portion of patients, emphasising the need for a whole system-based approach and personalised treatment strategies. Identifying key biomarkers to aid clinical decision-making is the first essential step in developing and appropriating therapies for at-risk patients, reducing toxicity and improving quality of life.
Collapse
Affiliation(s)
- Caitlin Phillips-Chavez
- Icon Cancer Centre, Southport, Australia.
- Endeavour College of Natural Health, 105 Scarborough Street, Southport, QLD, 4215, Australia.
| | - Michael Watson
- Endeavour College of Natural Health, 105 Scarborough Street, Southport, QLD, 4215, Australia
| | - Jermaine Coward
- Icon Cancer Centre, South Brisbane, Australia
- School of Medicine, University of Queensland, Brisbane, Australia
| | - Janet Schloss
- Endeavour College of Natural Health, Level 2, 269 Wickham Street, Fortitude Valley, Brisbane, QLD, 4006, Australia
| |
Collapse
|
10
|
Zhang F, Cui Y. Dysregulation of DNA methylation patterns may identify patients with breast cancer resistant to endocrine therapy: A predictive classifier based on differentially methylated regions. Oncol Lett 2019; 18:1287-1303. [PMID: 31423189 PMCID: PMC6607238 DOI: 10.3892/ol.2019.10405] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 04/15/2019] [Indexed: 02/06/2023] Open
Abstract
Endocrine therapy (ET) is one of a number of targeted therapies for estrogen receptor-positive breast cancer (BRCA); however, resistance to ET has become the primary issue affecting treatment outcome. In the present study, a predictive classifier was created using a DNA methylation dataset to identify patients susceptible to endocrine resistance. DNA methylation and RNA sequencing data, and the clinicopathological features of BRCA, were obtained from The Cancer Genome Atlas. Stringent criteria were set to select and classify patients into two groups, namely those resistant to ET (n=11) and sensitive to ET (n=21) groups. Bump hunting analysis revealed that 502 out of 135,418 genomic regions were differentially methylated between these two groups; these regions were differentially methylated regions (DMRs). The majority of the CpG sites contained in the DMRs mapped to the promoter region. Functional enrichment analyses indicated that a total of 562 specific genes encompassing these DMRs were primarily associated with 'biological progress of organ morphogenesis and development' and 'cell-cell adhesion' gene ontologies. Logistic regression and Pearson's correlation analysis were conducted to construct a predictive classifier for distinguishing patients resistant or sensitive to ET. The highest areas under the curve and relatively low Akaike information criterion values were associated with a total of 60 DMRs; a risk score retained from this classifier was revealed to be an unfavorable predictor of survival in two additional independent datasets. Furthermore, the majority of genes (55/63) exhibited a statistically significant association between DNA methylation and mRNA expression (P<0.05). The association between the mRNA expression of a number of genes (namely calcium release activated channel regulator 2A, Schlafen family member 12, chromosome 3 open reading frame 18, zinc finger protein 880, dual oxidase 1, major histocompatibility complex, class II, DP β1, C-terminal binding protein 1, ALG13 UDP-N-acetylglucosaminyltransferase subunit and RAS protein activator like 2) and the prognosis of patients with estrogen receptor-positive BRCA and ET resistance was determined using Kaplan-Meier Plotter. In summary, the predictive classifier proposed in the present study may aid the identification of patients sensitive or resistant to ET, and numerous genes maybe potential therapeutic targets to delay the development of resistance to ET.
Collapse
Affiliation(s)
- Fan Zhang
- Guangdong Provincial Key Laboratory for Breast Cancer Diagnosis and Treatment, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Yukun Cui
- Guangdong Provincial Key Laboratory for Breast Cancer Diagnosis and Treatment, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
- Correspondence to: Professor Yukun Cui, Guangdong Provincial Key Laboratory for Breast Cancer Diagnosis and Treatment, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou, Guangdong 515041, P.R. China, E-mail:
| |
Collapse
|
11
|
Hyaluronan-CD44 axis orchestrates cancer stem cell functions. Cell Signal 2019; 63:109377. [PMID: 31362044 DOI: 10.1016/j.cellsig.2019.109377] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 07/26/2019] [Accepted: 07/26/2019] [Indexed: 02/06/2023]
Abstract
The prominent role of CD44 in tumor cell signaling together with its establishment as a cancer stem cell (CSC) marker for various tumor entities imply a key role for CD44 in CSC functional properties. Hyaluronan, the main ligand of CD44, is a major constituent of CSC niche and, therefore, the hyaluronan-CD44 signaling axis is of functional importance in this special microenvironment. This review aims to provide recent advances in the importance of hyaluronan-CD44 interactions in the acquisition and maintenance of a CSC phenotype. Hyaluronan-CD44 axis has a substantial impact on stemness properties of CSCs and drug resistance through induction of EMT program, oxidative stress resistance, secretion of extracellular vesicles/exosomes and epigenetic control. Potential therapeutic approaches targeting CSCs based on the hyaluronan-CD44 axis are also presented.
Collapse
|
12
|
Schäfer SA, Hülsewig C, Barth P, von Wahlde MK, Tio J, Kolberg HC, Bernemann C, Blohmer JU, Kiesel L, Kolberg-Liedtke C. Correlation between SFRP1 expression and clinicopathological parameters in patients with triple-negative breast cancer. Future Oncol 2019; 15:1921-1938. [DOI: 10.2217/fon-2018-0564] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Aim: Breast cancer is a heterogeneous disease with distinct molecular and clinical behavior demanding reliable biomarkers, especially in triple-negative breast cancer (TNBC). This study seeks to improve the understanding of SFRP1 as a potential biomarker in breast cancer focusing on TNBC. Materials & methods: SFRP1 expression was investigated via immunohistochemistry with two anti-SFRP1-antibodies on tissue-microarrays of 376 invasive breast cancers. Results: Statistical analysis revealed a highly significant association between TNBC (n = 36) and SFRP1 expression (p < 0.001). SFRP1 expression was significantly associated with younger age, higher tumor stage, size and grade. Conclusion: SFRP1 expression is strongly correlated with TNBC on protein level. Associations with age and tumor grade support the role of SFRP1 as a biomarker for chemotherapy response in TNBC.
Collapse
Affiliation(s)
- Sarah Alexandra Schäfer
- Department of Pediatrics, Sana Kliniken Duisburg, Zu den Rehwiesen 9, 47055 Duisburg, Germany
| | - Carolin Hülsewig
- Molecular Health GmbH, Kurfürstenanlage 21, 69115 Heidelberg, Germany
| | - Peter Barth
- Gerhard-Domagk Departement for Pathology, University Münster, Albert-Schweitzer Campus 1 D17, 48149 Münster, Germany
| | - Marie-Kristin von Wahlde
- Department of Gynecology & Obstetrics, University Hospital Münster, Albert-Schweitzer Campus 1 A1, 48149 Münster, Germany
| | - Joke Tio
- Department of Gynecology & Obstetrics, University Hospital Münster, Albert-Schweitzer Campus 1 A1, 48149 Münster, Germany
| | - Hans-Christian Kolberg
- Department of Gynecology & Obstetrics, Marienhospital Bottrop, Josef-Albers-Str. 70, 46236 Bottrop, Germany
| | - Christof Bernemann
- Department of Urology, University Hospital Münster, Albert-Schweitzer Campus 1 A1, University Münster, Medical Faculty, Domagkstr, 48149 Münster, Germany
| | - Jens-Uwe Blohmer
- Department of Gynecology & Breast Center, Charité University Hospital Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Ludwig Kiesel
- Department of Gynecology & Obstetrics, University Hospital Münster, Albert-Schweitzer Campus 1 A1, 48149 Münster, Germany
| | - Cornelia Kolberg-Liedtke
- Department of Gynecology & Breast Center, Charité University Hospital Berlin, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
13
|
DNA Methylation Profiles and Their Diagnostic Utility in BC. DISEASE MARKERS 2019; 2019:6328503. [PMID: 31198475 PMCID: PMC6526564 DOI: 10.1155/2019/6328503] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 01/31/2019] [Accepted: 02/18/2019] [Indexed: 01/02/2023]
Abstract
Biomarkers, including DNA methylation, have shown a great potential for use in personalized medicine for BC and especially for the diagnosis of BC in developing countries. According to the bisulfite sequencing PCR in twelve specimens (BC and matched normal tissues), nine genetic probes were designed to detect the frequency of methylation of the promoters in a total of 302 paired cases of BC and matched normal breast tissues. Finally, a total of 900 serum samples were used to validate the use of these methylation biomarkers for clinical diagnosis of BC. A high frequency of promoter methylation of SFN, HOXA11, P16, RARβ, PCDHGB7, hMLH1, WNT5a, HOXD13, and RASSF1a was observed in BC tissues. The methylation frequencies of HOXD13 and hMLH1 increased with the progression of BC. The methylation frequencies of HOXD13 and WNT5a were significantly higher in BC. We found that methylation modification-positive samples were most consistently associated with luminal BC. Finally, we confirmed that RASSF1a, P16, and PCDHGB7 displayed a significant sensitivity and specificity as diagnostic biomarkers for BC (P < 0.001), and a panel that combined these three genes displayed increased significance (AUC, 0.781; P < 0.001). These data suggest that epigenetic markers in serum can potentially be used to diagnose BC. The identification of additional BC-specific methylated genes would improve the sensitivity and specificity of this approach. This study could also indicate that different molecular subtypes of BC are caused by distinct genetic and epigenetic mechanisms.
