1
|
Tan C, Wu D, Yang X, Zhang S, Liu S, Yu B, Yu X, Xiu Y, Huang Y. Low expression of TOX predicts poor prognosis of patients with breast cancer in the real world: A retrospective study. Heliyon 2025; 11:e41180. [PMID: 39758401 PMCID: PMC11699305 DOI: 10.1016/j.heliyon.2024.e41180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 11/11/2024] [Accepted: 12/11/2024] [Indexed: 01/07/2025] Open
Abstract
Background TOX is a transcription factor that is implicated in the regulation of T cell exhaustion in tumors. TOX has been proven to have prognostic value in some malignant tumors. We aim to analyze the expression of TOX in breast cancer patients, and the association between TOX and prognostic significance in patients with breast cancer. Methods 313 breast cancer patients were enrolled into this study. The expression of TOX was determined by immunohistochemistry assay. Survival curves were performed by Kaplan-Meier and log-rank test. The potential independent factors were assessed by Cox regression analyses. Nomogram models, calibration curve, decision curve analyses were applied to analyze the clinical utility of predictive models. Results According to semi-quantitative scoring, 129 patients were classified into low group, and 184 patients were classified into high group. Patients with high expression of TOX had a longer survival than those with low expression of TOX (DFS: 71.70 vs. 64.05 months, χ2 = 11.6300, P = 0.00065; OS: 81.03 vs. 73.72 months, χ2 = 11.4200, P = 0.00073). Based on Cox regression analyses, multivariate analysis indicated that TOX was the potential prognostic factor for both DFS (HR: 0.412, 95 % CI: 0.248-0.684, P = 0.001) and OS (HR: 0.395, 95 % CI: 0.237-0.660, P < 0.0001). Calibration curve analysis showed that the predicted line was well-matched with baseline regarding postoperative 1-, 3-, and 5-year survival rate. Conclusions The expression of TOX is a potential prognostic factor, and can be a promising biomarker for predicting survival in breast cancer patients.
Collapse
Affiliation(s)
- Chunlei Tan
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, PR China
| | - Danping Wu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, PR China
| | - Xiaotian Yang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, PR China
| | - Shiyuan Zhang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, PR China
| | - Shuqiang Liu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, PR China
| | - Boqian Yu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, PR China
| | - Xiao Yu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, PR China
| | - Yuting Xiu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, PR China
| | - Yuanxi Huang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, PR China
| |
Collapse
|
2
|
Pal M, Das D, Pandey M. Understanding genetic variations associated with familial breast cancer. World J Surg Oncol 2024; 22:271. [PMID: 39390525 PMCID: PMC11465949 DOI: 10.1186/s12957-024-03553-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 10/02/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Breast cancer is the most frequent cancer among women. Genetics are the main risk factor for breast cancer. Statistics show that 15-25% of breast cancers are inherited among those with cancer-prone relatives. BRCA1, BRCA2, TP53, CDH1, PTEN, and STK11 are the most frequent genes for familial breast cancer, which occurs 80% of the time. In rare situations, moderate-penetrance gene mutations such CHEK2, BRIP1, ATM, and PALB2 contribute 2-3%. METHODS A search of the PubMed database was carried out spanning from 2005 to July 2024, yielding a total of 768 articles that delve into the realm of familial breast cancer, concerning genes and genetic syndromes. After exclusion 150 articles were included in the final review. RESULTS We report on a set of 20 familial breast cancer -associated genes into high, moderate, and low penetrance levels. Additionally, 10 genetic disorders were found to be linked with familial breast cancer. CONCLUSION Familial breast cancer has been linked to several genetic diseases and mutations, according to studies. Screening for genetic disorders is recommended by National Comprehensive Cancer Network recommendations. Evaluation of breast cancer candidate variations and risk loci may improve individual risk assessment. Only high- and moderate-risk gene variations have clinical guidelines, whereas low-risk gene variants require additional investigation. With increasing use of NGS technology, more linkage with rare genes is being discovered.
Collapse
Affiliation(s)
- Manjusha Pal
- Department of Surgical Oncology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Doutrina Das
- Department of Surgical Oncology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Manoj Pandey
- Department of Surgical Oncology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
3
|
Wang Z, Ding S, Zhang C, Zhan H, Li Y, Yan J, Jia Y, Wang X, Wang Y. Revealing the impact of TOX3 on osteoarthritis: insights from bioinformatics. Front Med (Lausanne) 2023; 10:1256654. [PMID: 38020130 PMCID: PMC10663247 DOI: 10.3389/fmed.2023.1256654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 10/19/2023] [Indexed: 12/01/2023] Open
Abstract
Osteoarthritis, a prevalent long-term condition of the joints, primarily impacts older individuals, resulting in discomfort, restrictions in mobility, and a decrease in overall well-being. Although Osteoarthritis is widely spread, there is a lack of successful interventions to stop the advancement of the condition. Numerous signaling pathways have been emphasized in recent research on Osteoarthritis, yet the diagnostic significance of numerous genes has not been investigated. To identify genes that were expressed differently in osteoarthritis, we utilized the Gene Expression Omnibus database. To identify marker genes, we built machine learning models including Least Absolute Shrinkage and Selection Operator and Random Forest. We categorized Osteoarthritis samples and performed immune cell infiltration analysis based on the expression patterns of these characteristic genes. Both the Least Absolute Shrinkage and Selection Operator and Random Forest models selected six marker genes (TOX3, ARG1, CST7, RERGL, COL11A1, NCRNA00185) out of a total of 17 differentially expressed genes. The osteoarthritis samples were categorized into two groups, namely a high expression group and a low expression group, based on the median levels of TOX3 expression. Comparative analysis of these groups identified 85 differentially expressed genes, showing notable enrichment in pathways related to lipid metabolism in the group with high expression. Analysis of immune cell infiltration revealed noticeable differences in immune profiles among the two groups. The group with high expression of TOX3 showed a notable increase in Mast cells and Type II IFN Response, whereas B cells, Cytolytic activity, Inflammation-promoting cells, NK cells, pDCs, T cell co-inhibition, Th1 cells, and Th2 cells were significantly decreased. We constructed a ceRNA network for TOX3, revealing 57 lncRNAs and 18 miRNAs involved in 57 lncRNA-miRNA interactions, and 18 miRNA-mRNA interactions with TOX3. Validation of TOX3 expression was confirmed using an external dataset (GSE29746), revealing a notable increase in Osteoarthritis samples. In conclusion, our study presents a comprehensive analysis identifying TOX3 as a potential feature gene in Osteoarthritis. The distinct immune profiles and involvement in fat metabolism pathways associated with TOX3 expression suggest its significance in Osteoarthritis pathogenesis. The study establishes a basis for comprehending the intricate correlation between characteristic genes and Osteoarthritis, as well as for the formulation of individualized therapeutic approaches.
