1
|
Chen Y, Liao Y, Jia X, Lin H, Miao L, Yuan L, Huang X, Zhou J, Bian J, Zhuo Z. Association of ERCC1 and XPF polymorphisms with pediatric glioma susceptibility. Chin Med J (Engl) 2023; 136:746-748. [PMID: 36070459 PMCID: PMC10129197 DOI: 10.1097/cm9.0000000000002126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Indexed: 11/26/2022] Open
Affiliation(s)
- Yongping Chen
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
| | - Yuxiang Liao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Xingyu Jia
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
- School of Medicine, Jinan University, Guangzhou, Guangdong 510632, China
| | - Huiran Lin
- Faculty of Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Lei Miao
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
| | - Li Yuan
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
| | - Xiaokai Huang
- Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Jingying Zhou
- Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Jun Bian
- Department of General Surgery, Xi’an Children's Hospital, Xi’an Jiaotong University Affiliated Children's Hospital, Xi’an, Shaanxi 710003, China
| | - Zhenjian Zhuo
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
- Laboratory Animal Center, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, Guangdong 518055, China
| |
Collapse
|
2
|
Salari N, Rasoulpoor S, Shabani S, Mansouri K, Bokaee S, Fatahian R, Farshchian N, Mohammadi M, Hosseinian-Far M. ERCC2 rs13181 Polymorphism Association with Glioma Risk: an Update Meta-Analysis. Indian J Surg Oncol 2023; 14:60-68. [PMID: 36891435 PMCID: PMC9986186 DOI: 10.1007/s13193-022-01623-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 08/10/2022] [Indexed: 10/15/2022] Open
Abstract
Glioma is the most common type of primary brain tumour which accounts for about 30% of all brain and central nervous system tumours, and approximately 70% of adult malignant brain tumours. Numerous studies have been performed to assess the relationship between ERCC2 rs13181 polymorphism and the risk of glioma development, yet these findings of these studies are often inconsistent and contradictory. Therefore, the aim of this study is to conduct a systematic review and meta-analysis to assess the role of ERCC2 rs13181 in glioma developing. In this work, we have conducted a systematic review and meta-analysis. In order to collect the results of relevant studies on the association of ERCC2 rs13181 gene polymorphism with glioma, we initially searched the Scopus, Embase, Web of Science (WoS), PubMed, and ScienceDirect databases, without a lower time limit, and until June 2020. In order to analyse the eligible studies, the random effects model was used and the heterogeneity of the studies was investigated with the I 2 index. Data analysis was performed within the Comprehensive Meta-Analysis software (version 2). The total number of studies that focused on patients with glioma was 10. The odds ratio of GG vs TT genotype in patients with glioma based on meta-analysis was 1.08 (0.85-1.37: 95% confidence interval), which indicates the increasing effect of GG vs TT genotype by 0.08. The odds ratio of GG + TG vs TT genotype in patients with glioma was 1.22 (1.38-1.7: 95% confidence interval) based on meta-analysis, which indicates the increasing effect of GG + TG vs TT genotype as 0.22. The odds ratio of TG vs TT genotype in patients with glioma was 1.2 (0.38-1.4: 95% confidence interval), which shows the increasing effect of TG vs TT genotype by 0.2. The odds ratio of G vs T genotype in patients with glioma based on the meta-analysis was 1.15 (1.26-1.4: 95% confidence interval), which indicates the increasing effect of G vs T genotype by 0.15. The odds ratio of GG vs TG + TT genotype in patients with glioma based on meta-analysis was 1.22 (1.33-1.45: 95% confidence interval), which indicates the increasing effect of GG vs TG + TT genotype by 0.22. The results of this systematic review and meta-analysis show that ERCC2 rs13181 polymorphism and its genotypes are an important risk factor for genetic susceptibility to glioma tumour.
Collapse
Affiliation(s)
- Nader Salari
- Department of Biostatistics, School of Health, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Shna Rasoulpoor
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Shervin Shabani
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Kamran Mansouri
- Medical Biology Research Centre, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Shadi Bokaee
- Faculty of Health and Life Sciences, School of Life Sciences, Coventry University, Coventry, UK
| | - Reza Fatahian
- Department of Neurosurgery, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Negin Farshchian
- Department of Radiation Oncology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Masoud Mohammadi
- Cellular and Molecular Research Center, Gerash University of Medical Sciences, Gerash, Iran
| | - Melika Hosseinian-Far
- Department of Food Science & Technology, Faculty of Agriculture, Ferdowsi University of Mashhad (FUM), Mashhad, Iran
| |
Collapse
|
3
|
Association between XRCC3 p.Thr241Met polymorphism and risk of glioma: A systematic review and meta-analysis. PLoS One 2022; 17:e0276313. [PMID: 36264998 PMCID: PMC9584405 DOI: 10.1371/journal.pone.0276313] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 10/04/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The XRCC3 p.Thr241Met (rs861539) polymorphism has been extensively studied for its association with glioma risk, but results remain conflicting. Therefore, we performed a systematic review and meta-analysis to resolve this inconsistency. METHODS Studies published up to June 10, 2022, were searched in PubMed, Web of Science, Scopus, VIP, Wanfang, and China National Knowledge Infrastructure databases and screened for eligibility. Then, the combined odds ratio (OR) of the included studies was estimated based on five genetic models, i.e., homozygous (Met/Met vs. Thr/Thr), heterozygous (Thr/Met vs. Thr/Thr), dominant (Thr/Met + Met/Met vs. Thr/Thr), recessive (Met/Met vs. Thr/Thr + Thr/Met) and allele (Met vs. Thr). The study protocol was preregistered at PROSPERO (registration number: CRD42021235704). RESULTS Overall, our meta-analysis of 14 eligible studies involving 12,905 subjects showed that the p.Thr241Met polymorphism was significantly associated with increased glioma risk in both homozygous and recessive models (homozygous, OR = 1.381, 95% CI = 1.081-1.764, P = 0.010; recessive, OR = 1.305, 95% CI = 1.140-1.493, P<0.001). Subgroup analyses by ethnicity also revealed a statistically significant association under the two aforementioned genetic models, but only in the Asian population and not in Caucasians (P>0.05). CONCLUSION We demonstrated that the XRCC3 p.Thr241Met polymorphism is associated with an increased risk of glioma only in the homozygous and recessive models.
Collapse
|
4
|
Association between ERCC1 Gene Polymorphism (rs11615) and Colorectal Cancer Susceptibility: A Meta-Analysis of Medical Image Fusion and Safety Applications. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:9988513. [PMID: 36277013 PMCID: PMC9586779 DOI: 10.1155/2022/9988513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/19/2022] [Accepted: 09/24/2022] [Indexed: 11/29/2022]
Abstract
Colorectal cancer (CRC) is a malignant tumor of the colorectal mucosa epithelial tissue transformed. The fusion of data for medical imaging has become a central issue in such biomedical applications as image-guided surgery and radiotherapy. Currently, CRC has been one of the most threatening tumors affecting people's health worldwide. The excision repair cross-complementation group 1 (ERCC1) is a key enzyme for nucleotide excision repair (NER). Emerging epidemiological studies have indicated that the presence of colorectal cancer (CRC) may be relevant to the ERCC1 rs11615 genetic polymorphism. However, the results of ERCC1 rs11615 on CRC in these studies are controversial. We searched PubMed, Web of Science, Embase, CNKI, and CBM databases for the effects of ERCC1 rs11615 variant on CRC development. There was no meta-analysis focused on the diagnosis of colorectal cancer with ERCC1 rs11615 variant. We creatively carried out a meta-analysis of nine case-control studies and used Stata (version 12.0) software to integrate the pooled odds ratios (ORs) corresponding to a 95% confidence interval (CI) of overall and subgroup analysis. Our results suggest that a significant correlation was observed between rs11615 and the susceptibility of CRC OR 95% CI = 1.13 (1.04-1.23) under an allele genetic model and OR 95% CI = 1.14 (1.01-1.30) under a dominant genetic model for overall CRC. Significant statistical difference was also noted in Asians rather than Caucasians based on the ethnicity subgroups. These results suggested that there is a certain association between rs11615 and the susceptibility of colorectal cancer in the Asian populations.
Collapse
|
5
|
MGMT in glial carcinogenesis. Roles from prevention to treatment. Eur J Cancer Prev 2022; 31:568-576. [DOI: 10.1097/cej.0000000000000746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
6
|
Chen Y, Liao Y, Yuan L, Huang X, Ruan J, Lin H, Miao L, Zhuo Z. Genetic variants in XPD gene and glioma susceptibility in Chinese children: A multicenter case-control study. CANCER INNOVATION 2022; 1:70-79. [PMID: 38089451 PMCID: PMC10686151 DOI: 10.1002/cai2.6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/13/2022] [Accepted: 04/19/2022] [Indexed: 10/15/2024]
Abstract
Background Glioma is one of the central nervous system (CNS) tumors in children, accounting for 80% of malignant brain tumors. Nucleotide excision repair (NER) is a vital pathway during DNA damage repair progression. Xeroderma pigmentosum group D (XPD) or excision repair cross-complementing group 2 (ERCC2) is a critical factor in the NER pathway, playing an indispensable role in the DNA repair process. Therefore, the genetic variants in XPD may be associated with carcinogenesis induced by defects in DNA repair. Methods We are the first to conduct a multi-center case-control study to investigate the correlation between XPD gene polymorphisms and pediatric glioma risk. We chose three single nucleotide polymorphisms and genotyped them using the TaqMan assay. Results Although there is no significant association of these genetic variations with glioma susceptibility, the stratified analysis revealed that in the subtype of astrocytic tumors, the rs13181 TG/GG genotype enhanced glioma risk than the TT genotype, and carriers with two to three genotypes also elevated the tumor risk than 0-1 genotypes. Conclusion In conclusion, our findings provided an insight into the impact of XPD genetic variants on glioma risk.