Collapse
|
14
|
Abdallah MOE, Algizouli UK, Suliman MA, Abdulrahman RA, Koko M, Fessahaye G, Shakir JH, Fahal AH, Elhassan AM, Ibrahim ME, Mohamed HS. EBV Associated Breast Cancer Whole Methylome Analysis Reveals Viral and Developmental Enriched Pathways. Front Oncol 2018; 8:316. [PMID: 30151354 PMCID: PMC6099083 DOI: 10.3389/fonc.2018.00316] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 07/24/2018] [Indexed: 01/18/2023] Open
Abstract
Background: Breast cancer (BC) ranks among the most common cancers in Sudan and worldwide with hefty toll on female health and human resources. Recent studies have uncovered a common BC signature characterized by low frequency of oncogenic mutations and high frequency of epigenetic silencing of major BC tumor suppressor genes. Therefore, we conducted a pilot genome-wide methylome study to characterize aberrant DNA methylation in breast cancer. Results: Differential methylation analysis between primary tumor samples and normal samples from healthy adjacent tissues yielded 20,188 differentially methylated positions (DMPs), which is further divided into 13,633 hypermethylated sites corresponding to 5339 genes and 6,555 hypomethylated sites corresponding to 2811 genes. Moreover, bioinformatics analysis revealed epigenetic dysregulation of major developmental pathways including hippo signaling pathway. We also uncovered many clues to a possible role for EBV infection in BC. Conclusion: Our results clearly show the utility of epigenetic assays in interrogating breast cancer tumorigenesis, and pinpointing specific developmental and viral pathways dysregulation that might serve as potential biomarkers or targets for therapeutic interventions.
Collapse
Affiliation(s)
- Mohammad O E Abdallah
- Department of Molecular Biology, Institute of Endemic Disease, University of Khartoum, Khartoum, Sudan
| | - Ubai K Algizouli
- Department of Molecular Biology, Institute of Endemic Disease, University of Khartoum, Khartoum, Sudan
| | - Maram A Suliman
- Department of Molecular Biology, Institute of Endemic Disease, University of Khartoum, Khartoum, Sudan
| | - Rawya A Abdulrahman
- Department of Molecular Biology, Institute of Endemic Disease, University of Khartoum, Khartoum, Sudan
| | - Mahmoud Koko
- Department of Molecular Biology, Institute of Endemic Disease, University of Khartoum, Khartoum, Sudan
| | - Ghimja Fessahaye
- Department of Molecular Biology, Institute of Endemic Disease, University of Khartoum, Khartoum, Sudan
| | - Jamal H Shakir
- Department of Surgery, Khartoum Teaching Hospital, Khartoum, Sudan
| | - Ahmed H Fahal
- Department of Surgery, Faculty of Medicine, University of Khartoum, Khartoum, Sudan
| | - Ahmed M Elhassan
- Department of Molecular Biology, Institute of Endemic Disease, University of Khartoum, Khartoum, Sudan
| | - Muntaser E Ibrahim
- Department of Molecular Biology, Institute of Endemic Disease, University of Khartoum, Khartoum, Sudan
| | - Hiba S Mohamed
- Department of Molecular Biology, Institute of Endemic Disease, University of Khartoum, Khartoum, Sudan.,Department of Biology, Taibah University, Medina, Saudi Arabia
| |
Collapse
|
15
|
Cai Y, Lin JR, Zhang Q, O'Brien K, Montagna C, Zhang ZD. Epigenetic alterations to Polycomb targets precede malignant transition in a mouse model of breast cancer. Sci Rep 2018; 8:5535. [PMID: 29615825 PMCID: PMC5882905 DOI: 10.1038/s41598-018-24005-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 03/26/2018] [Indexed: 12/16/2022] Open
Abstract
Malignant breast cancer remains a major health threat to women of all ages worldwide and epigenetic variations on DNA methylation have been widely reported in cancers of different types. We profiled DNA methylation with ERRBS (Enhanced Reduced Representation Bisulfite Sequencing) across four main stages of tumor progression in the MMTV-PyMT mouse model (hyperplasia, adenoma/mammary intraepithelial neoplasia, early carcinoma and late carcinoma), during which malignant transition occurs. We identified a large number of differentially methylated cytosines (DMCs) in tumors relative to age-matched normal mammary glands from FVB mice. Despite similarities, the methylation differences of the premalignant stages were distinct from the malignant ones. Many differentially methylated loci were preserved from the first to the last stage throughout tumor progression. Genes affected by methylation gains were enriched in Polycomb repressive complex 2 (PRC2) targets, which may present biomarkers for early diagnosis and targets for treatment.
Collapse
Affiliation(s)
- Ying Cai
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Jhih-Rong Lin
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Quanwei Zhang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Kelly O'Brien
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Cristina Montagna
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, USA.,Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Zhengdong D Zhang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, USA.
| |
Collapse
|
16
|
Breast cancer subtype dictates DNA methylation and ALDH1A3-mediated expression of tumor suppressor RARRES1. Oncotarget 2018; 7:44096-44112. [PMID: 27286452 PMCID: PMC5190082 DOI: 10.18632/oncotarget.9858] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 05/09/2016] [Indexed: 11/25/2022] Open
Abstract
Breast cancer subtyping, based on the expression of hormone receptors and other genes, can determine patient prognosis and potential options for targeted therapy. Among breast cancer subtypes, tumors of basal-like and claudin-low subtypes are typically associated with worse patient outcomes, are primarily classified as triple-negative breast cancers (TNBC), and cannot be treated with existing hormone-receptor-targeted therapies. Understanding the molecular basis of these subtypes will lead to the development of more effective treatment options for TNBC. In this study, we focus on retinoic acid receptor responder 1 (RARRES1) as a paradigm to determine if breast cancer subtype dictates protein function and gene expression regulation. Patient tumor dataset analysis and gene expression studies of a 26 cell-line panel, representing the five breast cancer subtypes, demonstrate that RARRES1 expression is greatest in basal-like TNBCs. Cell proliferation and tumor growth assays reveal that RARRES1 is a tumor suppressor in TNBC. Furthermore, gene expression studies, Illumina HumanMethylation450 arrays, and chromatin immunoprecipitation demonstrate that expression of RARRES1 is retained in basal-like breast cancers due to hypomethylation of the promoter. Additionally, expression of the cancer stem cell marker, aldehyde dehydrogenase 1A3, which provides the required ligand (retinoic acid) for RARRES1 transcription, is also specific to the basal-like subtype. We functionally demonstrate that the combination of promoter methylation and retinoic acid signaling dictates expression of tumor suppressor RARRES1 in a subtype-specific manner. These findings provide a precedent for a therapeutically-inducible tumor suppressor and suggest novel avenues of therapeutic intervention for patients with basal-like breast cancer.
Collapse
|
17
|
Abstract
Breast cancer is the most common cancer among women and represents one of the top five leading causes of cancer-related mortality. Inherited and acquired genetic mutations as well as epigenetic aberrations are known to be important contributors to the development and progression of breast cancer. Recent developments in high-throughput technologies have increased our understanding of the molecular changes in breast cancer, leading to the identification of distinctive genetic and epigenetic modifications in different breast cancer molecular subtypes. These genetic and epigenetic changes in luminal A, luminal B, ERBB2/HER2-enriched, basal-like, and normal-like breast cancer subtypes are discussed in this chapter. Furthermore, recent epigenome studies provided more information about further stratification of breast cancer subtypes, with essential role in the appropriate diagnosis and treatment of breast cancer. Thus, the inclusion of both genetic and epigenetic information in breast cancer clinical care could provide critical scientific base for precision medicine in breast cancer.