Collapse
Affiliation(s)
- Zhengyan Wang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Shuang Ding
- Department of Orthopedics, The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | | | - Hongsheng Zhan
- Department of Orthopedics, Shuguang Hospital, Shanghai, China
| | - Yunfei Li
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Jing Yan
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yuyan Jia
- Department of Orthopedics, The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Xukai Wang
- Department of Orthopedics, The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Ying Wang
- Department of Orthopedics, The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
4
|
Zhang H, Fan F, Yu Y, Wang Z, Liu F, Dai Z, Zhang L, Liu Z, Cheng Q. Clinical characterization, genetic profiling, and immune infiltration of TOX in diffuse gliomas. J Transl Med 2020; 18:305. [PMID: 32762688 PMCID: PMC7409670 DOI: 10.1186/s12967-020-02460-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 07/26/2020] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Immunotherapies targeting glioblastoma (GBM) have led to significant improvements in patient outcomes. TOX is closely associated with the immune environment surrounding tumors, but its role in gliomas is not fully understood. METHODS Using data from The Cancer Genome Atlas (TCGA) and the Chinese Glioma Genome Atlas (CGGA), we analyzed the transcriptomes of 1691 WHO grade I-IV human glioma samples. The R language was used to perform most of the statistical analyses. Somatic mutations and somatic copy number variation (CNV) were analyzed using GISTIC 2.0. RESULTS TOX was down-regulated in malignant gliomas compared to low grade gliomas, and upregulated in the proneural and IDH mutant subtypes of GBM. TOXlow tumours are associated with the loss of PTEN and amplification of EGFR, while TOXhigh tumours harbor frequent mutations in IDH1 (91%). TOX was highly expressed in leading edge regions of tumours. Gene ontology and pathway analyses demonstrated that TOX was enriched in multiple immune related processes including lymphocyte migration in GBM. Finally, TOX had a negative association with the infiltration of several immune cell types in the tumour microenvironment. CONCLUSION TOX has the potential to be a new prognostic marker for GBM.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fan Fan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- Center for Medical Genetics and Hunan Provincial Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuanqiang Yu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zeyu Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fangkun Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ziyu Dai
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Liyang Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
- Clinical Diagnosis and Therapeutic Center of Glioma, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, People's Republic of China.
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
5
|
Akhtar M, Jamal T, Din JU, Hayat C, Rauf M, Ul Haq SM, Sher Khan R, Shah AA, Jamal M, Jalil F. An in silico approach to characterize nonsynonymous SNPs and regulatory SNPs in human TOX3 gene. J Genet 2019; 98:104. [PMID: 31819019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Cancer is one of the deadliest complex diseases having multigene nature where the role of single-nucleotide polymorphism (SNP) has been well explored in multiple genes. TOX high mobility group box family member 3 (TOX3) is one such gene, in which SNPs have been found to be associated with breast cancer. In this study, we have examined the potentially damaging nonsynonymous SNPs(nsSNPs) in TOX3 gene using in silico tools, namely PolyPhen2, SNP&GO, PhD-SNP and PROVEAN, which were further confirmed by I-Mutant, MutPred1.2 and ConSurf for their stability, functional and structural effects. nsSNPs rs368713418 (A266D), rs751141352 (P273S, P273T), rs200878352 (A275T) have been found to be the most deleterious that may have a vital role in breast cancer. Premature stop codon producing SNPs (Q527STOP), rs1259790811 (G495STOP), rs1294465822 (S395STOP) and rs1335372738 (G8STOP) were also found having prime importance in truncated and malfunctional protein formation. We also characterized regulatory SNPs for its potential effect on TOX3 gene regulation and found nine SNPs that may affect the gene regulation. Further, we have also designed 3D models using I-TASSER for the wild type and four mutant TOX3 proteins. Our study concludes that these SNPs can be of prime importance while studying breast cancer and other associated diseases as well. They are required to be studied in model organisms and cell cultures, and may have potential importance in personalized medicines and gene therapy.
Collapse
Affiliation(s)
- Mehran Akhtar
- Department of Biotechnology, Abdul Wali Khan University, Mardan 23200, Pakistan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
An in silico approach to characterize nonsynonymous SNPs and regulatory SNPs in human TOX3 gene. J Genet 2019. [DOI: 10.1007/s12041-019-1153-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
7
|
Al-Eitan LN, Rababa'h DM, Alghamdi MA, Khasawneh RH. Correlation between Candidate Single Nucleotide Variants and Several Clinicopathological Risk Factors Related to Breast Cancer in Jordanian Women: A Genotype-Phenotype Study. J Cancer 2019; 10:4647-4654. [PMID: 31528229 PMCID: PMC6746117 DOI: 10.7150/jca.33857] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 06/12/2019] [Indexed: 11/05/2022] Open
Abstract
This study aim to investigate the association of breast cancer risk and prognostic factors with single nucleotide variants of the BRCA1, BRCA2, DAPK1, MMP9, TOX3, and TP53 genes in Jordanian women. Blood samples were collected from 230 Jordanian breast cancer patients for use in DNA extraction followed by genotyping and subsequent statistical analysis. We found that two single nucleotide variants (SNVs) of the BRCA2 gene, namely rs1799944 and rs766173, were significantly associated with breastfeeding status. Likewise, the rs11141901 and rs1041326 SNVs of the DAPK1 gene were linked with co-morbidity (p-value = 0.002) and family history of BC (p-value = 0.015), while the rs1045042 SNV of the same gene was associated with both allergy (p-value = 0.001) and family history of BC (p-value = 0.02). Tumor differentiation was correlated with the DAPK1 SNVs rs11141901 (p-value = 0.041) and rs1041326 (p-value = 0.005). Additionally, the rs2250889 SNV of the MMP9 gene was significantly associated with HER2 status, whereas the TP53 SNVs rs12951053 and rs1042522 were associated with age at menarche (p-value = 0.043) and breastfeeding status (p-value = 0.013), respectively. In contrast, the TP53 SNV rs2287497 was significantly linked to age at first pregnancy (p-value = 0.001), smoking (p-value = 0.041), and axillary lymph node status (p-value = 6e-4). No such association was found for the BRCA1 and TOX3 SNVs. The current findings suggest significant associations between certain SNVs and breast cancer risk and prognosis in Jordanian women.
Collapse
Affiliation(s)
- Laith N Al-Eitan
- Department of Applied Biological Sciences, Jordan University of Science and Technology, Irbid 22110, Jordan.,Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Doaa M Rababa'h
- Department of Applied Biological Sciences, Jordan University of Science and Technology, Irbid 22110, Jordan
| | | | - Rame H Khasawneh
- Department of Hematopathology, King Hussein Medical Center (KHMC), Jordan Royal Medical Services (RMS), Amman 11118, Jordan
| |
Collapse
|
8
|
Mohamad Shah NS, Sulong S, Wan Sulaiman WA, Halim AS. Two novel genes TOX3 and COL21A1 in large extended Malay families with nonsyndromic cleft lip and/or palate. Mol Genet Genomic Med 2019; 7:e635. [PMID: 30924295 PMCID: PMC6503016 DOI: 10.1002/mgg3.635] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 01/09/2019] [Accepted: 02/11/2019] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Nonsyndromic cleft lip and/or palate is one of the most common human birth defects worldwide that affects the lip and/or palate. The incidence of clefts varies among populations through ethnic, race, or geographical differences. The focus on Malay nonsyndromic cleft lip and/or palate (NSCL/P) is because of a scarce report on genetic study in relation to this deformity in Malaysia. We are interested to discuss about the genes that are susceptible to cause orofacial cleft formation in the family. METHODS Genome-wide linkage analysis was carried out on eight large extended families of NSCL/P with the total of 91 individuals among Malay population using microarray platform. Based on linkage analyses findings, copy number variation (CNV) of LPHN2, SATB2, PVRL3, COL21A1, and TOX3 were identified in four large extended families that showed linkage evidence using quantitative polymerase chain reaction (qPCR) as for a validation purpose. Copy number calculated (CNC) for each genes were determined with Applied Biosystems CopyCallerTM Software v2.0. Normal CNC of the target sequence expected was set at two. RESULTS Genome-wide linkage analysis had discovered several genes including TOX3 and COL21A1 in four different loci 4p15.2-p16.1, 6p11.2-p12.3, 14q13-q21, and 16q12.1. There was significant decreased, p < 0.05 of SATB2, COL21A1, and TOX3 copy number in extended families compared to the normal controls. CONCLUSION Novel linkage evidence and significant low copy number of COL21A1 and TOX3 in NSCLP family was confirmed. These genes increased the risks toward NSCLP formation in that family traits.