Collapse
Affiliation(s)
- Yong‐Ping Chen
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Yuxiang Liao
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina
| | - Li Yuan
- Department of Pathology, Guangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Xiao‐Kai Huang
- Department of HematologyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| | - Ji‐Chen Ruan
- Department of HematologyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| | - Hui‐Ran Lin
- Faculty of MedicineMacau University of Science and TechnologyMacauChina
| | - Lei Miao
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Zhen‐Jian Zhuo
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhouGuangdongChina
- Laboratory Animal Center, School of Chemical Biology and BiotechnologyPeking University Shenzhen Graduate SchoolShenzhenChina
| |
Collapse
|
7
|
Comprehensive pharmacogenomics characterization of temozolomide response in gliomas. Eur J Pharmacol 2021; 912:174580. [PMID: 34678239 DOI: 10.1016/j.ejphar.2021.174580] [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: 07/07/2021] [Revised: 10/11/2021] [Accepted: 10/18/2021] [Indexed: 01/11/2023]
Abstract
Recent developments in pharmacogenomics have created opportunities for predicting temozolomide response in gliomas. Temozolomide is the main first-line alkylating chemotherapeutic drug together with radiotherapy as standard treatments of high-risk gliomas after surgery. However, there are great individual differences in temozolomide response. Besides the heterogeneity of gliomas, pharmacogenomics relevant genetic polymorphisms can not only affect pharmacokinetics of temozolomide but also change anti-tumor effects of temozolomide. This review will summarize pharmacogenomic studies of temozolomide in gliomas which can lay the foundation to personalized chemotherapy.
Collapse
|
8
|
Liu W, Ma S, Liang L, Kou Z, Zhang H, Yang J. The association between XRCC3 rs1799794 polymorphism and cancer risk: a meta-analysis of 34 case-control studies. BMC Med Genomics 2021; 14:117. [PMID: 33931047 PMCID: PMC8086287 DOI: 10.1186/s12920-021-00965-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 04/22/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Studies on the XRCC3 rs1799794 polymorphism show that this polymorphism is involved in a variety of cancers, but its specific relationships or effects are not consistent. The purpose of this meta-analysis was to investigate the association between rs1799794 polymorphism and susceptibility to cancer. METHODS PubMed, Embase, the Cochrane Library, Web of Science, and Scopus were searched for eligible studies through June 11, 2019. All analyses were performed with Stata 14.0. Subgroup analyses were performed by cancer type, ethnicity, source of control, and detection method. A total of 37 studies with 23,537 cases and 30,649 controls were included in this meta-analysis. RESULTS XRCC3 rs1799794 increased cancer risk in the dominant model and heterozygous model (GG + AG vs. AA: odds ratio [OR] = 1.04, 95% confidence interval [CI] = 1.00-1.08, P = 0.051; AG vs. AA: OR = 1.05, 95% CI = 1.00-1.01, P = 0.015). The existence of rs1799794 increased the risk of breast cancer and thyroid cancer, but reduced the risk of ovarian cancer. In addition, rs1799794 increased the risk of cancer in the Caucasian population. CONCLUSION This meta-analysis confirms that XRCC3 rs1799794 is related to cancer risk, especially increased risk for breast cancer and thyroid cancer and reduced risk for ovarian cancer. However, well-designed large-scale studies are required to further evaluate the results.
Collapse
Affiliation(s)
- Weiqing Liu
- Department of Internal Medicine-Oncology, the First Affiliated Hospital of Kunming Medical University, Yunnan Province, Kunming, 650032, People's Republic of China
| | - Shumin Ma
- Department of Oncology, the First Affiliated Hospital of Kunming Medical University, Yunnan Province, No. 295 Xichang Road, Kunming, 650032, People's Republic of China
| | - Lei Liang
- Department of Oncology, the First Affiliated Hospital of Kunming Medical University, Yunnan Province, No. 295 Xichang Road, Kunming, 650032, People's Republic of China
| | - Zhiyong Kou
- Department of Oncology, the First Affiliated Hospital of Kunming Medical University, Yunnan Province, No. 295 Xichang Road, Kunming, 650032, People's Republic of China
| | - Hongbin Zhang
- Department of Oncology, the First Affiliated Hospital of Kunming Medical University, Yunnan Province, No. 295 Xichang Road, Kunming, 650032, People's Republic of China
| | - Jun Yang
- Department of Oncology, the First Affiliated Hospital of Kunming Medical University, Yunnan Province, No. 295 Xichang Road, Kunming, 650032, People's Republic of China.
| |
Collapse
|
9
|
Yao J, Chen X, Liu Z, Zhang R, Zhang C, Yang Q, Yao P, Jiang Q, Wu J, Zhao S. The increasing expression of GPX7 related to the malignant clinical features leading to poor prognosis of glioma patients. Chin Neurosurg J 2021; 7:21. [PMID: 33750478 PMCID: PMC7945363 DOI: 10.1186/s41016-021-00235-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 01/26/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Glioma is the most common malignant brain tumor in adults. The standard treatment scheme of glioma is surgical resection combined alternative radio- and chemotherapy. However, the outcome of glioma patients was unsatisfied. Here, we aimed to explore the molecular and biological function characteristics of GPX7 in glioma. METHODS The multidimensional data of glioma samples were downloaded from Chinese Glioma Genome Atlas (CGGA). RT-qPCR method was used to identify the expression status of GPX7. Kaplan-Meier curves and Cox regression analysis were used to explore the prognostic value of GPX7. Gene Set Enrichment Analysis (GSEA) was applied to investigate the GPX7-related functions in glioma. RESULTS The results indicated that the expression of GPX7 in glioma was higher compared to that in normal brain tissue. Univariate and multivariate Cox regression analyses confirmed that the expression value of GPX7 was an independent prognostic factor in glioma. The GSEA analysis showed that GPX7 was significantly enriched in the cell cycle pathway, ECM pathway, focal adhesion pathway, and toll-like receptor pathway. CONCLUSIONS The GPX7 was recommended as an independent risk factor for patients diagnosed with glioma for the first time and GPX7 could be potentially used as the therapy target in future. Furthermore, we attempted to explore a potential biomarker for improving the diagnosis and prognosis of patients with glioma.
Collapse
Affiliation(s)
- Jiawei Yao
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, Harbin, 150001, Heilongjiang Province, China.,Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, 150001, Heilongjiang Province, China
| | - Xin Chen
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, Harbin, 150001, Heilongjiang Province, China.,Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, 150001, Heilongjiang Province, China
| | - Zhendong Liu
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, Harbin, 150001, Heilongjiang Province, China.,Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, 150001, Heilongjiang Province, China
| | - Ruotian Zhang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, Harbin, 150001, Heilongjiang Province, China.,Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, 150001, Heilongjiang Province, China
| | - Cheng Zhang
- North Broward Preparatory School, 7600 Lyons Rd, Coconut Creek, FL, 33073, USA
| | - Quan Yang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, Harbin, 150001, Heilongjiang Province, China.,Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, 150001, Heilongjiang Province, China
| | - Penglei Yao
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, Harbin, 150001, Heilongjiang Province, China.,Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, 150001, Heilongjiang Province, China
| | - Qiuyi Jiang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, Harbin, 150001, Heilongjiang Province, China.,Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, 150001, Heilongjiang Province, China
| | - Jianing Wu
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, Harbin, 150001, Heilongjiang Province, China. .,Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, 150001, Heilongjiang Province, China.
| | - Shiguang Zhao
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, Harbin, 150001, Heilongjiang Province, China. .,Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, 150001, Heilongjiang Province, China. .,Department of Neurosurgery, The Pinghu Hospital of Shenzhen University, Shenzhen, 518100, Guangdong Province, China. .,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, 150001, Heilongjiang Province, China.
| |
Collapse
|
10
|
Clinicopathological and Prognostic Roles of STAT3 and Its Phosphorylation in Glioma. DISEASE MARKERS 2020; 2020:8833885. [PMID: 33299498 PMCID: PMC7704152 DOI: 10.1155/2020/8833885] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 08/26/2020] [Accepted: 10/06/2020] [Indexed: 01/07/2023]
Abstract
Glioma is defined as a common brain tumor which causes severe disability or death. As many genes are reported to relate with glioma's occurrence and development, their prognostic and therapeutic value still remains uncertain. This study aimed at investigating the association between STAT3/p-STAT3 and glioma prognosis. Nine studies (12 trials) scored ≥5 on the Newcastle-Ottawa scale were meta-analysed from the Medline, Embase, and Web of Science databases. We found that STAT3/p-STAT3 overexpression in glioma patients was associated with worse overall survival (hazard ratio (HR) = 1.40, 95%confidence interval (CI) = 1.05 ~ 1.86, P = 0.020), progression-free survival (HR = 2.05, 95%CI = 1.63 ~ 2.58, P < 0.001), and better recurrence-free survival (HR = 0.37, 95%CI = 0.15 ~ 0.95, P < 0.039). Subgroup analysis implied that STAT3/p-STAT3 overexpression was associated with worse OS in standard treatment (HR = 1.80, 95%CI = 1.06 ~ 3.04, P = 0.030), and in China (HR = 2.18, 95%CI = 1.77 ~ 2.70, P < 0.001), and metaregression analysis indicated countries (P = 0.001) may be the source of heterogeneity in our study. In conclusion, we suggested STAT3/p-STAT3 was associated with poor prognosis in patients with glioma, which indicated that STAT3/p-STAT3 might be a valuable prognostic biomarker and a promising therapeutic target for glioma.
Collapse
|
11
|
Al-Khatib SM, Abdo N, Al-Eitan LN, Al-Mistarehi AHW, Zahran DJ, Al Ajlouni M, Kewan TZ. The Impact of the Genetic Polymorphism in DNA Repair Pathways on Increased Risk of Glioblastoma Multiforme in the Arab Jordanian Population: A Case-Control Study. APPLICATION OF CLINICAL GENETICS 2020; 13:115-126. [PMID: 32606887 PMCID: PMC7295542 DOI: 10.2147/tacg.s248994] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 05/22/2020] [Indexed: 01/06/2023]
Abstract
Introduction Among the Jordanian population, brain tumors are the tenth most common type of cancers in both males and females, comprising 2.8% of all newly diagnosed neoplasms. Diffuse gliomas are the most prevalent and the most aggressive primary brain tumors in adults. The incidence of diffuse gliomas varies among different populations; this variation is partially linked to genetic polymorphisms. The purpose of the study is to examine the association between (BRCA1 rs799917G>A, rs1799966T>C, EXO1 rs1047840G>A, EME1 rs12450550T>C, ERCC2 rs13181T>G, rs1799793C>T, and XRCC1 rs1799782G>A) DNA repair gene polymorphisms and glioblastoma multiforme (GBM) susceptibility, and survival in the Jordanian Arab population. Methods Eighty-four patients diagnosed with glioblastoma multiforme at the King Abdullah University Hospital (KAUH) between 2013 and 2018 and 225 healthy cancer-free control subjects with similar geographic and ethnic backgrounds to the patients were included in the study. Genomic DNA was extracted from the formalin-fixed paraffin-embedded tissues of the subjects. The Sequenom MassARRAY® sequencer system (iPLEX GOLD) was used. The analyses included assessments of population variability and survival. Results This study is the first to address the relationship between BRCA1 rs1799966 and rs799917 SNP, and the risk of GBM among the Arab Jordanian population. The findings of the study show that BRCA1 rs799917 is associated with decreased risk of GBM in the recessive model (AA vs G/G-A/G: OR, 0.46, 95% CI, 0.26-0.82, p=0.01) and the same SNP is associated with increased risk of GBM in the overdominant model (AG vs G/G-A/A: OR, 1.72, 95% CI, 1.02-2.89, p=0.04).