Collapse
|
18
|
Singla H, Ludhiadch A, Kaur RP, Chander H, Kumar V, Munshi A. Recent advances in HER2 positive breast cancer epigenetics: Susceptibility and therapeutic strategies. Eur J Med Chem 2017; 142:316-327. [DOI: 10.1016/j.ejmech.2017.07.075] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 07/27/2017] [Accepted: 07/31/2017] [Indexed: 12/31/2022]
|
19
|
Deb S, Gorringe KL, Pang JMB, Byrne DJ, Takano EA, Dobrovic A, Fox SB. BRCA2 carriers with male breast cancer show elevated tumour methylation. BMC Cancer 2017; 17:641. [PMID: 28893223 PMCID: PMC5594583 DOI: 10.1186/s12885-017-3632-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 08/28/2017] [Indexed: 12/19/2022] Open
Abstract
Background Male breast cancer (MBC) represents a poorly characterised group of tumours, the management of which is largely based on practices established for female breast cancer. However, recent studies demonstrate biological and molecular differences likely to impact on tumour behaviour and therefore patient outcome. The aim of this study was to investigate methylation of a panel of commonly methylated breast cancer genes in familial MBCs. Methods 60 tumours from 3 BRCA1 and 25 BRCA2 male mutation carriers and 32 males from BRCAX families were assessed for promoter methylation by methylation-sensitive high resolution melting in a panel of 10 genes (RASSF1A, TWIST1, APC, WIF1, MAL, RARβ, CDH1, RUNX3, FOXC1 and GSTP1). An average methylation index (AMI) was calculated for each case comprising the average of the methylation of the 10 genes tested as an indicator of overall tumour promoter region methylation. Promoter hypermethylation and AMI were correlated with BRCA carrier mutation status and clinicopathological parameters including tumour stage, grade, histological subtype and disease specific survival. Results Tumours arising in BRCA2 mutation carriers showed significantly higher methylation of candidate genes, than those arising in non-BRCA2 familial MBCs (average AMI 23.6 vs 16.6, p = 0.01, 45% of genes hypermethylated vs 34%, p < 0.01). RARβ methylation and AMI-high status were significantly associated with tumour size (p = 0.01 and p = 0.02 respectively), RUNX3 methylation with invasive carcinoma of no special type (94% vs 69%, p = 0.046) and RASSF1A methylation with coexistence of high grade ductal carcinoma in situ (33% vs 6%, p = 0.02). Cluster analysis showed MBCs arising in BRCA2 mutation carriers were characterised by RASSF1A, WIF1, RARβ and GTSP1 methylation (p = 0.02) whereas methylation in BRCAX tumours showed no clear clustering to particular genes. TWIST1 methylation (p = 0.001) and AMI (p = 0.01) were prognostic for disease specific survival. Conclusions Increased methylation defines a subset of familial MBC and with AMI may be a useful prognostic marker. Methylation might be predictive of response to novel therapeutics that are currently under investigation in other cancer types. Electronic supplementary material The online version of this article (10.1186/s12885-017-3632-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Siddhartha Deb
- Molecular Pathology Research and Development Laboratory, Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Vic, Parkville, 3010, Australia
| | - Kylie L Gorringe
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Vic, Parkville, 3010, Australia.,Cancer Genomics Program, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.,Department of Pathology, University of Melbourne, Parkville, VIC, 3012, Australia
| | - Jia-Min B Pang
- Molecular Pathology Research and Development Laboratory, Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - David J Byrne
- Molecular Pathology Research and Development Laboratory, Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Elena A Takano
- Molecular Pathology Research and Development Laboratory, Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | -
- Kathleen Cuningham Foundation Consortium for research into Familial Breast Cancer, Peter MacCallum Cancer Centre, Melbourne, 3000, Australia
| | - Alexander Dobrovic
- Molecular Pathology Research and Development Laboratory, Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.,Department of Pathology, University of Melbourne, Parkville, VIC, 3012, Australia.,Translational Genomics and Epigenomics Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia.,School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3084, Australia
| | - Stephen B Fox
- Molecular Pathology Research and Development Laboratory, Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia. .,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Vic, Parkville, 3010, Australia. .,Department of Pathology, University of Melbourne, Parkville, VIC, 3012, Australia. .,School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3084, Australia.
| |
Collapse
|
20
|
Chen E, Zheng F, Yuan X, Ye Y, Li X, Dai Y, Chen L. The effect of TMEFF2 methylation on the tumor stage and survival outcome of clear cell renal cell carcinoma. Cancer Biomark 2017; 19:207-212. [PMID: 28128743 DOI: 10.3233/cbm-161656] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Enjing Chen
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Fufu Zheng
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Xiaoxu Yuan
- Department of Urology, Jiangmen Central Hospital, Jiangmen, Guangdong 529030, China
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Yunlin Ye
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Xiaofei Li
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Yuping Dai
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Lingwu Chen
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| |
Collapse
|
21
|
Reaves DK, Hoadley KA, Fagan-Solis KD, Jima DD, Bereman M, Thorpe L, Hicks J, McDonald D, Troester MA, Perou CM, Fleming JM. Nuclear Localized LSR: A Novel Regulator of Breast Cancer Behavior and Tumorigenesis. Mol Cancer Res 2017; 15:165-178. [PMID: 27856957 PMCID: PMC5290211 DOI: 10.1158/1541-7786.mcr-16-0085-t] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 09/28/2016] [Accepted: 10/23/2016] [Indexed: 01/17/2023]
Abstract
Lipolysis-stimulated lipoprotein receptor (LSR) has been found in the plasma membrane and is believed to function in lipoprotein endocytosis and tight junctions. Given the impact of cellular metabolism and junction signaling pathways on tumor phenotypes and patient outcome, it is important to understand how LSR cellular localization mediates its functions. We conducted localization studies, evaluated DNA binding, and examined the effects of nuclear LSR in cells, xenografts, and clinical specimens. We found LSR within the membrane, cytoplasm, and the nucleus of breast cancer cells representing multiple intrinsic subtypes. Chromatin immunoprecipitation (ChIP) showed direct binding of LSR to DNA, and sequence analysis identified putative functional motifs and post-translational modifications of the LSR protein. While neither overexpression of transcript variants, nor pharmacologic manipulation of post-translational modification significantly altered localization, inhibition of nuclear export enhanced nuclear localization, suggesting a mechanism for nuclear retention. Coimmunoprecipitation and proximal ligation assays indicated LSR-pericentrin interactions, presenting potential mechanisms for nuclear-localized LSR. The clinical significance of LSR was evaluated using data from over 1,100 primary breast tumors, which showed high LSR levels in basal-like tumors and tumors from African-Americans. In tumors histosections, nuclear localization was significantly associated with poor outcomes. Finally, in vivo xenograft studies revealed that basal-like breast cancer cells that overexpress LSR exhibited both membrane and nuclear localization, and developed tumors with 100% penetrance, while control cells lacking LSR developed no tumors. These results show that nuclear LSR alters gene expression and may promote aggressive cancer phenotypes. IMPLICATIONS LSR functions in the promotion of aggressive breast cancer phenotypes and poor patient outcome via differential subcellular localization to alter cell signaling, bioenergetics, and gene expression. Mol Cancer Res; 15(2); 165-78. ©2016 AACR.
Collapse
Affiliation(s)
- Denise K Reaves
- Department of Biology, North Carolina Central University, Durham, North Carolina
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Katherine A Hoadley
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina
| | - Katerina D Fagan-Solis
- Department of Biology, North Carolina Central University, Durham, North Carolina
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Dereje D Jima
- Center for Human Health and the Environment, Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina
| | - Michael Bereman
- Center for Human Health and the Environment, Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina
| | - Lynnelle Thorpe
- Department of Biology, North Carolina Central University, Durham, North Carolina
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jyla Hicks
- Department of Biology, North Carolina Central University, Durham, North Carolina
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - David McDonald
- Department of Biology, North Carolina Central University, Durham, North Carolina
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Melissa A Troester
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Charles M Perou
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina
| | - Jodie M Fleming
- Department of Biology, North Carolina Central University, Durham, North Carolina.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Center for Human Health and the Environment, Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina
| |
Collapse
|
22
|
Yang Q, Pröll MJ, Salilew-Wondim D, Zhang R, Tesfaye D, Fan H, Cinar MU, Große-Brinkhaus C, Tholen E, Islam MA, Hölker M, Schellander K, Uddin MJ, Neuhoff C. LPS-induced expression of CD14 in the TRIF pathway is epigenetically regulated by sulforaphane in porcine pulmonary alveolar macrophages. Innate Immun 2016; 22:682-695. [PMID: 27688705 DOI: 10.1177/1753425916669418] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Pulmonary alveolar macrophages (AMs) are important in defense against bacterial lung inflammation. Cluster of differentiation 14 (CD14) is involved in recognizing bacterial lipopolysaccharide (LPS) through MyD88-dependent and TRIF pathways of innate immunity. Sulforaphane (SFN) shows anti-inflammatory activity and suppresses DNA methylation. To identify CD14 epigenetic changes by SFN in the LPS-induced TRIF pathway, an AMs model was investigated in vitro. CD14 gene expression was induced by 5 µg/ml LPS at the time point of 12 h and suppressed by 5 µM SFN. After 12 h of LPS stimulation, gene expression was significantly up-regulated, including TRIF, TRAF6, NF-κB, TRAF3, IRF7, TNF-α, IL-1β, IL-6, and IFN-β. LPS-induced TRAM, TRIF, RIPK1, TRAF3, TNF-α, IL-1β and IFN-β were suppressed by 5 µM SFN. Similarly, DNMT3a expression was increased by LPS but significantly down-regulated by 5 µM SFN. It showed positive correlation of CD14 gene body methylation with in LPS-stimulated AMs, and this methylation status was inhibited by SFN. This study suggests that SFN suppresses CD14 activation in bacterial inflammation through epigenetic regulation of CD14 gene body methylation associated with DNMT3a. The results provide insights into SFN-mediated epigenetic down-regulation of CD14 in LPS-induced TRIF pathway inflammation and may lead to new methods for controlling LPS-induced inflammation in pigs.