Collapse
Affiliation(s)
- Nurul Syazana Mohamad Shah
- Reconstructive Science Unit, School of Medical SciencesUniversiti Sains MalaysiaKubang KerianKelantanMalaysia
| | - Sarina Sulong
- Human Genome Centre, School of Medical SciencesUniversiti Sains MalaysiaKubang KerianKelantanMalaysia
| | - Wan Azman Wan Sulaiman
- Reconstructive Science Unit, School of Medical SciencesUniversiti Sains MalaysiaKubang KerianKelantanMalaysia
| | | |
Collapse
|
9
|
Pan-cancer screen for mutations in non-coding elements with conservation and cancer specificity reveals correlations with expression and survival. NPJ Genom Med 2018; 3:1. [PMID: 29354286 PMCID: PMC5765157 DOI: 10.1038/s41525-017-0040-5] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 11/22/2017] [Accepted: 11/29/2017] [Indexed: 01/01/2023] Open
Abstract
Cancer develops by accumulation of somatic driver mutations, which impact cellular function. Mutations in non-coding regulatory regions can now be studied genome-wide and further characterized by correlation with gene expression and clinical outcome to identify driver candidates. Using a new two-stage procedure, called ncDriver, we first screened 507 ICGC whole-genomes from 10 cancer types for non-coding elements, in which mutations are both recurrent and have elevated conservation or cancer specificity. This identified 160 significant non-coding elements, including the TERT promoter, a well-known non-coding driver element, as well as elements associated with known cancer genes and regulatory genes (e.g., PAX5, TOX3, PCF11, MAPRE3). However, in some significant elements, mutations appear to stem from localized mutational processes rather than recurrent positive selection in some cases. To further characterize the driver potential of the identified elements and shortlist candidates, we identified elements where presence of mutations correlated significantly with expression levels (e.g., TERT and CDH10) and survival (e.g., CDH9 and CDH10) in an independent set of 505 TCGA whole-genome samples. In a larger pan-cancer set of 4128 TCGA exomes with expression profiling, we identified mutational correlation with expression for additional elements (e.g., near GATA3, CDC6, ZNF217, and CTCF transcription factor binding sites). Survival analysis further pointed to MIR122, a known marker of poor prognosis in liver cancer. In conclusion, the screen for significant mutation patterns coupled with correlative mutational analysis identified new individual driver candidates and suggest that some non-coding mutations recurrently affect expression and play a role in cancer development. Mutations in the “non-coding” part of the genome have been identified that could be involved in driving cancer development. Jakob Pedersen, Henrik Hornshøj and colleagues from Aarhus University Hospital in Denmark and MIT in the United States developed a two-stage procedure to identify elements that could be driving cancer development in the part of DNA that does not code for proteins. They conducted statisical analyses on catalogs of tumor genomes to identify recurrent mutations. They then evaluated how specific these mutations were to different cancer types, their predicted functional impact, and their association with gene expression and patient survival. The analyses identified mutations in the non-coding part of cancer genomes that could be driving tumor development, but further analyses on larger sample sets need to be conducted to validate the results, which could provide a basis for biomarker discovery and precision medical treatment.
Collapse
|
10
|
Raghu VK, Ge X, Chrysanthis PK, Benos PV. Integrated Theory- and Data-driven Feature Selection in Gene Expression Data Analysis. PROCEEDINGS. INTERNATIONAL CONFERENCE ON DATA ENGINEERING 2017; 2017:1525-1532. [PMID: 29422764 PMCID: PMC5799807 DOI: 10.1109/icde.2017.223] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The exponential growth of high dimensional biological data has led to a rapid increase in demand for automated approaches for knowledge production. Existing methods rely on two general approaches to address this challenge: 1) the Theory-driven approach, which utilizes prior accumulated knowledge, and 2) the Data-driven approach, which solely utilizes the data to deduce scientific knowledge. Both of these approaches alone suffer from bias toward past/present knowledge, as they fail to incorporate all of the current knowledge that is available to make new discoveries. In this paper, we show how an integrated method can effectively address the high dimensionality of big biological data, which is a major problem for pure data-driven analysis approaches. We realize our approach in a novel two-step analytical workflow that incorporates a new feature selection paradigm as the first step to handling high-throughput gene expression data analysis and that utilizes graphical causal modeling as the second step to handle the automatic extraction of causal relationships. Our results, on real-world clinical datasets from The Cancer Genome Atlas (TCGA), demonstrate that our method is capable of intelligently selecting genes for learning effective causal networks.
Collapse
Affiliation(s)
- Vineet K Raghu
- Department of Computer Science, University of Pittsburgh
| | - Xiaoyu Ge
- Department of Computer Science, University of Pittsburgh
| | | | - Panayiotis V Benos
- Department of Computer Science, University of Pittsburgh
- Department of Computational and Systems Biology, University of Pittsburgh
| |
Collapse
|
11
|
Al-Eitan LN, Jamous RI, Khasawneh RH. Candidate Gene Analysis of Breast Cancer in the Jordanian Population of Arab Descent: A Case-Control Study. Cancer Invest 2017; 35:256-270. [PMID: 28272917 DOI: 10.1080/07357907.2017.1289217] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
This study aimed to investigate whether there are specific polymorphisms within six genes (BRCA1, BRCA2, TP53, DAPK1, MMP9 promoter, and TOX3) that are associated with breast cancer among the Jordanian population. Sequenom MassARRAY system was used to genotype 17 single nucleotide polymorphisms (SNPs) within these genes in 230 Jordanian breast cancer patients and 225 healthy individuals. Three SNPs (MMP9 (rs6065912), TOX3 (rs1420546), and DAPK1 (rs11141901) were found to be significantly associated with an increased risk of breast cancer (p < .05). This study is the first to provide evidence that genetic variation in MMP9, TOX3, and DAPK1 genes contribute to the development of breast cancer in the Jordanian population.
Collapse
Affiliation(s)
- Laith N Al-Eitan
- a Department of Applied Biological Sciences , Jordan University of Science and Technology , Irbid , Jordan.,b Department of Biotechnology and Genetic Engineering , Jordan University of Science and Technology , Irbid , Jordan
| | - Reem I Jamous
- a Department of Applied Biological Sciences , Jordan University of Science and Technology , Irbid , Jordan.,b Department of Biotechnology and Genetic Engineering , Jordan University of Science and Technology , Irbid , Jordan
| | - Rame H Khasawneh
- c Department of Hematopathology, King Hussein Medical Center (KHMC) , Jordan Royal Medical Services (RMS) , Amman , Jordan
| |
Collapse
|
12
|
Han YJ, Zhang J, Zheng Y, Huo D, Olopade OI. Genetic and Epigenetic Regulation of TOX3 Expression in Breast Cancer. PLoS One 2016; 11:e0165559. [PMID: 27806084 PMCID: PMC5091860 DOI: 10.1371/journal.pone.0165559] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 10/13/2016] [Indexed: 01/09/2023] Open
Abstract
Genome wide association studies (GWAS) have identified low penetrance and high frequency single nucleotide polymorphisms (SNPs) that contribute to genetic susceptibility of breast cancer. The SNPs at 16q12, close to the TOX3 and CASC16 genes, represent one of the susceptibility loci identified by GWAS, showing strong evidence for breast cancer association across various populations. To examine molecular mechanisms of TOX3 regulation in breast cancer, we investigated both genetic and epigenetic factors using cell lines and datasets derived from primary breast tumors available through The Cancer Genome Atlas (TCGA). TOX3 expression is highly up-regulated in luminal subtype tumors compared to normal breast tissues or basal-like tumors. Expression quantitative trait loci (eQTL) analyses revealed significant associations of rs3803662 and rs4784227 genotypes with TOX3 expression in breast tumors. Bisulfite sequencing of four CpG islands in the TOX3 promoter showed a clear difference between luminal and basal-like cancer cell lines. 5-Aza-2’-deoxycytidine treatment of a basal-like cancer cell line increased expression of TOX3. TCGA dataset verified significantly lower levels of methylation of the promoter in luminal breast tumors with an inverse correlation between methylation and expression of TOX3. Methylation QTL (mQTL) analyses showed a weak or no correlation of rs3803662 or rs4784227 with TOX3 promoter methylation in breast tumors, indicating an independent relationship between the genetic and epigenetic events. These data suggest a complex system of TOX3 regulation in breast tumors, driven by germline variants and somatic epigenetic modifications in a subtype specific manner.