Collapse
Affiliation(s)
- Sohaib M Al-Khatib
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Nour Abdo
- Department of Public Health, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Laith N Al-Eitan
- Department of Biotechnology and Genetic Engineering, Faculty of Science and Arts, Jordan University of Science and Technology, Irbid, Jordan
| | - Abdel-Hameed W Al-Mistarehi
- Department of Family Medicine, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Deeb Jamil Zahran
- Department of Internal Medicine, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Marwan Al Ajlouni
- Department of Public Health, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Tariq Zuheir Kewan
- Department of Internal Medicine, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| |
Collapse
|
12
|
Huang L, Xu W, Yan D, You X, Shi X, Zhang S, Hong H, Dai L. Genetic Predisposition to Glioma Mediated by a MAPKAP1 Enhancer Variant. Cell Mol Neurobiol 2020; 40:643-652. [PMID: 31773361 DOI: 10.1007/s10571-019-00763-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 11/18/2019] [Indexed: 12/12/2022]
Abstract
Mitogen-activated protein kinase-associated protein 1 (MAPKAP1) is a unique component of the mechanistic target of rapamycin (MTOR) pathway which plays a pivotal role in carcinogenesis. The role of enhancer variant in carcinogenesis receives increased attentions. However, the significance of enhancer variants of MAPKAP1 in glioma has not yet been investigated. The associations of enhancer variants of MAPKAP1 with glioma susceptibility were evaluated in a cohort of 400 glioma patients and 651 controls. The function of glioma susceptibility locus was examined by a set of biochemical assays. We found that an enhancer variant of MAPKAP1 rs473426 was associated with a significantly increased risk of glioma in a dominant manner (OR 1.53, 95% CI 1.13-2.06; P = 0.006). The association for rs1339499 located in the same enhancer approached the borderline of significance after multiple testing correction (OR 0.74, 95% CI 0.56-0.98; P = 0.037). Furthermore, cumulative associations of rs473426 and rs1339499 with glioma risk were observed (P = 0.011). Functional analyses showed that the risk allele rs473426 C downregulated the regulatory activity of enhancer by reducing the binding affinity of a transcriptional activator NFΙC, which resulted in lower gene expression both in vitro and in vivo. These results demonstrate for the first time that enhancer variant of MAPKAP1 confers susceptibility to glioma by downregulation of MAPKAP1 expression, and provide further evidence highlighting MAPKAP1 as a cancer suppressor in glioma carcinogenesis.
Collapse
Affiliation(s)
- Liming Huang
- Department of Medical Oncology (39th Section), The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
| | - Wenshen Xu
- Department of Laboratory Medicine, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Danfang Yan
- Department of Radiation Oncology, The First Affiliated Hospital, Zhejiang University, Hangzhou, 310003, China
| | - Xin You
- Department of Medical Oncology (39th Section), The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Xi Shi
- Department of Medical Oncology (39th Section), The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Shu Zhang
- Department of Medical Oncology (39th Section), The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Hualan Hong
- Department of Medical Oncology (39th Section), The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Lian Dai
- Department of Medicine, The Third Affiliated People's Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, 350108, China.
| |
Collapse
|
13
|
Ülgen E, Can Ö, Bilguvar K, Oktay Y, Akyerli CB, Danyeli AE, Yakıcıer MC, Sezerman OU, Pamir MN, Özduman K. Whole exome sequencing-based analysis to identify DNA damage repair deficiency as a major contributor to gliomagenesis in adult diffuse gliomas. J Neurosurg 2020; 132:1435-1446. [PMID: 30952131 DOI: 10.3171/2019.1.jns182938] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 01/10/2019] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Processes that cause or contribute to cancer, such as aging, exposure to carcinogens, or DNA damage repair deficiency (DDRd), create predictable and traceable nucleotide alterations in one's genetic code (termed "mutational signatures"). Large studies have previously identified various such mutational signatures across cancers that can be attributed to the specific causative processes. To gain further insight into the processes in glioma development, the authors analyzed mutational signatures in adult diffuse gliomas (DGs). METHODS Twenty-five DGs and paired blood samples were whole exome sequenced. Somatic mutational signatures were identified using 2 different methods. Associations of the signatures with age at diagnosis, molecular subset, and mutational load were investigated. As DDRd-related signatures were frequently observed, germline and somatic DDR gene mutations as well as microsatellite instability (MSI) status were determined for all samples. For validation of signature prevalence, publicly available data from The Cancer Genome Atlas (TCGA) were used. RESULTS Each tumor had a unique combination of signatures. The most common signatures were signature 1 (88%, aging related), signature 3 (52%, homologous recombination related), and signature 15 (56%, mismatch repair related). Eighty-four percent of the tumors contained at least 1 DDRd signature. The findings were validated using public TCGA data. The weight of signature 1 positively correlated with age (r = 0.43) while cumulative weight of DDRd signatures negatively correlated with age (r = -0.16). Each subject had at least 1 germline/somatic alteration in a DDR gene, the most common being the risk single nucleotide polymorphism rs1800734 in MLH1. The rs1800734-AA genotype had a higher cumulative DDRd weight as well as higher mutational load; TP53 was the most common somatically altered DDR gene. MSI was observed in 24% of the tumors. No significant associations of MSI status with mutational load, rs1800734, or the cumulative weight of DDRd signatures were identified. CONCLUSIONS Current findings suggest that DDRd may act as a fundamental mechanism in gliomagenesis rather than being a random, secondary event.
Collapse
Affiliation(s)
- Ege Ülgen
- Departments of1Biostatistics and Medical Informatics
| | - Özge Can
- 2Department of Medical Engineering, Acibadem Mehmet Ali Aydınlar University, School of Engineering, Istanbul, Turkey
| | - Kaya Bilguvar
- 3Department of Genetics, Yale Center for Genome Analysis, Yale University, School of Medicine, New Haven, Connecticut
| | - Yavuz Oktay
- 4Izmir International Biomedicine and Genome Institute (IBG), Dokuz Eylül University, Izmir
- 5Department of Medical Biology, Dokuz Eylül University, Izmir; and
| | | | | | - M Cengiz Yakıcıer
- 8Department of Molecular Biology, Acibadem Mehmet Ali Aydınlar University, School of Arts and Sciences, Istanbul, Turkey
| | | | - M Necmettin Pamir
- 9Neurosurgery, Acibadem Mehmet Ali Aydınlar University, School of Medicine, Istanbul
| | - Koray Özduman
- 9Neurosurgery, Acibadem Mehmet Ali Aydınlar University, School of Medicine, Istanbul
| |
Collapse
|
14
|
Viana-Pereira M, Moreno DA, Linhares P, Amorim J, Nabiço R, Costa S, Vaz R, Reis RM. Replication of GWAS identifies RTEL1, CDKN2A/B, and PHLDB1 SNPs as risk factors in Portuguese gliomas patients. Mol Biol Rep 2019; 47:877-886. [PMID: 31721021 DOI: 10.1007/s11033-019-05178-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 10/31/2019] [Indexed: 02/06/2023]
Abstract
Diffuse gliomas are the most common malignant primary brain tumors and remain incurable. A better knowledge of the tumor etiology is required. Specific single nucleotides polymorphisms (SNPs) rs4977756 (CDKN2A/B), rs6010620 (RTEL1), rs498872 (PHLDB1), rs2736100 (TERT), and rs4295627 (CCDC26) have been associated with glioma susceptibility and are potential risk biomarkers. This study aimed to analyze five SNPs associated with glioma susceptibility, in the Portuguese population. SNPs were genotyped using the Sequenom MassARRAY platform in 127 gliomas and 180 controls. Unconditional logistic regression models were used to calculate odds ratio (OR) and 95% confidence intervals. The false-positive report probability was also assessed. The associations between polymorphisms and survival were evaluated using the log-rank test. It was found that the AG and GG genotypes of the rs4977756 (CDKN2A/B) were associated with an increased risk of gliomas (OR 1.85 and OR 2.38) and glioblastomas (OR 2.77 and OR 3.94). The GA genotype of the rs6010620 (RTEL1) was associated with a decreased risk of glioblastomas (OR 0.45). We also observed that the GA genotype of the rs498872 (PHLDB1) was associated with an increased risk of gliomas (OR 2.92) and glioblastomas (OR 2.39). No significant risk associations were found for the rs2736100 (TERT) and rs4295627 (CCDC26). In addition, the genotype AA of the rs498872 (PHLDB1) was associated with poor overall survival of gliomas patients (AA vs. GA, p = 0.037). The rs6010620 (RTEL1), rs4977756 (CDKN2A/B), and rs498872 (PHLDB1) are associated with glioma risk in the Portuguese population and these data may contribute to understanding gliomas etiology.