Collapse
Affiliation(s)
- Qin Yang
- 1 Department of Animal Breeding and Husbandry, Institute of Animal Science, University of Bonn, Germany
| | - Maren J Pröll
- 1 Department of Animal Breeding and Husbandry, Institute of Animal Science, University of Bonn, Germany
| | - Dessie Salilew-Wondim
- 1 Department of Animal Breeding and Husbandry, Institute of Animal Science, University of Bonn, Germany
| | - Rui Zhang
- 1 Department of Animal Breeding and Husbandry, Institute of Animal Science, University of Bonn, Germany
| | - Dawit Tesfaye
- 1 Department of Animal Breeding and Husbandry, Institute of Animal Science, University of Bonn, Germany
| | - Huitao Fan
- 2 Department of Basic Medical Sciences, and Purdue Center for Cancer Research, Purdue University, USA
| | - Mehmet U Cinar
- 3 Department of Animal Science, Faculty of Agriculture, Erciyes University, Turkey
| | - Christine Große-Brinkhaus
- 1 Department of Animal Breeding and Husbandry, Institute of Animal Science, University of Bonn, Germany
| | - Ernst Tholen
- 1 Department of Animal Breeding and Husbandry, Institute of Animal Science, University of Bonn, Germany
| | - Mohammad A Islam
- 4 Department of Medicine, Faculty of Veterinary Science, Bangladesh Agricultural University, Bangladesh
| | - Michael Hölker
- 1 Department of Animal Breeding and Husbandry, Institute of Animal Science, University of Bonn, Germany
| | - Karl Schellander
- 1 Department of Animal Breeding and Husbandry, Institute of Animal Science, University of Bonn, Germany
| | - Muhammad J Uddin
- 4 Department of Medicine, Faculty of Veterinary Science, Bangladesh Agricultural University, Bangladesh
| | - Christiane Neuhoff
- 1 Department of Animal Breeding and Husbandry, Institute of Animal Science, University of Bonn, Germany
| |
Collapse
|
23
|
Song XY, Li BY, Zhou EX, Wu FX. The clinicopathological significance of RUNX3 hypermethylation and mRNA expression in human breast cancer, a meta-analysis. Onco Targets Ther 2016; 9:5339-47. [PMID: 27616890 PMCID: PMC5008647 DOI: 10.2147/ott.s77828] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Aberrant promoter methylation of RUNX3 has been reported in several tumors including human breast cancer (BC). However, the association between RUNX3 hypermethylation and incidence of BC remains elusive. In this study, a detailed literature search was performed in Medline and Google Scholar for related research publications. Analysis of pooled data were executed. Odds ratios with corresponding confidence intervals were determined and summarized, respectively. Finally, 13 studies were identified for the meta-analysis. Analysis of the pooled data showed that RUNX3 hypermethylation was significantly higher in both ductal carcinoma in situ and invasive ductal carcinoma (IDC) than in normal breast tissues. In addition, RUNX3 methylation was significantly higher in IDC than in benign tumor. However, RUNX3 methylation was not significantly higher in IDC than in ductal carcinoma in situ. We also determined that RUNX3 hypermethylation was significantly higher in ER positive BC than in ER negative BC. In addition, high RUNX3 mRNA expression was found to be correlated with better overall survival and relapse-free survival for all BC patients. Our results strongly support that RUNX3 hypermethylation may play an important role in BC incidence. RUNX3 methylation is a valuable early biomarker for the diagnosis of BC. Further large-scale studies will provide more insight into the role of RUNX3 hypermethylation in the carcinogenesis and clinical diagnosis of BC patients.
Collapse
Affiliation(s)
- Xiao-Yun Song
- Department of Thoracic Surgery, Beijing Chest Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Bo-Yan Li
- Department of Breast Surgery, Inner Mongolia Forestry General Hospital, Inner Mongolia, People's Republic of China
| | - En-Xiang Zhou
- Department of General Surgery, the Second Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| | - Feng-Xia Wu
- Department of Breast Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
24
|
Zhao C, Li P, Zhang L, Wang B, Xiao L, Guo F, Wei Y. An Observational Study on Aberrant Methylation of Runx3 With the Prognosis in Chronic Atrophic Gastritis Patients. Medicine (Baltimore) 2016; 95:e3356. [PMID: 27196446 PMCID: PMC4902388 DOI: 10.1097/md.0000000000003356] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The aim of this study is to discuss whether the methylation levels of Runx3 could be used as the early biomarker for predicting the prognosis in chronic atrophic gastritis (CAG) patients. A total of 200 subjects including 60 controls without CAG (Group 1), 70 patients with mild CAG (Group 2), and 70 patients with moderate and severe CAG (Group 3) were recruited for this cross-sectional investigation in the Department of Gastroenterology in Daqing Oilfield General Hospital from July 2013 to May 2014. The MlALDI-TOF-MS was used to measure the methylation levels of Runx3 in all of the subjects. Real-time quantitative reverse transcription polymerase chain reaction and western blotting were chosen to determine the expression levels of Runx3. The correlations between methylation levels of Runx3 among these CAG patients and their prognosis were shown by logistic regression models. The results demonstrated that the methylation levels of CpG13, CpG14, and CpG15 in Runx3 were higher in Group 3 than those in Groups 1 and 2 (P <0.05), whereas the mRNA and protein expression levels of Runx3 were lower in Group 3 than those in Groups 1 and 2 (P <0.05). There were significantly negative correlations between the methylation levels of Runx3 with its expression and the healing prognosis of CAG patients. In brief, this study proved that the hypermethylation modifications of CpG13, CpG14, and CpG15 in the promoter region of Runx3 could result in the down regulation of Runx3 expression to affect the prognosis of CAG. So the methylation levels of these CpG sites in Runx3 in the peripheral blood can be used as the biomarker for predicting the healing prognosis of CAG patients.
Collapse
Affiliation(s)
- Chunna Zhao
- From Department of Gastroenterology (CZ, LZ, BW, LX, FG, YW), Daqing Oilfield General Hospital, Daqing City, Heilongjiang Province; Department of Nutrition Research Laboratory (PL), Beijing, Children's Hospital, Beijing City, China
| | | | | | | | | | | | | |
Collapse
|
25
|
Győrffy B, Bottai G, Fleischer T, Munkácsy G, Budczies J, Paladini L, Børresen-Dale AL, Kristensen VN, Santarpia L. Aberrant DNA methylation impacts gene expression and prognosis in breast cancer subtypes. Int J Cancer 2015; 138:87-97. [PMID: 26174627 DOI: 10.1002/ijc.29684] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 06/19/2015] [Accepted: 07/02/2015] [Indexed: 02/02/2023]
Abstract
DNA methylation has a substantial impact on gene expression, affecting the prognosis of breast cancer (BC) patients dependent on molecular subtypes. In this study, we investigated the prognostic relevance of the expression of genes reported as aberrantly methylated, and the link between gene expression and DNA methylation in BC subtypes. The prognostic value of the expression of 144 aberrantly methylated genes was evaluated in ER+/HER2-, HER2+, and ER-/HER2- molecular BC subtypes, in a meta-analysis of two large transcriptomic cohorts of BC patients (n = 1,938 and n = 1,640). The correlation between gene expression and DNA methylation in distinct gene regions was also investigated in an independent dataset of 104 BCs. Survival and Pearson correlation analyses were computed for each gene separately. The expression of 48 genes was significantly associated with BC prognosis (p < 0.05), and 32 of these prognostic genes exhibited a direct expression-methylation correlation. The expression of several immune-related genes, including CD3D and HLA-A, was associated with both relapse-free survival (HR = 0.42, p = 3.5E-06; HR = 0.35, p = 1.7E-08) and overall survival (HR = 0.50, p = 5.5E-04; HR = 0.68, p = 4.5E-02) in ER-/HER2- BCs. On the overall, the distribution of both positive and negative expression-methylation correlation in distinct gene regions have different effects on gene expression and prognosis in BC subtypes. This large-scale meta-analysis allowed the identification of several genes consistently associated with prognosis, whose DNA methylation could represent a promising biomarker for prognostication and clinical stratification of patients with distinct BC subtypes.