Collapse
Affiliation(s)
- Yoo-Jeong Han
- Center for Clinical Cancer Genetics and Global Health; and Section of Hematology and Oncology, Department of Medicine, University of Chicago, Chicago, IL 60637, United States of America
- * E-mail: (OIO); (YJH)
| | - Jing Zhang
- Center for Clinical Cancer Genetics and Global Health; and Section of Hematology and Oncology, Department of Medicine, University of Chicago, Chicago, IL 60637, United States of America
| | - Yonglan Zheng
- Center for Clinical Cancer Genetics and Global Health; and Section of Hematology and Oncology, Department of Medicine, University of Chicago, Chicago, IL 60637, United States of America
| | - Dezheng Huo
- Department of Public Health Sciences, University of Chicago, Chicago, IL 60637, United States of America
| | - Olufunmilayo I. Olopade
- Center for Clinical Cancer Genetics and Global Health; and Section of Hematology and Oncology, Department of Medicine, University of Chicago, Chicago, IL 60637, United States of America
- * E-mail: (OIO); (YJH)
| |
Collapse
|
13
|
Deb S, Lakhani SR, Ottini L, Fox SB. The cancer genetics and pathology of male breast cancer. Histopathology 2016; 68:110-8. [PMID: 26768033 DOI: 10.1111/his.12862] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Male breast cancer (MBC) is an uncommon and poorly understood disease. Recent molecular studies have shown important differences from female breast cancer which are likely to influence treatment strategies from the current female-based management towards a more tailored approach. Significantly more MBCs than female breast cancers arise with an underlying germline cancer predisposition, and display a vastly different penetrance compared with females. Furthermore, the genophenotypical association of basal-like cancer with BRCA1 present in female breast cancer is not observed in male breast cancer. Differences in somatic changes between male and female breast cancer have also been reported, with particular enrichment of PIK3CA mutations and a paucity of TP53 mutations. In general, chromosomal-based changes, in particular regions of gains, are seen more frequently in male than female breast cancer and methylation is seen less frequently. Clinically, several molecular subtypes with prognostic relevance have been described, including chromosomal complex high and methylation high groups, and subgroups with profiling signatures pertaining to epithelial mesenchymal transition and hormonal therapy insensitivity. As with female breast cancer, attention to male specific multicentre trials based on the individual characteristics are needed, together with establishment of reliable preclinical models to understand more clearly the pathogenesis of male breast cancer and improve the general poor outcome of this disease.
Collapse
Affiliation(s)
- Siddhartha Deb
- Department of Pathology, Peter MacCallum Cancer Centre, Department of Pathology and Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Vic., Australia.,Department of Anatomical Pathology, Royal Melbourne Hospital, Parkville, Melbourne, Vic., Australia
| | - Sunil R Lakhani
- Department of Anatomical Pathology, Pathology Queensland, University of Queensland, Brisbane, Qld, Australia.,Department of Molecular and Cellular Pathology, School of Medicine, University of Queensland, Brisbane, Qld, Australia.,The Royal Brisbane and Women's Hospital, University of Queensland Centre for Clinical Research, Brisbane, Qld, Australia
| | - Laura Ottini
- Department of Molecular Medicine, 'Sapienza' University of Rome, Rome, Italy
| | - Stephen B Fox
- Department of Pathology, Peter MacCallum Cancer Centre, Department of Pathology and Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Vic., Australia
| |
Collapse
|
14
|
Association of single nucleotide polymorphism rs3803662 with the risk of breast cancer. Sci Rep 2016; 6:29008. [PMID: 27350156 PMCID: PMC4924094 DOI: 10.1038/srep29008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 06/09/2016] [Indexed: 02/02/2023] Open
Abstract
Large scale association studies have identified the single nucleotide polymorphism rs3803662 associated with breast cancer risk. However, the sample size of most studies is too small. Here, we performed this meta-analysis to make the result more convincing. Relevant articles published up to 2016 were identified by searching the PubMed database. 13 studies, involving a total of 29405 participants, were included in the meta-analysis. Odds Ratios (ORs) with 95% confidence intervals (CIs) was calculated with random or fixed effects model. All data analyses were analyzed by Review Manger 5.3 software. In Caucasian subgroup: Dominant model (TT + CT vs CC): OR = 1.17 (1.06, 1.29), Recessive model (TT vs CT + CC): OR = 1.25 (1.13, 1.39) and Allele frequency (T vs C): OR = 1.15 (1.08, 1.22). The present meta-analysis suggests that rs3803662 polymorphism is significantly associated with breast cancer risk in Caucasian women, and we did not find the association in Asian women.
Collapse
|
15
|
Yu X, Li Z. TOX gene: a novel target for human cancer gene therapy. Am J Cancer Res 2015; 5:3516-3524. [PMID: 26885442 PMCID: PMC4731627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 06/26/2015] [Indexed: 06/05/2023] Open
Abstract
Thymocyte selection-associated high mobility group box factor (TOX) is a member of an evolutionarily conserved DNA-binding protein family and is expressed in several immune-relevant cell subsets. TOX encodes a nuclear protein of the high-mobility group box superfamily. It contains a DNA-binding domain, which allows it to regulate transcription by modifying local chromatin structure and modulating the formation of multi-protein complexes. Previous studies have shown that TOX play important roles in immune system. More recently, several studies have described TOX expression is frequently upregulated in diverse types of human tumors and the overregulation often associates with tumor progression. Moreover, TOXis involved in the control of cell apoptosis, growth, metastasis, DNA repair and so on. In this review, we provide an overview of current knowledge concerning the role of TOX in tumor development and progression biology function. To our knowledge, this is the first review about the role of thisnew oncogene in tumor development and progression.