Collapse
Affiliation(s)
- Marta Viana-Pereira
- School of Medicine, Life and Health Sciences Research Institute (ICVS), University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Minho, Portugal
| | | | - Paulo Linhares
- Department of Neurosurgery, Hospital S. João, Porto, Portugal.,Faculty of Medicine, University of Porto, Porto, Portugal
| | - Júlia Amorim
- Department of Oncology, Hospital de Braga, Braga, Portugal
| | - Rui Nabiço
- Department of Oncology, Hospital de Braga, Braga, Portugal
| | - Sandra Costa
- School of Medicine, Life and Health Sciences Research Institute (ICVS), University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Minho, Portugal
| | - Rui Vaz
- Department of Neurosurgery, Hospital S. João, Porto, Portugal.,Faculty of Medicine, University of Porto, Porto, Portugal
| | - Rui Manuel Reis
- School of Medicine, Life and Health Sciences Research Institute (ICVS), University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal. .,ICVS/3B's - PT Government Associate Laboratory, Minho, Portugal. .,Barretos Cancer Hospital, Molecular Oncology Research Center, Barretos, SP, Brazil.
| |
Collapse
|
15
|
Cagney DN, Sul J, Huang RY, Ligon KL, Wen PY, Alexander BM. The FDA NIH Biomarkers, EndpointS, and other Tools (BEST) resource in neuro-oncology. Neuro Oncol 2019; 20:1162-1172. [PMID: 29294069 DOI: 10.1093/neuonc/nox242] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
In early 2016, the FDA and the National Institutes of Health (NIH) published the first version of the glossary included in the Biomarkers, EndpointS, and other Tools (BEST) resource.1 The BEST glossary was constructed to harmonize and clarify terms used in translational science and medical product development and to provide a common language used for communication by those agencies. It is considered a "living" document that will be updated in the future. This review will discuss the main biomarker and clinical outcome categories contained in the BEST glossary as they apply to neuro-oncology, as well as the overlapping and hierarchical relationships among them.
Collapse
Affiliation(s)
- Daniel N Cagney
- Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Joohee Sul
- Office of Hematology and Oncology Products, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Raymond Y Huang
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Keith L Ligon
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Patrick Y Wen
- Center For Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Brian M Alexander
- Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
16
|
Kewitz-Hempel S, Kurch L, Cepelova M, Volkmer I, Sauerbrey A, Conrad E, Knirsch S, Pöpperl G, Steinbach D, Beer AJ, Kramm CM, Sahlmann CO, Erdlenbruch B, Reinbold WD, Odparlik A, Sabri O, Kluge R, Staege MS. Impact of rs12917 MGMT Polymorphism on [ 18F]FDG-PET Response in Pediatric Hodgkin Lymphoma (PHL). Mol Imaging Biol 2019; 21:1182-1191. [PMID: 30945122 DOI: 10.1007/s11307-019-01350-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
PURPOSE The enzyme O6-methylguanine-DNA methyltransferase (MGMT) is an important component of the DNA repair machinery. MGMT removes O6-methylguanine from the DNA by transferring the methyl group to a cysteine residue in its active site. Recently, we detected the single nucleotide polymorphism (SNP) rs12917 (C/T) in the MGMT sequence adjacent to the active site in Hodgkin lymphoma (HL) cell line KM-H2. We now investigated whether this SNP is also present in other HL cell lines and patient samples. Furthermore, we asked whether this SNP might have an impact on metabolic response in 2-deoxy-2-[18F]fluoro-D-glucose positron emission tomography ([18F]FDG-PET), and on overall treatment outcome based on follow-up intervals of at least 34 months. PROCEDURES We determined the frequency of this MGMT polymorphism in 5 HL cell lines and in 29 pediatric HL (PHL) patients. The patient cohort included 17 female and 12 male patients aged between 4 and 18 years. After characterization of the sequence, we tested a possible association between rs12917 and age, gender, Ann Arbor stage, treatment group, metabolic response following two courses of OEPA (vincristine, etoposide, prednisone, and doxorubicin) chemotherapy, radiotherapy indication, and relapse status. RESULTS We detected the minor T allele in four of five HL cell lines. 11/29 patients carried the minor T allele whereas 18/29 patients showed homozygosity for the major C allele. Interestingly, we observed significantly better metabolic response in PHL patients carrying the rs12917 C allele resulting in a lower frequency of radiotherapy indication. CONCLUSION MGMT polymorphism rs12917 seems to affect chemotherapy response in PHL. The prognostic value of this polymorphism should be investigated in a larger patient cohort.
Collapse
Affiliation(s)
- Stefanie Kewitz-Hempel
- Department of Pediatrics I, Martin Luther University Halle-Wittenberg, Ernst-Grube-Str. 40, 06120, Halle, Germany.,Department of Pediatric Hematology and Oncology, Justus Liebig University, Giessen, Germany.,Department of Dermatology and Venereology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Lars Kurch
- Department of Nuclear Medicine, University Hospital of Leipzig, 04103, Leipzig, Germany
| | - Michaela Cepelova
- Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University in Prague and Motol University Hospital, Praha, Czech Republic
| | - Ines Volkmer
- Department of Pediatrics I, Martin Luther University Halle-Wittenberg, Ernst-Grube-Str. 40, 06120, Halle, Germany
| | | | - Elke Conrad
- Department of Nuclear Medicine, Helios Hospital Erfurt, Erfurt, Germany
| | - Stephanie Knirsch
- Pediatrics 5 (Oncology, Hematology, and Immunology), Klinikum Stuttgart, Olgahospital, Stuttgart, Germany
| | - Gabriele Pöpperl
- Department of Nuclear Medicine, Klinikum Stuttgart, Olgahospital, Stuttgart, Germany
| | - Daniel Steinbach
- Department of Pediatric Hematology and Oncology, University Hospital Ulm, Ulm, Germany
| | - Ambros J Beer
- Department of Nuclear Medicine, University Hospital, Ulm, Germany
| | - Christof M Kramm
- Department of Pediatrics I, Martin Luther University Halle-Wittenberg, Ernst-Grube-Str. 40, 06120, Halle, Germany.,Division of Pediatric Hematology and Oncology, University Medical Center Göttingen, Göttingen, Germany
| | | | - Bernhard Erdlenbruch
- University Hospital for Children and Adolescents, Johannes Wesling Klinikum Minden, Ruhr University Hospital, Bochum, Germany
| | - Wolf-Dieter Reinbold
- Universitätsinstitut für Diagnostische Radiologie, Neuroradiologie und Nuklearmedizin, Johannes Wesling Klinikum Minden, Ruhr University Hospital, Bochum, Germany
| | - Andreas Odparlik
- Department of Nuclear Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Osama Sabri
- Department of Nuclear Medicine, University Hospital of Leipzig, 04103, Leipzig, Germany
| | - Regine Kluge
- Department of Nuclear Medicine, University Hospital of Leipzig, 04103, Leipzig, Germany.
| | - Martin S Staege
- Department of Pediatrics I, Martin Luther University Halle-Wittenberg, Ernst-Grube-Str. 40, 06120, Halle, Germany.
| |
Collapse
|
17
|
The potential role of MGMT rs12917 polymorphism in cancer risk: an updated pooling analysis with 21010 cases and 34018 controls. Biosci Rep 2018; 38:BSR20180942. [PMID: 30232235 PMCID: PMC6435461 DOI: 10.1042/bsr20180942] [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: 06/13/2018] [Revised: 09/12/2018] [Accepted: 09/14/2018] [Indexed: 02/06/2023] Open
Abstract
In the present study, we aimed at determining the potential role of rs12917 polymorphism of the O-6-methylguanine-DNA methyltransferase (MGMT) gene in the occurrence of cancer. Based on the available data from the online database, we performed an updated meta-analysis. We retrieved 537 articles from our database research and finally selected a total of 54 case–control studies (21010 cases and 34018 controls) for a series of pooling analyses. We observed an enhanced risk in cancer cases compared with controls, using the genetic models T/T compared with C/C (P-value of association test <0.001; odds ratio (OR) = 1.29) and T/T compared with C/C+C/T (P<0.001; OR = 1.32). We detected similar positive results in the subgroups ‘Caucasian’, and ‘glioma’ (all P<0.05; OR > 1). However, we detected negative results in our analyses of most of the other subgroups (P>0.05). Begg’s and Egger’s tests indicated that the results were free of potential publication bias, and sensitivity analysis suggested the stability of the pooling results. In summary, the T/T genotype of MGMT rs12917 is likely to be linked to an enhanced susceptibility to cancer overall, especially glioma, in the Caucasian population.
Collapse
|
18
|
Kim J, Kim J, Lee Y. DNA polymerase β deficiency in the p53 null cerebellum leads to medulloblastoma formation. Biochem Biophys Res Commun 2018; 505:548-553. [PMID: 30274781 DOI: 10.1016/j.bbrc.2018.09.166] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 09/26/2018] [Indexed: 10/28/2022]
Abstract
Defects in DNA damage response or repair mechanisms during neurogenesis result in genomic instability, which is causative for several neural defects. These include brain tumors, particularly medulloblastoma, which occurs in the cerebellum with a high incidence in children. We generated an animal model with defective base excision repair during brain development through selective inactivation of DNA polymerase β (Polb) in neuroprogenitor cells. All of Polb conditional knockout mice developed medulloblastoma in a p53 null background, similar to the Xrcc1 and p53 double deficient animal model. XRCC1 is a scaffolding protein which is involved in DNA damage repair and binds to POLB. In both animal models, the histopathological characteristics of the medulloblastoma were similar to those of human classic medulloblastoma. Brain tumor development was slower in the Polb and p53 double null animals than in the Xrcc1 and p53 double knockout animals. Molecular marker analysis suggested that Polb- and Xrcc1-deficient medulloblastomas belonged to the SHHα subtype, underscoring the important role of genomic stability in preventing this devastating pediatric cerebellar tumor.