Collapse
Affiliation(s)
- Balázs Győrffy
- MTA TTK Lendület Cancer Biomarker Research Group, Budapest, Hungary.,2nd Dept. of Pediatrics, Semmelweis University, Budapest, Hungary.,MTA-SE Pediatrics and Nephrology Research Group, Budapest, Hungary
| | - Giulia Bottai
- Oncology Experimental Therapeutics Unit, IRCCS Clinical and Research Institute Humanitas, Rozzano - Milan, Italy
| | - Thomas Fleischer
- Department of Genetics, Institute for Cancer Research, OUS Radiumhospitalet, Oslo, Norway.,The K.G. Jebsen Center for Breast Cancer Research, Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Norway
| | - Gyöngyi Munkácsy
- MTA TTK Lendület Cancer Biomarker Research Group, Budapest, Hungary
| | - Jan Budczies
- Institute of Pathology, Campus Charité Mitte, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Laura Paladini
- Oncology Experimental Therapeutics Unit, IRCCS Clinical and Research Institute Humanitas, Rozzano - Milan, Italy
| | - Anne-Lise Børresen-Dale
- Department of Genetics, Institute for Cancer Research, OUS Radiumhospitalet, Oslo, Norway.,The K.G. Jebsen Center for Breast Cancer Research, Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Norway
| | - Vessela N Kristensen
- Department of Genetics, Institute for Cancer Research, OUS Radiumhospitalet, Oslo, Norway.,The K.G. Jebsen Center for Breast Cancer Research, Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Norway.,Department of Clinical Molecular Biology and Laboratory Science (EpiGen), Akershus University Hospital, Division of Medicine, Lørenskog, Norway
| | - Libero Santarpia
- Oncology Experimental Therapeutics Unit, IRCCS Clinical and Research Institute Humanitas, Rozzano - Milan, Italy
| |
Collapse
|
26
|
Milioli HH, Vimieiro R, Riveros C, Tishchenko I, Berretta R, Moscato P. The Discovery of Novel Biomarkers Improves Breast Cancer Intrinsic Subtype Prediction and Reconciles the Labels in the METABRIC Data Set. PLoS One 2015; 10:e0129711. [PMID: 26132585 PMCID: PMC4488510 DOI: 10.1371/journal.pone.0129711] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 05/12/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The prediction of breast cancer intrinsic subtypes has been introduced as a valuable strategy to determine patient diagnosis and prognosis, and therapy response. The PAM50 method, based on the expression levels of 50 genes, uses a single sample predictor model to assign subtype labels to samples. Intrinsic errors reported within this assay demonstrate the challenge of identifying and understanding the breast cancer groups. In this study, we aim to: a) identify novel biomarkers for subtype individuation by exploring the competence of a newly proposed method named CM1 score, and b) apply an ensemble learning, as opposed to the use of a single classifier, for sample subtype assignment. The overarching objective is to improve class prediction. METHODS AND FINDINGS The microarray transcriptome data sets used in this study are: the METABRIC breast cancer data recorded for over 2000 patients, and the public integrated source from ROCK database with 1570 samples. We first computed the CM1 score to identify the probes with highly discriminative patterns of expression across samples of each intrinsic subtype. We further assessed the ability of 42 selected probes on assigning correct subtype labels using 24 different classifiers from the Weka software suite. For comparison, the same method was applied on the list of 50 genes from the PAM50 method. CONCLUSIONS The CM1 score portrayed 30 novel biomarkers for predicting breast cancer subtypes, with the confirmation of the role of 12 well-established genes. Intrinsic subtypes assigned using the CM1 list and the ensemble of classifiers are more consistent and homogeneous than the original PAM50 labels. The new subtypes show accurate distributions of current clinical markers ER, PR and HER2, and survival curves in the METABRIC and ROCK data sets. Remarkably, the paradoxical attribution of the original labels reinforces the limitations of employing a single sample classifiers to predict breast cancer intrinsic subtypes.
Collapse
Affiliation(s)
- Heloisa Helena Milioli
- Priority Research Centre for Bioinformatics, Biomarker Discovery and Information-Based Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Environmental and Life Science, The University of Newcastle, Callaghan, NSW, Australia
| | - Renato Vimieiro
- Priority Research Centre for Bioinformatics, Biomarker Discovery and Information-Based Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- Centro de Informática, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Carlos Riveros
- Priority Research Centre for Bioinformatics, Biomarker Discovery and Information-Based Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Electrical Engineering and Computer Science, The University of Newcastle, Callaghan, NSW, Australia
| | - Inna Tishchenko
- Priority Research Centre for Bioinformatics, Biomarker Discovery and Information-Based Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Electrical Engineering and Computer Science, The University of Newcastle, Callaghan, NSW, Australia
| | - Regina Berretta
- Priority Research Centre for Bioinformatics, Biomarker Discovery and Information-Based Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Electrical Engineering and Computer Science, The University of Newcastle, Callaghan, NSW, Australia
| | - Pablo Moscato
- Priority Research Centre for Bioinformatics, Biomarker Discovery and Information-Based Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Electrical Engineering and Computer Science, The University of Newcastle, Callaghan, NSW, Australia
| |
Collapse
|
27
|
Abstract
Cancer stem cells (CSCs) are rare, tumour-initiating cells that exhibit stem cell properties: capacity of self-renewal, pluripotency, highly tumorigenic potential, and resistance to therapy. Cancer stem cells have been characterised and isolated from many cancers, including breast cancer. Developmental pathways, such as the Wnt/β-catenin, Notch/γ-secretase/Jagged, Shh (sonic hedgehog), and BMP signalling pathways, which direct proliferation and differentiation of normal stem cells, have emerged as major signalling pathways that contribute to the self-renewal of stem and/or progenitor cells in a variety of organs and cancers. Deregulation of these signalling pathways is frequently linked to an epithelial-mesenchymal transition (EMT), and breast CSCs often possess properties of cells that have undergone the EMT process. Signalling networks mediated by microRNAs and EMT-inducing transcription factors tie the EMT process to regulatory networks that maintain "stemness". Recent studies have elucidated epigenetic mechanisms that control pluripotency and stemness, which allows an assessment on how embryonic and normal tissue stem cells are deregulated during cancerogenesis to give rise to CSCs. Epigenetic-based mechanisms are reversible, and the possibility of "resetting" the abnormal cancer epigenome by applying pharmacological compounds targeting epigenetic enzymes is a promising new therapeutic strategy. Chemoresistance of CSCs is frequently driven by various mechanisms, including aberrant expression/activity of ABC transporters, aldehyde dehydrogenase and anti-oncogenic proteins (i.e. BCL2, B-cell lymphoma-2), enhanced DNA damage response, activation of pro-survival signalling pathways, and epigenetic deregulations. Despite controversy surrounding the CSC hypothesis, there is substantial evidence for their role in cancer, and a number of drugs intended to specifically target CSCs have entered clinical trials.
Collapse
|
28
|
Conway K, Edmiston SN, May R, Kuan PF, Chu H, Bryant C, Tse CK, Swift-Scanlan T, Geradts J, Troester MA, Millikan RC. DNA methylation profiling in the Carolina Breast Cancer Study defines cancer subclasses differing in clinicopathologic characteristics and survival. Breast Cancer Res 2014; 16:450. [PMID: 25287138 PMCID: PMC4303129 DOI: 10.1186/s13058-014-0450-6] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 09/24/2014] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION Breast cancer is a heterogeneous disease, with several intrinsic subtypes differing by hormone receptor (HR) status, molecular profiles, and prognosis. However, the role of DNA methylation in breast cancer development and progression and its relationship with the intrinsic tumor subtypes are not fully understood. METHODS A microarray targeting promoters of cancer-related genes was used to evaluate DNA methylation at 935 CpG sites in 517 breast tumors from the Carolina Breast Cancer Study, a population-based study of invasive breast cancer. RESULTS Consensus clustering using methylation (β) values for the 167 most variant CpG loci defined four clusters differing most distinctly in HR status, intrinsic subtype (luminal versus basal-like), and p53 mutation status. Supervised analyses for HR status, subtype, and p53 status identified 266 differentially methylated CpG loci with considerable overlap. Genes relatively hypermethylated in HR+, luminal A, or p53 wild-type breast cancers included FABP3, FGF2, FZD9, GAS7, HDAC9, HOXA11, MME, PAX6, POMC, PTGS2, RASSF1, RBP1, and SCGB3A1, whereas those more highly methylated in HR-, basal-like, or p53 mutant tumors included BCR, C4B, DAB2IP, MEST, RARA, SEPT5, TFF1, THY1, and SERPINA5. Clustering also defined a hypermethylated luminal-enriched tumor cluster 3 that gene ontology analysis revealed to be enriched for homeobox and other developmental genes (ASCL2, DLK1, EYA4, GAS7, HOXA5, HOXA9, HOXB13, IHH, IPF1, ISL1, PAX6, TBX1, SOX1, and SOX17). Although basal-enriched cluster 2 showed worse short-term survival, the luminal-enriched cluster 3 showed worse long-term survival but was not independently prognostic in multivariate Cox proportional hazard analysis, likely due to the mostly early stage cases in this dataset. CONCLUSIONS This study demonstrates that epigenetic patterns are strongly associated with HR status, subtype, and p53 mutation status and may show heterogeneity within tumor subclass. Among HR+ breast tumors, a subset exhibiting a gene signature characterized by hypermethylation of developmental genes and poorer clinicopathologic features may have prognostic value and requires further study. Genes differentially methylated between clinically important tumor subsets have roles in differentiation, development, and tumor growth and may be critical to establishing and maintaining tumor phenotypes and clinical outcomes.