Collapse
Affiliation(s)
- Xin Yu
- Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing 100042, China
| | - Zheng Li
- Department of Orthopaedic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| |
Collapse
|
16
|
Bawa P, Zackaria S, Verma M, Gupta S, Srivatsan R, Chaudhary B, Srinivasan S. Integrative Analysis of Normal Long Intergenic Non-Coding RNAs in Prostate Cancer. PLoS One 2015; 10:e0122143. [PMID: 25933431 PMCID: PMC4416928 DOI: 10.1371/journal.pone.0122143] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 02/10/2015] [Indexed: 02/03/2023] Open
Abstract
Recently, large numbers of normal human tissues have been profiled for non-coding RNAs and more than fourteen thousand long intergenic non-coding RNAs (lincRNAs) are found expressed in normal human tissues. The functional roles of these normal lincRNAs (nlincRNAs) in the regulation of protein coding genes in normal and disease biology are yet to be established. Here, we have profiled two RNA-seq datasets including cancer and matched non-neoplastic tissues from 12 individuals from diverse demography for both coding genes and nlincRNAs. We find 130 nlincRNAs significantly regulated in cancer, with 127 regulated in the same direction in the two datasets. Interestingly, according to Illumina Body Map, significant numbers of these nlincRNAs display baseline null expression in normal prostate tissues but are specific to other tissues such as thyroid, kidney, liver and testis. A number of the regulated nlincRNAs share loci with coding genes, which are either co-regulated or oppositely regulated in all cancer samples studied here. For example, in all cancer samples i) the nlincRNA, TCONS_00029157, and a neighboring tumor suppressor factor, SIK1, are both down regulated; ii) several thyroid-specific nlincRNAs in the neighborhood of the thyroid-specific gene TPO, are both up-regulated; and iii) the TCONS_00010581, an isoform of HEIH, is down-regulated while the neighboring EZH2 gene is up-regulated in cancer. Several nlincRNAs from a prostate cancer associated chromosomal locus, 8q24, are up-regulated in cancer along with other known prostate cancer associated genes including PCAT-1, PVT1, and PCAT-92. We observe that there is significant bias towards up-regulation of nlincRNAs with as high as 118 out of 127 up-regulated in cancer, even though regulation of coding genes is skewed towards down-regulation. Considering that all reported cancer associated lincRNAs (clincRNAs) are biased towards up-regulation, we conclude that this bias may be functionally relevant.
Collapse
Affiliation(s)
- Pushpinder Bawa
- IBAB, Institute of Bioinformatics and Applied Biotechnology, Bangalore, Karnataka, India
- Manipal University, Manipal, Karnataka, India
| | - Sajna Zackaria
- IBAB, Institute of Bioinformatics and Applied Biotechnology, Bangalore, Karnataka, India
| | - Mohit Verma
- IBAB, Institute of Bioinformatics and Applied Biotechnology, Bangalore, Karnataka, India
| | - Saurabh Gupta
- IBAB, Institute of Bioinformatics and Applied Biotechnology, Bangalore, Karnataka, India
| | - R Srivatsan
- IBAB, Institute of Bioinformatics and Applied Biotechnology, Bangalore, Karnataka, India
| | - Bibha Chaudhary
- IBAB, Institute of Bioinformatics and Applied Biotechnology, Bangalore, Karnataka, India
| | - Subhashini Srinivasan
- IBAB, Institute of Bioinformatics and Applied Biotechnology, Bangalore, Karnataka, India
- * E-mail:
| |
Collapse
|
17
|
Seksenyan A, Kadavallore A, Walts AE, de la Torre B, Berel D, Strom SP, Aliahmad P, Funari VA, Kaye J. TOX3 is expressed in mammary ER(+) epithelial cells and regulates ER target genes in luminal breast cancer. BMC Cancer 2015; 15:22. [PMID: 25632947 PMCID: PMC4324787 DOI: 10.1186/s12885-015-1018-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 01/13/2015] [Indexed: 01/13/2023] Open
Abstract
Background A breast cancer susceptibility locus has been mapped to the gene encoding TOX3. Little is known regarding the expression pattern or biological role of TOX3 in breast cancer or in the mammary gland. Here we analyzed TOX3 expression in murine and human mammary glands and in molecular subtypes of breast cancer, and assessed its ability to alter the biology of breast cancer cells. Methods We used a cell sorting strategy, followed by quantitative real-time PCR, to study TOX3 gene expression in the mouse mammary gland. To study the expression of this nuclear protein in human mammary glands and breast tumors, we generated a rabbit monoclonal antibody specific for human TOX3. In vitro studies were performed on MCF7, BT474 and MDA-MB-231 cell lines to study the effects of TOX3 modulation on gene expression in the context of breast cancer cells. Results We found TOX3 expression in estrogen receptor-positive mammary epithelial cells, including progenitor cells. A subset of breast tumors also highly expresses TOX3, with poor outcome associated with high expression of TOX3 in luminal B breast cancers. We also demonstrate the ability of TOX3 to alter gene expression in MCF7 luminal breast cancer cells, including cancer relevant genes TFF1 and CXCR4. Knockdown of TOX3 in a luminal B breast cancer cell line that highly expresses TOX3 is associated with slower growth. Surprisingly, TOX3 is also shown to regulate TFF1 in an estrogen-independent and tamoxifen-insensitive manner. Conclusions These results demonstrate that high expression of this protein likely plays a crucial role in breast cancer progression. This is in sharp contrast to previous studies that indicated breast cancer susceptibility is associated with lower expression of TOX3. Together, these results suggest two different roles for TOX3, one in the initiation of breast cancer, potentially related to expression of TOX3 in mammary epithelial cell progenitors, and another role for this nuclear protein in the progression of cancer. In addition, these results can begin to shed light on the reported association of TOX3 expression and breast cancer metastasis to the bone, and point to TOX3 as a novel regulator of estrogen receptor-mediated gene expression. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1018-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Akop Seksenyan
- Research Division of Immunology, Departments of Biomedical Sciences and Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Davis 5089, Los Angeles, 90048, CA, USA.
| | - Asha Kadavallore
- Research Division of Immunology, Departments of Biomedical Sciences and Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Davis 5089, Los Angeles, 90048, CA, USA.
| | - Ann E Walts
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | - Brian de la Torre
- Research Division of Immunology, Departments of Biomedical Sciences and Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Davis 5089, Los Angeles, 90048, CA, USA.
| | - Dror Berel
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA. .,Center for Applied Molecular Medicine, University of Southern California, Keck School of Medicine, Los Angeles, CA, USA.
| | - Samuel P Strom
- Genomics Core Facility, Cedars-Sinai Medical Center, Los Angeles, CA, USA. .,Department of Pathology and Laboratory Medicine, University of California Los Angeles David Geffen School of Medicine, Los Angeles, CA, USA.
| | - Parinaz Aliahmad
- Research Division of Immunology, Departments of Biomedical Sciences and Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Davis 5089, Los Angeles, 90048, CA, USA.
| | - Vincent A Funari
- Genomics Core Facility, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | - Jonathan Kaye
- Research Division of Immunology, Departments of Biomedical Sciences and Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Davis 5089, Los Angeles, 90048, CA, USA. .,Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA. .,Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| |
Collapse
|
18
|
Elbelt U, Trovato A, Kloth M, Gentz E, Finke R, Spranger J, Galas D, Weber S, Wolf C, König K, Arlt W, Büttner R, May P, Allolio B, Schneider JG. Molecular and clinical evidence for an ARMC5 tumor syndrome: concurrent inactivating germline and somatic mutations are associated with both primary macronodular adrenal hyperplasia and meningioma. J Clin Endocrinol Metab 2015; 100:E119-28. [PMID: 25279498 PMCID: PMC4283009 DOI: 10.1210/jc.2014-2648] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 09/19/2014] [Indexed: 12/26/2022]
Abstract
CONTEXT Primary macronodular adrenal hyperplasia (PMAH) is a rare cause of Cushing's syndrome, which may present in the context of different familial multitumor syndromes. Heterozygous inactivating germline mutations of armadillo repeat containing 5 (ARMC5) have very recently been described as cause for sporadic PMAH. Whether this genetic condition also causes familial PMAH in association with other neoplasias is unclear. OBJECTIVE The aim of the present study was to delineate the molecular cause in a large family with PMAH and other neoplasias. PATIENTS AND METHODS Whole-genome sequencing and comprehensive clinical and biochemical phenotyping was performed in members of a PMAH affected family. Nodules derived from adrenal surgery and pancreatic and meningeal tumor tissue were analyzed for accompanying somatic mutations in the identified target genes. RESULTS PMAH presenting either as overt or subclinical Cushing's syndrome was accompanied by a heterozygous germline mutation in ARMC5 (p.A110fs*9) located on chromosome 16. Analysis of tumor tissue showed different somatic ARMC5 mutations in adrenal nodules supporting a second hit hypothesis with inactivation of a tumor suppressor gene. A damaging somatic ARMC5 mutation was also found in a concomitant meningioma (p.R502fs) but not in a pancreatic tumor, suggesting biallelic inactivation of ARMC5 as causal also for the intracranial meningioma. CONCLUSIONS Our analysis further confirms inherited inactivating ARMC5 mutations as a cause of familial PMAH and suggests an additional role for the development of concomitant intracranial meningiomas.