Collapse
Affiliation(s)
- Jusik Kim
- Genomic Instability Research Center, Ajou University School of Medicine, Suwon, Republic of Korea; Department of Biomedical Sciences, The Graduate School, Ajou University, Suwon, Republic of Korea
| | - Jaemi Kim
- Genomic Instability Research Center, Ajou University School of Medicine, Suwon, Republic of Korea; Genome Stability Institute, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Youngsoo Lee
- Genomic Instability Research Center, Ajou University School of Medicine, Suwon, Republic of Korea; Genome Stability Institute, Ajou University School of Medicine, Suwon, Republic of Korea; Department of Biomedical Sciences, The Graduate School, Ajou University, Suwon, Republic of Korea.
| |
Collapse
|
19
|
Tao S, Liu M, Shen D, Zhang W, Wang T, Bai Y. TGF-β/Smads Signaling Affects Radiation Response and Prolongs Survival by Regulating DNA Repair Genes in Malignant Glioma. DNA Cell Biol 2018; 37:909-916. [PMID: 30230914 DOI: 10.1089/dna.2018.4310] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
To understand the molecular mechanism underlying the causal relationship between aberrant upregulation of transforming growth factor beta (TGF-β) and radio-resistance in glioma. The mouse glioma cell GL261 was irradiated, and relative expression of TGF-β/Smad signaling genes was determined by real-time PCR and western blotting. The DNA repair response on exogenous TGF-β or LY2109761 was evaluated by quantification of diverse genes by real-time PCR and western blotting. Xenograft mice were employed for in vivo investigation to assess the response to irradiation and LY2109761 either alone or in combination. The expression of DNA repair genes was further determined in the xenograft tumor. The TGF-β/Smad signaling pathway was activated by radiation in the GL261 cell line. The exogenous complement of TGF-β significantly stimulated DNA repair response. Administration of LY2109761 suppressed DNA repair genes. Simultaneous treatment with LY2109761 abrogated the upregulation of DNA repair genes in GL261. In the xenograft tumor model, LY2109761 synergistically improved the therapeutic effect of radiation via improvement of sensitivity. Our data suggested that LY2109761 treatment re-sensitized glioma to radiation via antagonizing TGF-β/Smad-induced DNA repair.
Collapse
Affiliation(s)
- Sichen Tao
- Neurosurgery Department, Yidu Central Hospital of Weifang , Qingzhou, Shandong Province, China
| | - Minli Liu
- Neurosurgery Department, Yidu Central Hospital of Weifang , Qingzhou, Shandong Province, China
| | - Dawei Shen
- Neurosurgery Department, Yidu Central Hospital of Weifang , Qingzhou, Shandong Province, China
| | - Wei Zhang
- Neurosurgery Department, Yidu Central Hospital of Weifang , Qingzhou, Shandong Province, China
| | - Tongxin Wang
- Neurosurgery Department, Yidu Central Hospital of Weifang , Qingzhou, Shandong Province, China
| | - Yunan Bai
- Neurosurgery Department, Yidu Central Hospital of Weifang , Qingzhou, Shandong Province, China
| |
Collapse
|
20
|
Linhares P, Viana-Pereira M, Ferreira M, Amorim J, Nabiço R, Pinto F, Costa S, Vaz R, Reis RM. Genetic variants of vascular endothelial growth factor predict risk and survival of gliomas. Tumour Biol 2018; 40:1010428318766273. [PMID: 29584591 DOI: 10.1177/1010428318766273] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The vascular endothelial growth factor regulates angiogenesis that is increased in glioma. VEGF polymorphisms are thought to modulate vascular endothelial growth factor plasma levels and therefore may be implicated in glioma risk. We aimed to clarify the role of VEGF and von Willebrand factor polymorphisms in glioma susceptibility and prognosis. A case-control study of 126 glioma patients and 180 cancer-free controls was performed. Using Sequenom MassARRAY platform, 11 VEGF and 1 VWF polymorphisms were genotyped. Unconditional multivariate logistic regression models were used to calculate odds ratios and 95% confidence intervals. The associations between polymorphisms and survival were evaluated using a Cox regression model. Bonferroni's adjustment was used to correct for multiple testing. The VEGF polymorphism rs833061 was strongly associated with increased risk for glioma (odds ratio = 164.85) and glioblastoma (odds ratio = 155.66), confirmed after Bonferroni correction. Also, the VEGF polymorphisms rs3024994, rs2010963, and particularly the homozygous carriers of rs1005230 were associated with a worse prognosis for glioma and glioblastoma. Our data support a role of VEGF and VWF polymorphisms as glioma biomarkers, with additional potential relevance for molecular stratification of patients for anti-angiogenic therapies.
Collapse
Affiliation(s)
- Paulo Linhares
- 1 Department of Neurosurgery, Hospital S. João, Porto, Portugal.,2 Faculty of Medicine, University of Porto, Porto, Portugal
| | - Marta Viana-Pereira
- 3 Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,4 ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Mónica Ferreira
- 3 Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,4 ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Júlia Amorim
- 5 Department of Oncology, Hospital de Braga, Braga, Portugal
| | - Rui Nabiço
- 5 Department of Oncology, Hospital de Braga, Braga, Portugal
| | - Filipe Pinto
- 3 Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,4 ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Sandra Costa
- 3 Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,4 ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rui Vaz
- 1 Department of Neurosurgery, Hospital S. João, Porto, Portugal.,2 Faculty of Medicine, University of Porto, Porto, Portugal
| | - Rui Manuel Reis
- 3 Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,4 ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.,6 Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
| |
Collapse
|
21
|
Wang X, Sun CL, Hageman L, Smith K, Singh P, Desai S, Hawkins DS, Hudson MM, Mascarenhas L, Neglia JP, Oeffinger KC, Ritchey AK, Robison LL, Villaluna D, Landier W, Bhatia S. Clinical and Genetic Risk Prediction of Subsequent CNS Tumors in Survivors of Childhood Cancer: A Report From the COG ALTE03N1 Study. J Clin Oncol 2017; 35:3688-3696. [PMID: 28976792 DOI: 10.1200/jco.2017.74.7444] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose Survivors of childhood cancer treated with cranial radiation therapy are at risk for subsequent CNS tumors. However, significant interindividual variability in risk suggests a role for genetic susceptibility and provides an opportunity to identify survivors of childhood cancer at increased risk for these tumors. Methods We curated candidate genetic variants from previously published studies in adult-onset primary CNS tumors and replicated these in survivors of childhood cancer with and without subsequent CNS tumors (82 participants and 228 matched controls). We developed prediction models to identify survivors at high or low risk for subsequent CNS tumors and validated these models in an independent matched case-control sample (25 participants and 54 controls). Results We demonstrated an association between six previously published single nucleotide polymorphisms (rs15869 [ BRCA2], rs1805389 [ LIG4], rs8079544 [ TP53], rs25489 [ XRCC1], rs1673041 [ POLD1], and rs11615 [ ERCC1]) and subsequent CNS tumors in survivors of childhood cancer. Including genetic variants in a Final Model containing age at primary cancer, sex, and cranial radiation therapy dose yielded an area under the curve of 0.81 (95% CI, 0.76 to 0.86), which was superior ( P = .002) to the Clinical Model (area under the curve, 0.73; 95% CI, 0.66 to 0.80). The prediction model was successfully validated. The sensitivity and specificity of predicting survivors of childhood cancer at highest or lowest risk of subsequent CNS tumors was 87.5% and 83.5%, respectively. Conclusion It is possible to identify survivors of childhood cancer at high or low risk for subsequent CNS tumors on the basis of genetic and clinical information. This information can be used to inform surveillance for early detection of subsequent CNS tumors.
Collapse
Affiliation(s)
- Xuexia Wang
- Xuexia Wang, University of North Texas, Denton, TX; Can-Lan Sun, City of Hope, Duarte; Leo Mascarenhas, Children's Hospital Los Angeles, University of Southern California, Los Angeles; Doojduen Villaluna, Children's Oncology Group, Monrovia, CA; Lindsey Hageman, Kandice Smith, Purnima Singh, Wendy Landier, and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL; Sunil Desai, University of Alberta, Edmonton, Alberta, Canada; Douglas S. Hawkins, Seattle Children's Hospital, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA; Melissa M. Hudson and Leslie L. Robison, St Jude Children's Research Hospital, Memphis, TN; Joseph P. Neglia, University of Minnesota Medical School, Minneapolis, MN; Kevin C. Oeffinger, Duke University Medical Center, Durham, NC; and A. Kim Ritchey, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Can-Lan Sun
- Xuexia Wang, University of North Texas, Denton, TX; Can-Lan Sun, City of Hope, Duarte; Leo Mascarenhas, Children's Hospital Los Angeles, University of Southern California, Los Angeles; Doojduen Villaluna, Children's Oncology Group, Monrovia, CA; Lindsey Hageman, Kandice Smith, Purnima Singh, Wendy Landier, and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL; Sunil Desai, University of Alberta, Edmonton, Alberta, Canada; Douglas S. Hawkins, Seattle Children's Hospital, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA; Melissa M. Hudson and Leslie L. Robison, St Jude Children's Research Hospital, Memphis, TN; Joseph P. Neglia, University of Minnesota Medical School, Minneapolis, MN; Kevin C. Oeffinger, Duke University Medical Center, Durham, NC; and A. Kim Ritchey, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Lindsey Hageman
- Xuexia Wang, University of North Texas, Denton, TX; Can-Lan Sun, City of Hope, Duarte; Leo Mascarenhas, Children's Hospital Los Angeles, University of Southern California, Los Angeles; Doojduen Villaluna, Children's Oncology Group, Monrovia, CA; Lindsey Hageman, Kandice Smith, Purnima Singh, Wendy Landier, and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL; Sunil Desai, University of Alberta, Edmonton, Alberta, Canada; Douglas S. Hawkins, Seattle Children's Hospital, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA; Melissa M. Hudson and Leslie L. Robison, St Jude Children's Research Hospital, Memphis, TN; Joseph P. Neglia, University of Minnesota Medical School, Minneapolis, MN; Kevin C. Oeffinger, Duke University Medical Center, Durham, NC; and A. Kim Ritchey, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Kandice Smith
- Xuexia Wang, University of North Texas, Denton, TX; Can-Lan Sun, City of Hope, Duarte; Leo Mascarenhas, Children's Hospital Los Angeles, University of Southern California, Los Angeles; Doojduen Villaluna, Children's Oncology Group, Monrovia, CA; Lindsey Hageman, Kandice Smith, Purnima Singh, Wendy Landier, and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL; Sunil Desai, University of Alberta, Edmonton, Alberta, Canada; Douglas S. Hawkins, Seattle Children's Hospital, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA; Melissa M. Hudson and Leslie L. Robison, St Jude Children's Research Hospital, Memphis, TN; Joseph P. Neglia, University of Minnesota Medical School, Minneapolis, MN; Kevin C. Oeffinger, Duke University Medical Center, Durham, NC; and A. Kim Ritchey, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Purnima Singh