Collapse
|
29
|
MicroRNA-9 is associated with epithelial-mesenchymal transition, breast cancer stem cell phenotype, and tumor progression in breast cancer. Breast Cancer Res Treat 2014; 147:39-49. [PMID: 25086633 DOI: 10.1007/s10549-014-3069-5] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Accepted: 07/19/2014] [Indexed: 12/11/2022]
Abstract
MicroRNAs (miRNAs) are involved in the progression of breast cancer. Some miRNAs, especially the miR-200 family, miR-9, and miR-155 have been reported to be associated with epithelial-mesenchymal transition (EMT) and breast cancer stem cell (BCSC) phenotypes. This study was designed to evaluate the expression levels of these miRNAs in human breast cancer samples and analyzed their relationship with clinicopathologic features of the tumor including breast cancer subtype, EMT, BCSC phenotype, and prognosis. Expression levels of the miR-200 family, miR-9, and miR-155 were quantified using qRT-PCR. Breast cancer subtype, BCSC phenotype (CD44+/CD24- and ALDH1+), and expression of EMT markers (vimentin expression and E-cadherin loss) were evaluated by immunohistochemistry. miR-9 was more highly expressed in HER2+ and triple-negative subtypes than in luminal subtypes. Its expression level was significantly higher in tumors with high T stage, high histologic grade, p53 overexpression, and high proliferation index. Expression of miR-9 was also higher in tumors showing the CD44+/CD24- phenotype, vimentin expression, and E-cadherin loss. Furthermore, high level of miR-9 expression was found to be an independent prognostic factor for poor disease-free survival of the patients. Expression of miR-200a and miR-141 was highest in luminal A subtype, and miR-155 expression was highest in triple-negative subtype. Although the expression levels of some miR-200 family members and miR-155 showed difference with regard to EMT or BCSC phenotype, they were not associated with patients' prognosis. In conclusion, overexpression of miR-9 is found in tumors with aggressive phenotypes and is associated with poor prognosis in breast cancer, suggesting that it may serve as a potential biomarker for breast cancer progression and a target for treatment.
Collapse
|
30
|
Marcato P, Dean CA, Liu RZ, Coyle KM, Bydoun M, Wallace M, Clements D, Turner C, Mathenge EG, Gujar SA, Giacomantonio CA, Mackey JR, Godbout R, Lee PWK. Aldehyde dehydrogenase 1A3 influences breast cancer progression via differential retinoic acid signaling. Mol Oncol 2014; 9:17-31. [PMID: 25106087 DOI: 10.1016/j.molonc.2014.07.010] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 07/12/2014] [Accepted: 07/13/2014] [Indexed: 01/08/2023] Open
Abstract
Aldehyde dehydrogenase (ALDH) 1A enzymes produce retinoic acid (RA), a transcription induction molecule. To investigate if ALDH1A1 or ALDH1A3-mediated RA signaling has an active role in breast cancer tumorigenesis, we performed gene expression and tumor xenograft studies. Analysis of breast patient tumors revealed that high levels of ALDH1A3 correlated with expression of RA-inducible genes with retinoic acid response elements (RAREs), poorer patient survival and triple-negative breast cancers. This suggests a potential link between ALDH1A3 expression and RA signaling especially in aggressive and/or triple-negative breast cancers. In MDA-MB-231, MDA-MB-468 and MDA-MB-435 cells, ALDH1A3 and RA increased expression of RA-inducible genes. Interestingly, ALDH1A3 had opposing effects in tumor xenografts, increasing tumor growth and metastasis of MDA-MB-231 and MDA-MB-435 cells, but decreasing tumor growth of MDA-MB-468 cells. Exogenous RA replaced ALDH1A3 in inducing the same opposing tumor growth and metastasis effects, suggesting that ALDH1A3 mediates these effects by promoting RA signaling. Genome expression analysis revealed that ALDH1A3 induced largely divergent gene expression in MDA-MB-231 and MDA-MB-468 cells which likely resulted in the opposing tumor growth effects. Treatment with DNA methylation inhibitor 5-aza-2'deoxycytidine restored uniform RA-inducibility of RARE-containing HOXA1 and MUC4 in MDA-MB-231 and MDA-MB-468 cells, suggesting that differences in epigenetic modifications contribute to differential ALDH1A3/RA-induced gene expression in breast cancer. In summary, ALDH1A3 induces differential RA signaling in breast cancer cells which affects the rate of breast cancer progression.
Collapse
Affiliation(s)
- Paola Marcato
- Department of Pathology, Dalhousie University, Halifax, NS, Canada; Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada.
| | - Cheryl A Dean
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada
| | - Rong-Zong Liu
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Krysta M Coyle
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| | - Moamen Bydoun
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| | - Melissa Wallace
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| | - Derek Clements
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| | - Colin Turner
- Department of Surgery, Dalhousie University, Halifax, NS, Canada
| | | | - Shashi A Gujar
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada; Strategy and Organizational Performance, IWK Health Centre, Halifax, NS, Canada
| | | | - John R Mackey
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Roseline Godbout
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Patrick W K Lee
- Department of Pathology, Dalhousie University, Halifax, NS, Canada; Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada.
| |
Collapse
|
31
|
Park SY, Seo AN, Jung HY, Gwak JM, Jung N, Cho NY, Kang GH. Alu and LINE-1 hypomethylation is associated with HER2 enriched subtype of breast cancer. PLoS One 2014; 9:e100429. [PMID: 24971511 PMCID: PMC4074093 DOI: 10.1371/journal.pone.0100429] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 05/28/2014] [Indexed: 01/31/2023] Open
Abstract
The changes in DNA methylation status in cancer cells are characterized by hypermethylation of promoter CpG islands and diffuse genomic hypomethylation. Alu and long interspersed nucleotide element-1 (LINE-1) are non-coding genomic repetitive sequences and methylation of these elements can be used as a surrogate marker for genome-wide methylation status. This study was designed to evaluate the changes of Alu and LINE-1 hypomethylation during breast cancer progression from normal to pre-invasive lesions and invasive breast cancer (IBC), and their relationship with characteristics of IBC. We analyzed the methylation status of Alu and LINE-1 in 145 cases of breast samples including normal breast tissue, atypical ductal hyperplasia/flat epithelial atypia (ADH/FEA), ductal carcinoma in situ (DCIS) and IBC, and another set of 129 cases of IBC by pyrosequencing. Alu methylation showed no significant changes during multistep progression of breast cancer, although it tended to decrease during the transition from DCIS to IBC. In contrast, LINE-1 methylation significantly decreased from normal to ADH/FEA, while it was similar in ADH/FEA, DCIS and IBC. In IBC, Alu hypomethylation correlated with negative estrogen receptor (ER) status, and LINE-1 hypomethylation was associated with negative ER status, ERBB2 (HER2) amplification and p53 overexpression. Alu and LINE-1 methylation status was significantly different between breast cancer subtypes, and the HER2 enriched subtype had lowest methylation levels. In survival analyses, low Alu methylation status tended to be associated with poor disease-free survival of the patients. Our findings suggest that LINE-1 hypomethylation is an early event and Alu hypomethylation is probably a late event during breast cancer progression, and prominent hypomethylation of Alu and LINE-1 in HER2 enriched subtype may be related to chromosomal instability of this specific subtype.