Collapse
Affiliation(s)
| | - Alessia Trovato
- Department of Endocrinology, Diabetes, and Nutrition (U.E., A.T., J.S.), Department of Hepatology and Gastroenterology (E.G.), Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany; Institute of Pathology (M.K., K.K., R.B.), University of Cologne, 50937 Cologne, Germany; Praxisgemeinschaft an der Kaisereiche (R.F.), 12159 Berlin, Germany; Luxembourg Centre for Systems Biomedicine (D.G., C.W., P.M., J.G.S.), University of Luxembourg, 4362 Luxembourg, Luxembourg; Pacific Northwest Diabetes Research Institute (D.G.), Seattle, Washington 98122; Department of Internal Medicine II (S.W., C.W.), Saarland University Medical Center, 66421 Homburg/Saar, Germany; Centre for Endocrinology, Diabetes, and Metabolism (W.A.), School of Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, United Kingdom; Family Genomes Group (P.M.), Institute for Systems Biology, Seattle, Washington 98109; and Department of Internal Medicine I (B.A.), Endocrine and Diabetes Unit, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Michael Kloth
- Department of Endocrinology, Diabetes, and Nutrition (U.E., A.T., J.S.), Department of Hepatology and Gastroenterology (E.G.), Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany; Institute of Pathology (M.K., K.K., R.B.), University of Cologne, 50937 Cologne, Germany; Praxisgemeinschaft an der Kaisereiche (R.F.), 12159 Berlin, Germany; Luxembourg Centre for Systems Biomedicine (D.G., C.W., P.M., J.G.S.), University of Luxembourg, 4362 Luxembourg, Luxembourg; Pacific Northwest Diabetes Research Institute (D.G.), Seattle, Washington 98122; Department of Internal Medicine II (S.W., C.W.), Saarland University Medical Center, 66421 Homburg/Saar, Germany; Centre for Endocrinology, Diabetes, and Metabolism (W.A.), School of Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, United Kingdom; Family Genomes Group (P.M.), Institute for Systems Biology, Seattle, Washington 98109; and Department of Internal Medicine I (B.A.), Endocrine and Diabetes Unit, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Enno Gentz
- Department of Endocrinology, Diabetes, and Nutrition (U.E., A.T., J.S.), Department of Hepatology and Gastroenterology (E.G.), Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany; Institute of Pathology (M.K., K.K., R.B.), University of Cologne, 50937 Cologne, Germany; Praxisgemeinschaft an der Kaisereiche (R.F.), 12159 Berlin, Germany; Luxembourg Centre for Systems Biomedicine (D.G., C.W., P.M., J.G.S.), University of Luxembourg, 4362 Luxembourg, Luxembourg; Pacific Northwest Diabetes Research Institute (D.G.), Seattle, Washington 98122; Department of Internal Medicine II (S.W., C.W.), Saarland University Medical Center, 66421 Homburg/Saar, Germany; Centre for Endocrinology, Diabetes, and Metabolism (W.A.), School of Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, United Kingdom; Family Genomes Group (P.M.), Institute for Systems Biology, Seattle, Washington 98109; and Department of Internal Medicine I (B.A.), Endocrine and Diabetes Unit, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Reinhard Finke
- Department of Endocrinology, Diabetes, and Nutrition (U.E., A.T., J.S.), Department of Hepatology and Gastroenterology (E.G.), Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany; Institute of Pathology (M.K., K.K., R.B.), University of Cologne, 50937 Cologne, Germany; Praxisgemeinschaft an der Kaisereiche (R.F.), 12159 Berlin, Germany; Luxembourg Centre for Systems Biomedicine (D.G., C.W., P.M., J.G.S.), University of Luxembourg, 4362 Luxembourg, Luxembourg; Pacific Northwest Diabetes Research Institute (D.G.), Seattle, Washington 98122; Department of Internal Medicine II (S.W., C.W.), Saarland University Medical Center, 66421 Homburg/Saar, Germany; Centre for Endocrinology, Diabetes, and Metabolism (W.A.), School of Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, United Kingdom; Family Genomes Group (P.M.), Institute for Systems Biology, Seattle, Washington 98109; and Department of Internal Medicine I (B.A.), Endocrine and Diabetes Unit, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Joachim Spranger
- Department of Endocrinology, Diabetes, and Nutrition (U.E., A.T., J.S.), Department of Hepatology and Gastroenterology (E.G.), Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany; Institute of Pathology (M.K., K.K., R.B.), University of Cologne, 50937 Cologne, Germany; Praxisgemeinschaft an der Kaisereiche (R.F.), 12159 Berlin, Germany; Luxembourg Centre for Systems Biomedicine (D.G., C.W., P.M., J.G.S.), University of Luxembourg, 4362 Luxembourg, Luxembourg; Pacific Northwest Diabetes Research Institute (D.G.), Seattle, Washington 98122; Department of Internal Medicine II (S.W., C.W.), Saarland University Medical Center, 66421 Homburg/Saar, Germany; Centre for Endocrinology, Diabetes, and Metabolism (W.A.), School of Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, United Kingdom; Family Genomes Group (P.M.), Institute for Systems Biology, Seattle, Washington 98109; and Department of Internal Medicine I (B.A.), Endocrine and Diabetes Unit, University Hospital Würzburg, 97080 Würzburg, Germany
| | - David Galas
- Department of Endocrinology, Diabetes, and Nutrition (U.E., A.T., J.S.), Department of Hepatology and Gastroenterology (E.G.), Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany; Institute of Pathology (M.K., K.K., R.B.), University of Cologne, 50937 Cologne, Germany; Praxisgemeinschaft an der Kaisereiche (R.F.), 12159 Berlin, Germany; Luxembourg Centre for Systems Biomedicine (D.G., C.W., P.M., J.G.S.), University of Luxembourg, 4362 Luxembourg, Luxembourg; Pacific Northwest Diabetes Research Institute (D.G.), Seattle, Washington 98122; Department of Internal Medicine II (S.W., C.W.), Saarland University Medical Center, 66421 Homburg/Saar, Germany; Centre for Endocrinology, Diabetes, and Metabolism (W.A.), School of Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, United Kingdom; Family Genomes Group (P.M.), Institute for Systems Biology, Seattle, Washington 98109; and Department of Internal Medicine I (B.A.), Endocrine and Diabetes Unit, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Susanne Weber
- Department of Endocrinology, Diabetes, and Nutrition (U.E., A.T., J.S.), Department of Hepatology and Gastroenterology (E.G.), Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany; Institute of Pathology (M.K., K.K., R.B.), University of Cologne, 50937 Cologne, Germany; Praxisgemeinschaft an der Kaisereiche (R.F.), 12159 Berlin, Germany; Luxembourg Centre for Systems Biomedicine (D.G., C.W., P.M., J.G.S.), University of Luxembourg, 4362 Luxembourg, Luxembourg; Pacific Northwest Diabetes Research Institute (D.G.), Seattle, Washington 98122; Department of Internal Medicine II (S.W., C.W.), Saarland University Medical Center, 66421 Homburg/Saar, Germany; Centre for Endocrinology, Diabetes, and Metabolism (W.A.), School of Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, United Kingdom; Family Genomes Group (P.M.), Institute for Systems Biology, Seattle, Washington 98109; and Department of Internal Medicine I (B.A.), Endocrine and Diabetes Unit, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Cristina Wolf
- Department of Endocrinology, Diabetes, and Nutrition (U.E., A.T., J.S.), Department of Hepatology and Gastroenterology (E.G.), Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany; Institute of Pathology (M.K., K.K., R.B.), University of Cologne, 50937 Cologne, Germany; Praxisgemeinschaft an der Kaisereiche (R.F.), 12159 Berlin, Germany; Luxembourg Centre for Systems Biomedicine (D.G., C.W., P.M., J.G.S.), University of Luxembourg, 4362 Luxembourg, Luxembourg; Pacific Northwest Diabetes Research Institute (D.G.), Seattle, Washington 98122; Department of Internal Medicine II (S.W., C.W.), Saarland University Medical Center, 66421 Homburg/Saar, Germany; Centre for Endocrinology, Diabetes, and Metabolism (W.A.), School of Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, United Kingdom; Family Genomes Group (P.M.), Institute for Systems Biology, Seattle, Washington 98109; and Department of Internal Medicine I (B.A.), Endocrine and Diabetes Unit, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Katharina König