- Xuexia Wang, University of North Texas, Denton, TX; Can-Lan Sun, City of Hope, Duarte; Leo Mascarenhas, Children's Hospital Los Angeles, University of Southern California, Los Angeles; Doojduen Villaluna, Children's Oncology Group, Monrovia, CA; Lindsey Hageman, Kandice Smith, Purnima Singh, Wendy Landier, and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL; Sunil Desai, University of Alberta, Edmonton, Alberta, Canada; Douglas S. Hawkins, Seattle Children's Hospital, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA; Melissa M. Hudson and Leslie L. Robison, St Jude Children's Research Hospital, Memphis, TN; Joseph P. Neglia, University of Minnesota Medical School, Minneapolis, MN; Kevin C. Oeffinger, Duke University Medical Center, Durham, NC; and A. Kim Ritchey, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Sunil Desai
- Xuexia Wang, University of North Texas, Denton, TX; Can-Lan Sun, City of Hope, Duarte; Leo Mascarenhas, Children's Hospital Los Angeles, University of Southern California, Los Angeles; Doojduen Villaluna, Children's Oncology Group, Monrovia, CA; Lindsey Hageman, Kandice Smith, Purnima Singh, Wendy Landier, and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL; Sunil Desai, University of Alberta, Edmonton, Alberta, Canada; Douglas S. Hawkins, Seattle Children's Hospital, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA; Melissa M. Hudson and Leslie L. Robison, St Jude Children's Research Hospital, Memphis, TN; Joseph P. Neglia, University of Minnesota Medical School, Minneapolis, MN; Kevin C. Oeffinger, Duke University Medical Center, Durham, NC; and A. Kim Ritchey, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Douglas S Hawkins
- Xuexia Wang, University of North Texas, Denton, TX; Can-Lan Sun, City of Hope, Duarte; Leo Mascarenhas, Children's Hospital Los Angeles, University of Southern California, Los Angeles; Doojduen Villaluna, Children's Oncology Group, Monrovia, CA; Lindsey Hageman, Kandice Smith, Purnima Singh, Wendy Landier, and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL; Sunil Desai, University of Alberta, Edmonton, Alberta, Canada; Douglas S. Hawkins, Seattle Children's Hospital, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA; Melissa M. Hudson and Leslie L. Robison, St Jude Children's Research Hospital, Memphis, TN; Joseph P. Neglia, University of Minnesota Medical School, Minneapolis, MN; Kevin C. Oeffinger, Duke University Medical Center, Durham, NC; and A. Kim Ritchey, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Melissa M Hudson
- Xuexia Wang, University of North Texas, Denton, TX; Can-Lan Sun, City of Hope, Duarte; Leo Mascarenhas, Children's Hospital Los Angeles, University of Southern California, Los Angeles; Doojduen Villaluna, Children's Oncology Group, Monrovia, CA; Lindsey Hageman, Kandice Smith, Purnima Singh, Wendy Landier, and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL; Sunil Desai, University of Alberta, Edmonton, Alberta, Canada; Douglas S. Hawkins, Seattle Children's Hospital, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA; Melissa M. Hudson and Leslie L. Robison, St Jude Children's Research Hospital, Memphis, TN; Joseph P. Neglia, University of Minnesota Medical School, Minneapolis, MN; Kevin C. Oeffinger, Duke University Medical Center, Durham, NC; and A. Kim Ritchey, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Leo Mascarenhas
- Xuexia Wang, University of North Texas, Denton, TX; Can-Lan Sun, City of Hope, Duarte; Leo Mascarenhas, Children's Hospital Los Angeles, University of Southern California, Los Angeles; Doojduen Villaluna, Children's Oncology Group, Monrovia, CA; Lindsey Hageman, Kandice Smith, Purnima Singh, Wendy Landier, and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL; Sunil Desai, University of Alberta, Edmonton, Alberta, Canada; Douglas S. Hawkins, Seattle Children's Hospital, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA; Melissa M. Hudson and Leslie L. Robison, St Jude Children's Research Hospital, Memphis, TN; Joseph P. Neglia, University of Minnesota Medical School, Minneapolis, MN; Kevin C. Oeffinger, Duke University Medical Center, Durham, NC; and A. Kim Ritchey, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Joseph P Neglia
- Xuexia Wang, University of North Texas, Denton, TX; Can-Lan Sun, City of Hope, Duarte; Leo Mascarenhas, Children's Hospital Los Angeles, University of Southern California, Los Angeles; Doojduen Villaluna, Children's Oncology Group, Monrovia, CA; Lindsey Hageman, Kandice Smith, Purnima Singh, Wendy Landier, and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL; Sunil Desai, University of Alberta, Edmonton, Alberta, Canada; Douglas S. Hawkins, Seattle Children's Hospital, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA; Melissa M. Hudson and Leslie L. Robison, St Jude Children's Research Hospital, Memphis, TN; Joseph P. Neglia, University of Minnesota Medical School, Minneapolis, MN; Kevin C. Oeffinger, Duke University Medical Center, Durham, NC; and A. Kim Ritchey, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Kevin C Oeffinger
- Xuexia Wang, University of North Texas, Denton, TX; Can-Lan Sun, City of Hope, Duarte; Leo Mascarenhas, Children's Hospital Los Angeles, University of Southern California, Los Angeles; Doojduen Villaluna, Children's Oncology Group, Monrovia, CA; Lindsey Hageman, Kandice Smith, Purnima Singh, Wendy Landier, and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL; Sunil Desai, University of Alberta, Edmonton, Alberta, Canada; Douglas S. Hawkins, Seattle Children's Hospital, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA; Melissa M. Hudson and Leslie L. Robison, St Jude Children's Research Hospital, Memphis, TN; Joseph P. Neglia, University of Minnesota Medical School, Minneapolis, MN; Kevin C. Oeffinger, Duke University Medical Center, Durham, NC; and A. Kim Ritchey, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - A Kim Ritchey
- Xuexia Wang, University of North Texas, Denton, TX; Can-Lan Sun, City of Hope, Duarte; Leo Mascarenhas, Children's Hospital Los Angeles, University of Southern California, Los Angeles; Doojduen Villaluna, Children's Oncology Group, Monrovia, CA; Lindsey Hageman, Kandice Smith, Purnima Singh, Wendy Landier, and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL; Sunil Desai, University of Alberta, Edmonton, Alberta, Canada; Douglas S. Hawkins, Seattle Children's Hospital, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA; Melissa M. Hudson and Leslie L. Robison, St Jude Children's Research Hospital, Memphis, TN; Joseph P. Neglia, University of Minnesota Medical School, Minneapolis, MN; Kevin C. Oeffinger, Duke University Medical Center, Durham, NC; and A. Kim Ritchey, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Leslie L Robison
- Xuexia Wang, University of North Texas, Denton, TX; Can-Lan Sun, City of Hope, Duarte; Leo Mascarenhas, Children's Hospital Los Angeles, University of Southern California, Los Angeles; Doojduen Villaluna, Children's Oncology Group, Monrovia, CA; Lindsey Hageman, Kandice Smith, Purnima Singh, Wendy Landier, and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL; Sunil Desai, University of Alberta, Edmonton, Alberta, Canada; Douglas S. Hawkins, Seattle Children's Hospital, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA; Melissa M. Hudson and Leslie L. Robison, St Jude Children's Research Hospital, Memphis, TN; Joseph P. Neglia, University of Minnesota Medical School, Minneapolis, MN; Kevin C. Oeffinger, Duke University Medical Center, Durham, NC; and A. Kim Ritchey, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Doojduen Villaluna
- Xuexia Wang, University of North Texas, Denton, TX; Can-Lan Sun, City of Hope, Duarte; Leo Mascarenhas, Children's Hospital Los Angeles, University of Southern California, Los Angeles; Doojduen Villaluna, Children's Oncology Group, Monrovia, CA; Lindsey Hageman, Kandice Smith, Purnima Singh, Wendy Landier, and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL; Sunil Desai, University of Alberta, Edmonton, Alberta, Canada; Douglas S. Hawkins, Seattle Children's Hospital, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA; Melissa M. Hudson and Leslie L. Robison, St Jude Children's Research Hospital, Memphis, TN; Joseph P. Neglia, University of Minnesota Medical School, Minneapolis, MN; Kevin C. Oeffinger, Duke University Medical Center, Durham, NC; and A. Kim Ritchey, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Wendy Landier
- Xuexia Wang, University of North Texas, Denton, TX; Can-Lan Sun, City of Hope, Duarte; Leo Mascarenhas, Children's Hospital Los Angeles, University of Southern California, Los Angeles; Doojduen Villaluna, Children's Oncology Group, Monrovia, CA; Lindsey Hageman, Kandice Smith, Purnima Singh, Wendy Landier, and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL; Sunil Desai, University of Alberta, Edmonton, Alberta, Canada; Douglas S. Hawkins, Seattle Children's Hospital, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA; Melissa M. Hudson and Leslie L. Robison, St Jude Children's Research Hospital, Memphis, TN; Joseph P. Neglia, University of Minnesota Medical School, Minneapolis, MN; Kevin C. Oeffinger, Duke University Medical Center, Durham, NC; and A. Kim Ritchey, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Smita Bhatia
- Xuexia Wang, University of North Texas, Denton, TX; Can-Lan Sun, City of Hope, Duarte; Leo Mascarenhas, Children's Hospital Los Angeles, University of Southern California, Los Angeles; Doojduen Villaluna, Children's Oncology Group, Monrovia, CA; Lindsey Hageman, Kandice Smith, Purnima Singh, Wendy Landier, and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL; Sunil Desai, University of Alberta, Edmonton, Alberta, Canada; Douglas S. Hawkins, Seattle Children's Hospital, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA; Melissa M. Hudson and Leslie L. Robison, St Jude Children's Research Hospital, Memphis, TN; Joseph P. Neglia, University of Minnesota Medical School, Minneapolis, MN; Kevin C. Oeffinger, Duke University Medical Center, Durham, NC; and A. Kim Ritchey, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
22
|
Meta-analysis showing that ERCC1 polymorphism is predictive of osteosarcoma prognosis. Oncotarget 2017; 8:62769-62779. [PMID: 28977987 PMCID: PMC5617547 DOI: 10.18632/oncotarget.19370] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 07/11/2017] [Indexed: 12/14/2022] Open
Abstract
To investigate correlations between excision repair cross-complementation group 1 (ERCC1) and 2 (ERCC2) polymorphisms and osteosarcoma prognosis, we conducted a meta-analysis of studies published through October 2016. Studies were identified in the PubMed, ScienceDirect, Springer, and Web of Science databases using preferred reporting items for systematic reviews and meta-analyses (PRISMA). Odds ratios (ORs) or hazard ratios (HRs) and their 95% confidence intervals (CIs) for overall survival (OS), tumor response (TR), and event-free survival (EFS) were estimated. Our meta-analysis included eleven studies in which four SNPs (ERCC1 rs11615 and rs3212986, ERCC2 rs13181 and rs1799793) reportedly associated with osteosarcoma prognosis were investigated. Each of these studies scored > 6 on the Newcastle-Ottawa Scale (NOS). We found that only one SNP, ERCC1 rs11615, correlated with improved OS and TR. The HR of T vs. C for OS was 1.455 (T/C, 95% CI = 1.151–1.839, P = 0.002, I2 = 37.80%). The OR of T vs. C for good TR was 0.554 (T/C, 95% CI = 0.437–0.702, P < 0.001, I2 = 0%). Few significant outcome was observed in subgroup analyses stratified based on study characteristics with adjustments for potential confounders. Our results suggest that ERCC1 rs11615 CC is associated with a better clinical outcome. This suggests rs11615 may be a useful genetic marker for predicting osteosarcoma prognosis.