Collapse
Affiliation(s)
- So Yeon Park
- Department of Pathology, Seoul National University College of Medicine, Jongno-gu, Seoul, Korea
- Department of Pathology, Seoul National University Bundang Hospital, Bundang-gu, Seongnam, Gyeonggi, Korea
| | - An Na Seo
- Department of Pathology, Seoul National University Bundang Hospital, Bundang-gu, Seongnam, Gyeonggi, Korea
| | - Hae Yoen Jung
- Department of Pathology, Seoul National University Bundang Hospital, Bundang-gu, Seongnam, Gyeonggi, Korea
| | - Jae Moon Gwak
- Department of Pathology, Seoul National University College of Medicine, Jongno-gu, Seoul, Korea
| | - Namhee Jung
- Laboratory of Epigenetics, Cancer Research Institute, Seoul National University, Jongno-gu, Seoul, Korea
| | - Nam-Yun Cho
- Laboratory of Epigenetics, Cancer Research Institute, Seoul National University, Jongno-gu, Seoul, Korea
| | - Gyeong Hoon Kang
- Department of Pathology, Seoul National University College of Medicine, Jongno-gu, Seoul, Korea
- Laboratory of Epigenetics, Cancer Research Institute, Seoul National University, Jongno-gu, Seoul, Korea
- * E-mail:
| |
Collapse
|
32
|
Yu YY, Chen C, Kong FF, Zhang W. Clinicopathological significance and potential drug target of RUNX3 in breast cancer. Drug Des Devel Ther 2014; 8:2423-30. [PMID: 25525332 PMCID: PMC4266273 DOI: 10.2147/dddt.s71815] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Previous reports indicate that RUNX3 is a tumor suppressor in several types of human tumors, including breast cancer (BC). However, the correlation between RUNX3 hypermethylation and the incidence of BC remains unclear. In this study, we conducted a systematic review and meta-analysis aiming to comprehensively assess the potential role of RUNX3 hypermethylation in the pathogenesis of BC. METHODS A detailed literature search was made to identify studies for related research publications. Methodological quality of the studies was evaluated. Analysis of pooled data was performed. Odds ratio (OR) was calculated and summarized respectively. RESULTS Final analysis of 565 BC patients from eleven eligible studies was performed. The results showed that RUNX3 hypermethylation was significantly higher in BC than in normal breast tissue, the pooled OR from nine studies including 339 BC and 248 normal breast tissue (OR =24.12, 95% confidence interval [CI] =13.50-43.11, Z=10.75, P<0.00001). Further analysis also showed significantly increased OR of RUNX3 hypermethylation in estrogen receptor (ER)-positive than in ER-negative BC patients (OR =5.67, 95% CI =2.69-11.95, Z=4.57, P<0.00001). In addition, RUNX3 messenger RNA (mRNA) high expression was found to be correlated to better overall survival in 3,455 cases of BC patients that were followed up for 20 years (hazard ratio [HR] 0.79, P=8.8×10(-5)). Interestingly, RUNX3 mRNA overexpression was found to be correlated to better overall survival in only 668 cases of ER-negative patients (HR 0.72, P=0.01), but not in 1,767 cases of ER-positive patients (HR 0.87, P=0.13). CONCLUSION The results of this meta-analysis suggest that RUNX3 hypermethylation may be implicated in the pathogenesis of BC. Detection of RUNX3 mRNA may be a helpful and valuable biomarker for diagnosis of BC, especially in ER-negative BC. We also discussed the significance of RUNX3 as a potential drug target.
Collapse
Affiliation(s)
- Ying-Ying Yu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Chao Chen
- Department of Gynecology, Shanghai First Maternity and Infant Hospital Affiliated to TongJi University, Shanghai, People’s Republic of China
| | - Fan-fei Kong
- Department of Gynecology, Shanghai First Maternity and Infant Hospital Affiliated to TongJi University, Shanghai, People’s Republic of China
| | - Wei Zhang
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, People’s Republic of China
- Correspondence: Wei Zhang, Obstetrics and Gynecology Hospital, Fudan University, 413 Zhaozhou Road, Shanghai 200011, People’s Republic of China, Email
| |
Collapse
|
33
|
Bu D, Lewis CM, Sarode V, Chen M, Ma X, Lazorwitz AM, Rao R, Leitch M, Moldrem A, Andrews V, Gazdar A, Euhus D. Identification of Breast Cancer DNA Methylation Markers Optimized for Fine-Needle Aspiration Samples. Cancer Epidemiol Biomarkers Prev 2013; 22:2212-21. [DOI: 10.1158/1055-9965.epi-13-0208] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
34
|
Cohen AL, Piccolo SR, Cheng L, Soldi R, Han B, Johnson WE, Bild AH. Genomic pathway analysis reveals that EZH2 and HDAC4 represent mutually exclusive epigenetic pathways across human cancers. BMC Med Genomics 2013; 6:35. [PMID: 24079712 PMCID: PMC3850967 DOI: 10.1186/1755-8794-6-35] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 09/19/2013] [Indexed: 12/19/2022] Open
Abstract
Background Alterations in epigenetic marks, including methylation or acetylation, are common in human cancers. For many epigenetic pathways, however, direct measures of activity are unknown, making their role in various cancers difficult to assess. Gene expression signatures facilitate the examination of patterns of epigenetic pathway activation across and within human cancer types allowing better understanding of the relationships between these pathways. Methods We used Bayesian regression to generate gene expression signatures from normal epithelial cells before and after epigenetic pathway activation. Signatures were applied to datasets from TCGA, GEO, CaArray, ArrayExpress, and the cancer cell line encyclopedia. For TCGA data, signature results were correlated with copy number variation and DNA methylation changes. GSEA was used to identify biologic pathways related to the signatures. Results We developed and validated signatures reflecting downstream effects of enhancer of zeste homolog 2(EZH2), histone deacetylase(HDAC) 1, HDAC4, sirtuin 1(SIRT1), and DNA methyltransferase 2(DNMT2). By applying these signatures to data from cancer cell lines and tumors in large public repositories, we identify those cancers that have the highest and lowest activation of each of these pathways. Highest EZH2 activation is seen in neuroblastoma, hepatocellular carcinoma, small cell lung cancer, and melanoma, while highest HDAC activity is seen in pharyngeal cancer, kidney cancer, and pancreatic cancer. Across all datasets studied, activation of both EZH2 and HDAC4 is significantly underrepresented. Using breast cancer and glioblastoma as examples to examine intrinsic subtypes of particular cancers, EZH2 activation was highest in luminal breast cancers and proneural glioblastomas, while HDAC4 activation was highest in basal breast cancer and mesenchymal glioblastoma. EZH2 and HDAC4 activation are associated with particular chromosome abnormalities: EZH2 activation with aberrations in genes from the TGF and phosphatidylinositol pathways and HDAC4 activation with aberrations in inflammatory and chemokine related genes. Conclusion Gene expression patterns can reveal the activation level of epigenetic pathways. Epigenetic pathways define biologically relevant subsets of human cancers. EZH2 activation and HDAC4 activation correlate with growth factor signaling and inflammation, respectively, and represent two distinct states for cancer cells. This understanding may allow us to identify targetable drivers in these cancer subsets.
Collapse
Affiliation(s)
- Adam L Cohen
- Huntsman Cancer Institute, 2000 Circle of Hope, Salt Lake City, UT 84112, USA.
| | | | | | | | | | | | | |
Collapse
|
35
|
Balic M, Schwarzenbacher D, Stanzer S, Heitzer E, Auer M, Geigl JB, Cote RJ, Datar RH, Dandachi N. Genetic and epigenetic analysis of putative breast cancer stem cell models. BMC Cancer 2013; 13:358. [PMID: 23883436 PMCID: PMC3727963 DOI: 10.1186/1471-2407-13-358] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Accepted: 07/22/2013] [Indexed: 12/17/2022] Open
Abstract
Background Cancer stem cell model hypothesizes existence of a small proportion of tumor cells capable of sustaining tumor formation, self-renewal and differentiation. In breast cancer, these cells were found to be associated with CD44+CD24-low and ALDH+ phenotype. Our study was performed to evaluate the suitability of current approaches for breast cancer stem cell analyses to evaluate heterogeneity of breast cancer cells through their extensive genetic and epigenetic characterization. Methods Breast cancer cell lines MCF7 and SUM159 were cultured in adherent conditions and as mammospheres. Flow cytometry sorting for CD44, CD24 and ALDH was performed. Sorted and unsorted populations, mammospheres and adherent cell cultures were subjected to DNA profiling by array CGH and methylation profiling by Epitect Methyl qPCR array. Methylation status of selected genes was further evaluated by pyrosequencing. Functional impact of methylation was evaluated by mRNA analysis for selected genes. Results Array CGH did not reveal any genomic differences. In contrast, putative breast cancer stem cells showed altered methylation levels of several genes compared to parental tumor cells. Conclusions Our results underpin the hypothesis that epigenetic mechanisms seem to play a major role in the regulation of CSCs. However, it is also clear that more efficient methods for CSC enrichment are needed. This work underscores requirement of additional approaches to reveal heterogeneity within breast cancer.