- Department of Endocrinology, Diabetes, and Nutrition (U.E., A.T., J.S.), Department of Hepatology and Gastroenterology (E.G.), Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany; Institute of Pathology (M.K., K.K., R.B.), University of Cologne, 50937 Cologne, Germany; Praxisgemeinschaft an der Kaisereiche (R.F.), 12159 Berlin, Germany; Luxembourg Centre for Systems Biomedicine (D.G., C.W., P.M., J.G.S.), University of Luxembourg, 4362 Luxembourg, Luxembourg; Pacific Northwest Diabetes Research Institute (D.G.), Seattle, Washington 98122; Department of Internal Medicine II (S.W., C.W.), Saarland University Medical Center, 66421 Homburg/Saar, Germany; Centre for Endocrinology, Diabetes, and Metabolism (W.A.), School of Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, United Kingdom; Family Genomes Group (P.M.), Institute for Systems Biology, Seattle, Washington 98109; and Department of Internal Medicine I (B.A.), Endocrine and Diabetes Unit, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Wiebke Arlt
- Department of Endocrinology, Diabetes, and Nutrition (U.E., A.T., J.S.), Department of Hepatology and Gastroenterology (E.G.), Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany; Institute of Pathology (M.K., K.K., R.B.), University of Cologne, 50937 Cologne, Germany; Praxisgemeinschaft an der Kaisereiche (R.F.), 12159 Berlin, Germany; Luxembourg Centre for Systems Biomedicine (D.G., C.W., P.M., J.G.S.), University of Luxembourg, 4362 Luxembourg, Luxembourg; Pacific Northwest Diabetes Research Institute (D.G.), Seattle, Washington 98122; Department of Internal Medicine II (S.W., C.W.), Saarland University Medical Center, 66421 Homburg/Saar, Germany; Centre for Endocrinology, Diabetes, and Metabolism (W.A.), School of Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, United Kingdom; Family Genomes Group (P.M.), Institute for Systems Biology, Seattle, Washington 98109; and Department of Internal Medicine I (B.A.), Endocrine and Diabetes Unit, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Reinhard Büttner
- Department of Endocrinology, Diabetes, and Nutrition (U.E., A.T., J.S.), Department of Hepatology and Gastroenterology (E.G.), Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany; Institute of Pathology (M.K., K.K., R.B.), University of Cologne, 50937 Cologne, Germany; Praxisgemeinschaft an der Kaisereiche (R.F.), 12159 Berlin, Germany; Luxembourg Centre for Systems Biomedicine (D.G., C.W., P.M., J.G.S.), University of Luxembourg, 4362 Luxembourg, Luxembourg; Pacific Northwest Diabetes Research Institute (D.G.), Seattle, Washington 98122; Department of Internal Medicine II (S.W., C.W.), Saarland University Medical Center, 66421 Homburg/Saar, Germany; Centre for Endocrinology, Diabetes, and Metabolism (W.A.), School of Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, United Kingdom; Family Genomes Group (P.M.), Institute for Systems Biology, Seattle, Washington 98109; and Department of Internal Medicine I (B.A.), Endocrine and Diabetes Unit, University Hospital Würzburg, 97080 Würzburg, Germany
| | | | | | | |
Collapse
|
19
|
Associations of two common genetic variants with breast cancer risk in a chinese population: a stratified interaction analysis. PLoS One 2014; 9:e115707. [PMID: 25531440 PMCID: PMC4274096 DOI: 10.1371/journal.pone.0115707] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 11/26/2014] [Indexed: 12/31/2022] Open
Abstract
Recent genome-wide association studies (GWAS) have identified a series of new genetic susceptibility loci for breast cancer (BC). However, the correlations between these variants and breast cancer are still not clear. In order to explore the role of breast cancer susceptibility variants in a Southeast Chinese population, we genotyped two common SNPs at chromosome 6q25 (rs2046210) and in TOX3 (rs4784227) in a case-control study with a total of 702 breast cancer cases and 794 healthy-controls. In addition, we also evaluated the multiple interactions among genetic variants, risk factors, and tumor subtypes. Associations of genotypes with breast cancer risk was evaluated using multivariate logistic regression to estimate odds ratios (OR) and their 95% confidence intervals (95% CI). The results indicated that both polymorphisms were significantly associated with the risk of breast cancer, with per allele OR = 1.35, (95%CI = 1.17-1.57) for rs2046210 and per allele OR = 1.24 (95%CI = 1.06-1.45) for rs4784227. Furthermore, in subgroup stratified analyses, we observed that the T allele of rs4784227 was significantly associated with elevated OR among postmenopausal populations (OR = 1.44, 95%CI 1.11-1.87) but not in premenopausal populations, with the heterogeneity P value of P = 0.064. These findings suggest that the genetic variants at chromosome 6q25 and in the TOX3 gene may play important roles in breast cancer development in a Chinese population and the underlying biological mechanisms need to be further elucidated.
Collapse
|
20
|
Garrisi VM, Strippoli S, De Summa S, Pinto R, Perrone A, Guida G, Azzariti A, Guida M, Stefania T. Proteomic profile and in silico analysis in metastatic melanoma with and without BRAF mutation. PLoS One 2014; 9:e112025. [PMID: 25437182 PMCID: PMC4249853 DOI: 10.1371/journal.pone.0112025] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 10/11/2014] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION Selective inhibitors of BRAF, vemurafenib and dabrafenib are the standard of care for metastatic melanoma patients with BRAF V600, while chemotherapy continued to be widely used in BRAF wild type patients. MATERIALS AND METHODS In order to discover novel candidate biomarkers predictive to treatment, serum of 39 metastatic melanoma vemurafenib (n = 19) or chemotherapy (n = 20) treated patients at baseline, at disease control and at progression, were analyzed using SELDI-TOF technology. In silico analysis was used to identify more significant peaks. RESULTS In patients with different BRAF status, we found 5 peptides significantly deregulated, with the down-regulation of the m/z 9176 peak strongly associated with BRAF mutation. At baseline as predictive biomarkers we identified 2 peptides - m/z 6411, 4075 - as significantly up-regulated in responders to chemotherapy and 4 peaks - m/z 5900, 12544, 49124 and 11724 - significantly up-regulated in longer vs shorter responders to vemurafenib. After response, 3 peptides (m/z 4658, 18639, and 9307) resulted significantly down regulated while 3 peptides m/z 9292, 7765 and 9176 appeared up-regulated respectively in chemotherapy and vemurafenib responder patients. In vemurafenib treated patients, 16 peaks appeared deregulated at progression compared to baseline time. In silico analysis identified proteins involved in invasiveness (SLAIN1) and resistance (ABCC12) as well as in the pathway of detoxification (NQO1) and apoptosis (RBM10, TOX3, MTEFD1, TSPO2). Proteins associated with the modulation of neuronal plasticity (RIN1) and regulatory activity factors of gene transcription (KLF17, ZBTB44) were also highlighted. CONCLUSION Our exploratory study highlighted some factors that deserve to be further investigated in order to provide a framework for improving melanoma treatment management through the development of biomarkers which could act as the strongest surrogates of the key biological events in stage IV melanoma.