Collapse
|
23
|
Guan Q, Chen Z, Chen Q, Zhi X. XRCC1 and XPD polymorphisms and their relation to the clinical course in hepatocarcinoma patients. Oncol Lett 2017; 14:2783-2788. [PMID: 28927037 PMCID: PMC5588103 DOI: 10.3892/ol.2017.6522] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 06/29/2017] [Indexed: 02/07/2023] Open
Abstract
In this study genotyping of hepatocellular carcinoma (HCC) patients was conducted to detect polymorphisms on the X-ray repair cross-complementing 1 (XRCC1) and xeroderma pigmentosum complementary group D (XPD) genes and analyze the relationship of their presence with the clinical features of the cancer. A total of 172 patients with HCC were selected in Qilu Hospital, Shandong University, from January 2010 to September 2011. All patients underwent resection of HCC and no tumor metastases were found. Peripheral venous blood samples (3–5 ml) were collected from the patients to extract genomic DNA. Genotyping was performed by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) and gene sequencing. During the five-year follow-up, the survival of patients with various genotypes of XRCC1 and XPD genes were observed and compared. Logistic regression analysis was used to analyze the association between single nucleotide polymorphisms of XRCC1 and XPD genes and the prognosis of patients with HCC. χ2 tests showed that XRCC1-194, XRCC1-280 and XPD-312 gene polymorphisms were significantly correlated with the number, location and diameter of the tumors (p<0.05). No significant difference was found in the survival curve of patients presenting different genotypes of the XRCC1-194 locus (p>0.05). Nevertheless, a significant difference was found in the survival curve of patients with AA and GG genotypes of the XRCC1-280 locus and in the patients with AA, GA and GG genotypes of the XPD-312 locus (p<0.05). Logistic regression analysis showed that the XRCC1-194 genotype was not an independent risk factor for HCC mortality risk (p>0.05), but XRCC1-280 (OR=1.815, p<0.01) and XPD-312 (OR=1.815, p<0.01) genotypes were independent risk factors for a poor prognosis. Taken together our results point to polymorphisms in XRCC1 and XPD genes as being related to the clinical characteristics of HCC, making them suitable prognostic markers of HCC.
Collapse
Affiliation(s)
- Qinghai Guan
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China.,Department of General Surgery, The Affiliated Hospital of Binzhou Medical College, Binzhou, Shandong 256603, P.R. China
| | - Zhiqiang Chen
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Qiangpu Chen
- Department of General Surgery, The Affiliated Hospital of Binzhou Medical College, Binzhou, Shandong 256603, P.R. China
| | - Xuting Zhi
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
24
|
Qian T, Zhang B, Qian C, He Y, Li Y. Association between common polymorphisms in ERCC gene and glioma risk: A meta-analysis of 15 studies. Medicine (Baltimore) 2017; 96:e6832. [PMID: 28514298 PMCID: PMC5440135 DOI: 10.1097/md.0000000000006832] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND A number of studies have investigated the roles of excision repair cross complementation group 1 (ERCC1), ERCC2, and ERCC5 genes polymorphisms in the development of glioma; however, the results were inconsistent. Here, we performed a meta-analysis to investigate the association between 6 polymorphisms in the ERCC genes (rs3212986, rs11615, rs13181, rs1799793, rs238406, rs17655) and glioma risk. METHODS The PubMed, Embase, and Web of science were searched up to September 6, 2016, for studies on the association between ERCC polymorphisms and glioma risk. A fixed-effects or random-effects model was used to calculate the pooled odds ratios based on the results from the heterogeneity tests. Sensitivity and cumulative meta-analyses were also performed. RESULTS A total of 15 studies were eligible for the pooled analysis, conducted in 2 populations of ethnic descent: 8 Europeans and 7 Asians. The results showed that ERCC1 rs3212986 polymorphism was positively associated with glioma [AA vs CC: odds ratio (OR) = 1.298, 95% confidence interval (95% CI) = 1.043-1.230, P = .025]. Association of the ERCC2 rs13181 and rs1799793 polymorphisms was only observed in Asians (CC vs AA for rs13181: OR = 1.539, 95% CI = 1.122-2.109, P = .007; AA vs GG for rs1799793: OR = 1.474, 95% CI = 1.090-1.994, P = .012). However, no association was observed between glioma risk and ERCC1 rs11615, ERCC2 rs238406, and ERCC5 rs17655 polymorphisms. Moreover, sensitivity and cumulative meta-analyses confirmed the stability of the results. CONCLUSIONS Our meta-analysis indicated that the ERCC1 rs3212986 polymorphism and 2 polymorphisms in ERCC2 gene (rs13181 and rs1799793) contributed to the susceptibility of glioma.
Collapse
|
25
|
Altinoz MA, Elmaci I, Bolukbasi FH, Ekmekci CG, Yenmis G, Sari R, Sav A. MGMT gene variants, temozolomide myelotoxicity and glioma risk. A concise literature survey including an illustrative case. J Chemother 2017; 29:238-244. [PMID: 28436299 DOI: 10.1080/1120009x.2017.1312752] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Temozolomide may cause thrombocytopenia or neutropenia in 3-4% of glioblastoma patients, respectively. However, pancytopenia is rarely reported. MGMT (O6-methylguanine-DNA-methyltransferase) enzyme repairs temozolomide-induced DNA mutations and associates both with antitumour efficacy and myelosuppression. Many studies on the effects of MGMT gene-methylation on temozolomide's effects exist, but much fewer publications concerning MGMT variants were documented. A full sequencing of the MGMT gene was performed in a female glioblastoma patient, who developed pancytopenia following temozolomide treatment. Results indicated the presence of all the rs2308321 (I143 V), rs2308327 (K178R) and rs12917 (L84F) MGMT-variants, which were previously associated with temozolomide myelotoxicity. rs12917 (L84F) variant was reported as associating with lesser risk of gallbladder tumours, yet with higher risk of non-Hodgkin lymphomas related with exposure to chlorinated solvents or hair dyes. DNA repair proteins may exert diverging effects on DNA injuries caused by different chemicals and therefore exerting complex effects on myelotoxicity, antitumour activity and carcinogenesis.
Collapse
Affiliation(s)
| | - Ilhan Elmaci
- a Neuroacademy Group , Istanbul , Turkey.,b Department of Neurosurgery , Memorial Hospital , Istanbul , Turkey
| | - Fatih Han Bolukbasi
- a Neuroacademy Group , Istanbul , Turkey.,b Department of Neurosurgery , Memorial Hospital , Istanbul , Turkey
| | | | - Guven Yenmis
- c Department of Genetics , Acibadem University , Istanbul , Turkey
| | - Ramazan Sari
- d Department of Neurosurgery , Hizmet Hastanesi , Istanbul , Turkey
| | - Aydin Sav
- e Nisantasi Pathology Group , Istanbul , Turkey
| |
Collapse
|
26
|
Azevedo AP, Silva SN, De Lima JP, Reichert A, Lima F, Júnior E, Rueff J. DNA repair genes polymorphisms and genetic susceptibility to Philadelphia-negative myeloproliferative neoplasms in a Portuguese population: The role of base excision repair genes polymorphisms. Oncol Lett 2017; 13:4641-4650. [PMID: 28599464 PMCID: PMC5452988 DOI: 10.3892/ol.2017.6065] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 01/26/2017] [Indexed: 02/06/2023] Open
Abstract
The role of base excision repair (BER) genes in Philadelphia-negative (PN)-myeloproliferative neoplasms (MPNs) susceptibility was evaluated by genotyping eight polymorphisms [apurinic/apyrimidinic endodeoxyribonuclease 1, mutY DNA glycosylase, earlier mutY homolog (E. coli) (MUTYH), 8-oxoguanine DNA glycosylase 1, poly (ADP-ribose) polymerase (PARP) 1, PARP4 and X-ray repair cross-complementing 1 (XRCC1)] in a case-control study involving 133 Caucasian Portuguese patients. The results did not reveal a correlation between individual BER polymorphisms and PN-MPNs when considered as a whole. However, stratification for essential thrombocythaemia revealed i) borderline effect/tendency to increased risk when carrying at least one variant allele for XRCC1_399 single-nucleotide polymorphism (SNP); ii) decreased risk for Janus kinase 2-positive patients carrying at least one variant allele for XRCC1_399 SNP; and iii) decreased risk in females carrying at least one variant allele for MUTYH SNP. Combination of alleles demonstrated an increased risk to PN-MPNs for one specific haplogroup. These findings may provide evidence for gene variants in susceptibility to MPNs. Indeed, common variants in DNA repair genes may hamper the capacity to repair DNA, thus increasing cancer susceptibility.