Collapse
Affiliation(s)
- Marija Balic
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, A-8036 Graz, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Chimge NO, Frenkel B. The RUNX family in breast cancer: relationships with estrogen signaling. Oncogene 2013; 32:2121-30. [PMID: 23045283 PMCID: PMC5770236 DOI: 10.1038/onc.2012.328] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Revised: 06/20/2012] [Accepted: 06/20/2012] [Indexed: 12/22/2022]
Abstract
The three RUNX family members are lineage specific master regulators, which also have important, context-dependent roles in carcinogenesis as either tumor suppressors or oncogenes. Here we review evidence for such roles in breast cancer (BCa). RUNX1, the predominant RUNX family member in breast epithelial cells, has a tumor suppressor role reflected by many somatic mutations found in primary tumor biopsies. The classical tumor suppressor gene RUNX3 does not consist of such a mutation hot spot, but it too seems to inhibit BCa; it is often inactivated in human BCa tumors and its haploinsufficiency in mice leads to spontaneous BCa development. The tumor suppressor activities of RUNX1 and RUNX3 are mediated in part by antagonism of estrogen signaling, a feature recently attributed to RUNX2 as well. Paradoxically, however RUNX2, a master osteoblast regulator, has been implicated in various aspects of metastasis in general and bone metastasis in particular. Reciprocating the anti-estrogenic tumor suppressor activity of RUNX proteins, inhibition of RUNX2 by estrogens may help explain their context-dependent anti-metastatic roles. Such roles are reserved to non-osseous metastasis, because ERα is associated with increased, not decreased skeletal dissemination of BCa cells. Finally, based on diverse expression patterns in BCa subtypes, the successful use of future RUNX-based therapies will most likely require careful patient selection.
Collapse
Affiliation(s)
- N-O Chimge
- Department of Biochemistry and Molecular Biology, Institute for Genetic Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - B Frenkel
- Departments of Orthopaedic Surgery and Biochemistry and Molecular Biology, Institute for Genetic Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
37
|
Ogino S, Lochhead P, Chan AT, Nishihara R, Cho E, Wolpin BM, Meyerhardt JA, Meissner A, Schernhammer ES, Fuchs CS, Giovannucci E. Molecular pathological epidemiology of epigenetics: emerging integrative science to analyze environment, host, and disease. Mod Pathol 2013; 26:465-84. [PMID: 23307060 PMCID: PMC3637979 DOI: 10.1038/modpathol.2012.214] [Citation(s) in RCA: 166] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Epigenetics acts as an interface between environmental/exogenous factors, cellular responses, and pathological processes. Aberrant epigenetic signatures are a hallmark of complex multifactorial diseases (including neoplasms and malignancies such as leukemias, lymphomas, sarcomas, and breast, lung, prostate, liver, and colorectal cancers). Epigenetic signatures (DNA methylation, mRNA and microRNA expression, etc) may serve as biomarkers for risk stratification, early detection, and disease classification, as well as targets for therapy and chemoprevention. In particular, DNA methylation assays are widely applied to formalin-fixed, paraffin-embedded archival tissue specimens as clinical pathology tests. To better understand the interplay between etiological factors, cellular molecular characteristics, and disease evolution, the field of 'molecular pathological epidemiology (MPE)' has emerged as an interdisciplinary integration of 'molecular pathology' and 'epidemiology'. In contrast to traditional epidemiological research including genome-wide association studies (GWAS), MPE is founded on the unique disease principle, that is, each disease process results from unique profiles of exposomes, epigenomes, transcriptomes, proteomes, metabolomes, microbiomes, and interactomes in relation to the macroenvironment and tissue microenvironment. MPE may represent a logical evolution of GWAS, termed 'GWAS-MPE approach'. Although epigenome-wide association study attracts increasing attention, currently, it has a fundamental problem in that each cell within one individual has a unique, time-varying epigenome. Having a similar conceptual framework to systems biology, the holistic MPE approach enables us to link potential etiological factors to specific molecular pathology, and gain novel pathogenic insights on causality. The widespread application of epigenome (eg, methylome) analyses will enhance our understanding of disease heterogeneity, epigenotypes (CpG island methylator phenotype, LINE-1 (long interspersed nucleotide element-1; also called long interspersed nuclear element-1; long interspersed element-1; L1) hypomethylation, etc), and host-disease interactions. In this article, we illustrate increasing contribution of modern pathology to broader public health sciences, which attests pivotal roles of pathologists in the new integrated MPE science towards our ultimate goal of personalized medicine and prevention.
Collapse
Affiliation(s)
- Shuji Ogino
- Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA 02215, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Short-term single treatment of chemotherapy results in the enrichment of ovarian cancer stem cell-like cells leading to an increased tumor burden. Mol Cancer 2013; 12:24. [PMID: 23537295 PMCID: PMC3668985 DOI: 10.1186/1476-4598-12-24] [Citation(s) in RCA: 163] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Accepted: 03/19/2013] [Indexed: 01/03/2023] Open
Abstract
Over 80% of women diagnosed with advanced-stage ovarian cancer die as a result of disease recurrence due to failure of chemotherapy treatment. In this study, using two distinct ovarian cancer cell lines (epithelial OVCA 433 and mesenchymal HEY) we demonstrate enrichment in a population of cells with high expression of CSC markers at the protein and mRNA levels in response to cisplatin, paclitaxel and the combination of both. We also demonstrate a significant enhancement in the sphere forming abilities of ovarian cancer cells in response to chemotherapy drugs. The results of these in vitro findings are supported by in vivo mouse xenograft models in which intraperitoneal transplantation of cisplatin or paclitaxel-treated residual HEY cells generated significantly higher tumor burden compared to control untreated cells. Both the treated and untreated cells infiltrated the organs of the abdominal cavity. In addition, immunohistochemical studies on mouse tumors injected with cisplatin or paclitaxel treated residual cells displayed higher staining for the proliferative antigen Ki67, oncogeneic CA125, epithelial E-cadherin as well as cancer stem cell markers such as Oct4 and CD117, compared to mice injected with control untreated cells. These results suggest that a short-term single treatment of chemotherapy leaves residual cells that are enriched in CSC-like traits, resulting in an increased metastatic potential. The novel findings in this study are important in understanding the early molecular mechanisms by which chemoresistance and subsequent relapse may be triggered after the first line of chemotherapy treatment.
Collapse
|
39
|
|
40
|
Lu H, Yang X, Zhang Y, Lu R, Wang X. Epigenetic disorder may cause downregulation of HOXA10 in the eutopic endometrium of fertile women with endometriosis. Reprod Sci 2012; 20:78-84. [PMID: 22915150 DOI: 10.1177/1933719112451146] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
HOXA10 is an important gene for endometrial receptivity and plays a regulatory role in the adult female reproductive tract. It is regulated by epigenetic modulation in the CpG clusters of promoter in some cases. The aim of this study was to investigate HOXA10 expression and the epigenetic regulation in the eutopic endometrium of fertile women with endometriosis by quantitative real-time polymerase chain reaction (RT-PCR) and Western blot. The effect of 5-azacytidine (5-ac), a demethylation agent on HOXA10 expression was determined on endometrium stromal cells (ESCs) from these women with endometriosis. Results revealed that in normal endometrium (NE), HOXA10 messenger RNA (mRNA) and protein expression at the secretory phase were significantly higher than that at the proliferative phase. The HOXA10 mRNA and protein expression in the eutopic endometrium of endometriosis were significantly lower than in NE. The HOXA10 mRNA and protein levels in cultured stromal cells from endometriosis in vitro were significantly increased in a 5-ac treatment group compared with a nontreatment group. Our results indicated that the level of HOXA10 decrease in the eutopic endometrium of patients with endometriosis. Upregulation of HOXA10 in ESCs after treatment with 5-ac suggests that HOXA10 expression is controlled by methylation of the promoter. An epigenetic aberration is likely the main cause of endometriosis.
Collapse
Affiliation(s)
- Hong Lu
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | | | | | | | | |
Collapse
|
41
|
Abstract
Changes in gene expression that reset a cell program from a normal to a diseased state involve multiple genetic circuitries, creating a characteristic signature of gene expression that defines the cell's unique identity. Such signatures have been demonstrated to classify subtypes of breast cancers. Because DNA methylation is critical in programming gene expression, a change in methylation from a normal to diseased state should be similarly reflected in a signature of DNA methylation that involves multiple gene pathways. Whole-genome approaches have recently been used with different levels of success to delineate breast-cancer-specific DNA methylation signatures, and to test whether they can classify breast cancer and whether they could be associated with specific clinical outcomes. Recent work suggests that DNA methylation signatures will extend our ability to classify breast cancer and predict outcome beyond what is currently possible. DNA methylation is a robust biomarker, vastly more stable than RNA or proteins, and is therefore a promising target for the development of new approaches for diagnosis and prognosis of breast cancer and other diseases. Here, I review the scientific basis for using DNA methylation signatures in breast cancer classification and prognosis. I discuss the role of DNA methylation in normal gene regulation, the aberrations in DNA methylation in cancer, and candidate-gene and whole-genome approaches to classify breast cancer subtypes using DNA methylation markers.
Collapse
Affiliation(s)
- Moshe Szyf
- Department of Pharmacology and Therapeutics, Sackler Program in Epigenetics and Psychobiology, McGill University, 3,655 Sir William Osler Promenade, Montreal H3G1Y6, Canada
| |
Collapse
|