Collapse
Affiliation(s)
- Vito Michele Garrisi
- National Cancer Research Centre, Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Sabino Strippoli
- National Cancer Research Centre, Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Simona De Summa
- National Cancer Research Centre, Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Rosamaria Pinto
- National Cancer Research Centre, Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Antonella Perrone
- National Cancer Research Centre, Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Gabriella Guida
- Dept. of Basic Medical Sciences, Faculty of Medicine and Surgery, School of Medicine, University of Bari, Bari, Italy
| | - Amalia Azzariti
- National Cancer Research Centre, Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Michele Guida
- National Cancer Research Centre, Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Tommasi Stefania
- National Cancer Research Centre, Istituto Tumori “Giovanni Paolo II”, Bari, Italy
- * E-mail:
| |
Collapse
|
21
|
Cui Y, Zhao S, Zhao H, Lv Y, Yu M, Wang Y, Chen ZJ. Mutational analysis of TOX3 in Chinese Han women with polycystic ovary syndrome. Reprod Biomed Online 2014; 29:752-5. [PMID: 25311971 DOI: 10.1016/j.rbmo.2014.08.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2014] [Revised: 07/25/2014] [Accepted: 08/12/2014] [Indexed: 10/24/2022]
Abstract
A previous genome-wide association study of polycystic ovary syndrome (PCOS) identified several susceptibility loci. TOX3 is the nearest gene to signal rs4784165. In the present study, all exons and exon-intron boundaries of TOX3 were amplified and sequenced in 200 Chinese women with PCOS. A 3-bp nucleotide deletion of CAG repeat and two known single nucleotide polymorphisms were identified. No plausible pathogenic mutations were detected. The results suggest that mutations in TOX3 are not common in Chinese Han women with PCOS.
Collapse
Affiliation(s)
- Yuqian Cui
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200127, China; Center for Reproductive Medicine, Provincial Hospital Affiliated to Shandong University, 324 Jingwu Road, Jinan 250021, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, 324 Jingwu Road, Jinan 250021, China; The Key Laboratory for Reproductive Endocrinology of Ministry of Education, 324 Jingwu Road, Jinan 250021, China; Shandong Provincial Key Laboratory of Reproductive Medicine, 324 Jingwu Road, Jinan 250021, China; Center for Reproductive Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan 250012, China
| | - Shigang Zhao
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200127, China; Center for Reproductive Medicine, Provincial Hospital Affiliated to Shandong University, 324 Jingwu Road, Jinan 250021, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, 324 Jingwu Road, Jinan 250021, China; The Key Laboratory for Reproductive Endocrinology of Ministry of Education, 324 Jingwu Road, Jinan 250021, China; Shandong Provincial Key Laboratory of Reproductive Medicine, 324 Jingwu Road, Jinan 250021, China
| | - Han Zhao
- Center for Reproductive Medicine, Provincial Hospital Affiliated to Shandong University, 324 Jingwu Road, Jinan 250021, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, 324 Jingwu Road, Jinan 250021, China; The Key Laboratory for Reproductive Endocrinology of Ministry of Education, 324 Jingwu Road, Jinan 250021, China; Shandong Provincial Key Laboratory of Reproductive Medicine, 324 Jingwu Road, Jinan 250021, China
| | - Yue Lv
- Center for Reproductive Medicine, Provincial Hospital Affiliated to Shandong University, 324 Jingwu Road, Jinan 250021, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, 324 Jingwu Road, Jinan 250021, China; The Key Laboratory for Reproductive Endocrinology of Ministry of Education, 324 Jingwu Road, Jinan 250021, China; Shandong Provincial Key Laboratory of Reproductive Medicine, 324 Jingwu Road, Jinan 250021, China
| | - Mengru Yu
- Center for Reproductive Medicine, Provincial Hospital Affiliated to Shandong University, 324 Jingwu Road, Jinan 250021, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, 324 Jingwu Road, Jinan 250021, China; The Key Laboratory for Reproductive Endocrinology of Ministry of Education, 324 Jingwu Road, Jinan 250021, China; Shandong Provincial Key Laboratory of Reproductive Medicine, 324 Jingwu Road, Jinan 250021, China
| | - Yu Wang
- Institute of Obstetrics and Gynecologic Oncology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200127, China; Center for Reproductive Medicine, Provincial Hospital Affiliated to Shandong University, 324 Jingwu Road, Jinan 250021, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, 324 Jingwu Road, Jinan 250021, China; The Key Laboratory for Reproductive Endocrinology of Ministry of Education, 324 Jingwu Road, Jinan 250021, China; Shandong Provincial Key Laboratory of Reproductive Medicine, 324 Jingwu Road, Jinan 250021, China.
| |
Collapse
|
22
|
Mizoo T, Taira N, Nishiyama K, Nogami T, Iwamoto T, Motoki T, Shien T, Matsuoka J, Doihara H, Ishihara S, Kawai H, Kawasaki K, Ishibe Y, Ogasawara Y, Komoike Y, Miyoshi S. Effects of lifestyle and single nucleotide polymorphisms on breast cancer risk: a case-control study in Japanese women. BMC Cancer 2013; 13:565. [PMID: 24289300 PMCID: PMC3879029 DOI: 10.1186/1471-2407-13-565] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 11/18/2013] [Indexed: 01/06/2023] Open
Abstract
Background Lifestyle factors, including food and nutrition, physical activity, body composition and reproductive factors, and single nucleotide polymorphisms (SNPs) are associated with breast cancer risk, but few studies of these factors have been performed in the Japanese population. Thus, the goals of this study were to validate the association between reported SNPs and breast cancer risk in the Japanese population and to evaluate the effects of SNP genotypes and lifestyle factors on breast cancer risk. Methods A case–control study in 472 patients and 464 controls was conducted from December 2010 to November 2011. Lifestyle was examined using a self-administered questionnaire. We analyzed 16 breast cancer-associated SNPs based on previous GWAS or candidate-gene association studies. Age or multivariate-adjusted odds ratios (OR) and 95% confidence intervals (95% CI) were estimated from logistic regression analyses. Results High BMI and current or former smoking were significantly associated with an increased breast cancer risk, while intake of meat, mushrooms, yellow and green vegetables, coffee, and green tea, current leisure-time exercise, and education were significantly associated with a decreased risk. Three SNPs were significantly associated with a breast cancer risk in multivariate analysis: rs2046210 (per allele OR = 1.37 [95% CI: 1.11-1.70]), rs3757318 (OR = 1.33[1.05-1.69]), and rs3803662 (OR = 1.28 [1.07-1.55]). In 2046210 risk allele carriers, leisure-time exercise was associated with a significantly decreased risk for breast cancer, whereas current smoking and high BMI were associated with a significantly decreased risk in non-risk allele carriers. Conclusion In Japanese women, rs2046210 and 3757318 located near the ESR1 gene are associated with a risk of breast cancer, as in other Asian women. However, our findings suggest that exercise can decrease this risk in allele carriers.
Collapse
Affiliation(s)
| | - Naruto Taira
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama-city, Okayama 700-8558, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|