Collapse
Affiliation(s)
- Ana P Azevedo
- Centre for Toxicogenomics and Human Health (ToxOmics), Genetics, Oncology and Human Toxicology, NOVA Medical School, Faculty of Medical Sciences, NOVA University of Lisbon, 1169-056 Lisbon, Portugal.,Department of Clinical Pathology, Hospital of São Francisco Xavier, West Lisbon Hospital Centre, 1449-005 Lisbon, Portugal
| | - Susana N Silva
- Centre for Toxicogenomics and Human Health (ToxOmics), Genetics, Oncology and Human Toxicology, NOVA Medical School, Faculty of Medical Sciences, NOVA University of Lisbon, 1169-056 Lisbon, Portugal
| | - João P De Lima
- Centre for Toxicogenomics and Human Health (ToxOmics), Genetics, Oncology and Human Toxicology, NOVA Medical School, Faculty of Medical Sciences, NOVA University of Lisbon, 1169-056 Lisbon, Portugal
| | - Alice Reichert
- Department of Clinical Haematology, Hospital of São Francisco Xavier, West Lisbon Hospital Centre, 1449-005 Lisbon, Portugal
| | - Fernando Lima
- Department of Clinical Haematology, Hospital of São Francisco Xavier, West Lisbon Hospital Centre, 1449-005 Lisbon, Portugal
| | - Esmeraldina Júnior
- Department of Clinical Pathology, Hospital of São Francisco Xavier, West Lisbon Hospital Centre, 1449-005 Lisbon, Portugal
| | - José Rueff
- Centre for Toxicogenomics and Human Health (ToxOmics), Genetics, Oncology and Human Toxicology, NOVA Medical School, Faculty of Medical Sciences, NOVA University of Lisbon, 1169-056 Lisbon, Portugal
| |
Collapse
|
27
|
Reappraisal of XRCC1 Arg194Trp polymorphism and glioma risk: a cumulative meta-analysis. Oncotarget 2017; 8:21599-21608. [PMID: 28423490 PMCID: PMC5400609 DOI: 10.18632/oncotarget.15376] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 01/11/2017] [Indexed: 12/18/2022] Open
Abstract
The association between XRCC1 Arg194Trp polymorphism and glioma risk were inconsistent from published meta-analyses and epidemiological studies. Hence, we performed this updated and cumulative meta-analysis to reappraisal this relationship. PubMed, Embase, CBM (Chinese Biomedical Database), and CNKI (China National Knowledge Internet) databases were comprehensively searched up to August 13, 2016 (updated on December 22, 2016). After study selection and data extraction from eligible studies, the association was evaluated by odds ratios (ORs) and its 95% confidence intervals (95%CIs) using Comprehensive Meta-Analysis software. Finally 16 case-control studies involving 7011 patients and 9519 healthy controls were yielded. The results indicated that XRCC1 Arg194Trp polymorphism was significantly correlated with the increased risk of glioma [Trp vs. Arg: OR = 1.18(1.05-1.34); TrpTrp vs. ArgArg: OR = 1.66(1.31-2.12); ArgTrp vs. ArgArg: OR = 1.34(1.02-1.77); TrpTrp vs. ArgArg+ArgTrp: OR = 1.47(1.26-1.72); TrpTrp+ArgTrp vs. ArgArg: OR = 1.17(1.01-1.35)]. Cumulative analysis showed the results changed from non-significant to significant when new studies accumulated, and sensitivity analysis indicated the results were stable. Subgroup analysis showed the significant association existed in Asians but not in Caucasians. Current evidence indicated that XRCC1 Arg194Trp polymorphism was associated with increased risk for glioma, especially in Asians; however, relevant studies involving other ethnic groups are required to validate our findings in further.
Collapse
|
28
|
Liu K, Jiang Y. Polymorphisms in DNA Repair Gene and Susceptibility to Glioma: A Systematic Review and Meta-Analysis Based on 33 Studies with 15 SNPs in 9 Genes. Cell Mol Neurobiol 2017; 37:263-274. [PMID: 27055523 DOI: 10.1007/s10571-016-0367-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Accepted: 03/22/2016] [Indexed: 11/29/2022]
Abstract
At present, many publications have evaluated the correlation between the DNA repair gene polymorphisms and glioma susceptibility. However, the results remain inconclusive. The aim of this research is to exhaustively assess the association of genetic polymorphisms in DNA repair genes with glioma risk in human. Meta-analysis method was conducted, and 33 studies with 15 SNPs in 9 genes were included (12553 glioma cases and 17178 controls). Correlation strength was evaluated by odds ratio with a 95 % confidence interval. Rs1799782 T allele and rs25487A allele might bring about higher risk of glioma in Asian population. Rs1805377 G allele was an increased risk genetic factor of glioma. Asian carried with rs3212986 A allele was more likely to have glioma. Rs1800067 G allele was a risk factor of developing glioma. Carriers with rs12917 CC genotype in MGMT gene had higher risk of glioma in Caucasian than other non-CC genotype carriers. Carriers with rs1136410 T allele in PARP1 gene could more likely to develop glioma in Caucasian. This meta-analysis suggests that glioma susceptibility is associated with rs1799782 and rs25487 of X-ray repair complementing defective repair in Chinese hamster cells 1 (XRCC1), rs1805377 of XRCC4, rs1800067 of excision repair cross-complementing rodent repair deficiency complementation group 4 (ERCC4) and rs3212986 of ERCC1 in Asian population, and rs12917 of O-6-methylguanine-DNA methyltransferase (MGMT) and rs1136410 of poly(ADP-ribose) polymerase 1 (PARP1) in Caucasian population.
Collapse
Affiliation(s)
- Kun Liu
- Department of neurosurgery, The Second Xiangya Hospital of Central South University, 139 Renmin(M) Road, Changsha, 410011, Hunan, China
- Department of neurosurgery, Brains Hospital of Hunan Province, Changsha, China
| | - Yugang Jiang
- Department of neurosurgery, The Second Xiangya Hospital of Central South University, 139 Renmin(M) Road, Changsha, 410011, Hunan, China.
| |
Collapse
|
29
|
Du C, Pan P, Jiang Y, Zhang Q, Bao J, Liu C. Microarray data analysis to identify crucial genes regulated by CEBPB in human SNB19 glioma cells. World J Surg Oncol 2016; 14:258. [PMID: 27716259 PMCID: PMC5054626 DOI: 10.1186/s12957-016-0997-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 08/30/2016] [Indexed: 01/18/2023] Open
Abstract
Background Glioma is one of the most common primary malignancies in the brain or spine. The transcription factor (TF) CCAAT/enhancer binding protein beta (CEBPB) is important for maintaining the tumor initiating capacity and invasion ability. To investigate the regulation mechanism of CEBPB in glioma, microarray data GSE47352 was analyzed. Methods GSE47352 was downloaded from Gene Expression Omnibus, including three samples of SNB19 human glioma cells transduced with non-target control small hairpin RNA (shRNA) lentiviral vectors for 72 h (normal glioma cells) and three samples of SNB19 human glioma cells transduced with CEBPB shRNA lentiviral vectors for 72 h (CEBPB-silenced glioma cells). The differentially expressed genes (DEGs) were screened using limma package and then annotated. Afterwards, the Database for Annotation, Visualization, and Integrated Discovery (DAVID) software was applied to perform enrichment analysis for the DEGs. Furthermore, the protein-protein interaction (PPI) network and transcriptional regulatory network were constructed using Cytoscape software. Results Total 529 DEGs were identified in the normal glioma cells compared with the CEBPB-silenced glioma cells, including 336 up-regulated and 193 down-regulated genes. The significantly enriched pathways included chemokine signaling pathway (which involved CCL2), focal adhesion (which involved THBS1 and THBS2), TGF-beta signaling pathway (which involved THBS1, THBS2, SMAD5, and SMAD6) and chronic myeloid leukemia (which involved TGFBR2 and CCND1). In the PPI network, CCND1 (degree = 29) and CCL2 (degree = 12) were hub nodes. Additionally, CEBPB and TCF12 might function in glioma through targeting others (CEBPB → TCF12, CEBPB → TGFBR2, and TCF12 → TGFBR2). Conclusions CEBPB might act in glioma by regulating CCL2, CCND1, THBS1, THBS2, SMAD5, SMAD6, TGFBR2, and TCF12.
Collapse
Affiliation(s)
- Chenghua Du
- Department of Neurosurgery, The Affiliated Hospital of Inner Mongolia University for the Nationalities, Huolinhe Street No.1742, Tongliao, Inner Mongolia, 028007, China.
| | - Pan Pan
- Department of Hepatology, Tongliao City Hospital for Infectious Diseases, Tongliao, Inner Mongolia, 028007, China
| | - Yan Jiang
- Department of Neurosurgery, The Affiliated Hospital of Inner Mongolia University for the Nationalities, Huolinhe Street No.1742, Tongliao, Inner Mongolia, 028007, China
| | - Qiuli Zhang
- Department of Neurosurgery, The Affiliated Hospital of Inner Mongolia University for the Nationalities, Huolinhe Street No.1742, Tongliao, Inner Mongolia, 028007, China
| | - Jinsuo Bao
- Department of Neurosurgery, The Affiliated Hospital of Inner Mongolia University for the Nationalities, Huolinhe Street No.1742, Tongliao, Inner Mongolia, 028007, China
| | - Chang Liu
- Department of Neurosurgery, The Affiliated Hospital of Inner Mongolia University for the Nationalities, Huolinhe Street No.1742, Tongliao, Inner Mongolia, 028007, China
| |
Collapse
|
30
|
Fahmideh MA, Lavebratt C, Schüz J, Röösli M, Tynes T, Grotzer MA, Johansen C, Kuehni CE, Lannering B, Prochazka M, Schmidt LS, Feychting M. Common genetic variations in cell cycle and DNA repair pathways associated with pediatric brain tumor susceptibility. Oncotarget 2016; 7:63640-63650. [PMID: 27613841 PMCID: PMC5325391 DOI: 10.18632/oncotarget.11575] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 08/15/2016] [Indexed: 01/11/2023] Open
Abstract
Knowledge on the role of genetic polymorphisms in the etiology of pediatric brain tumors (PBTs) is limited. Therefore, we investigated the association between single nucleotide polymorphisms (SNPs), identified by candidate gene-association studies on adult brain tumors, and PBT risk.The study is based on the largest series of PBT cases to date. Saliva DNA from 245 cases and 489 controls, aged 7-19 years at diagnosis/reference date, was genotyped for 68 SNPs. Data were analyzed using unconditional logistic regression.The results showed EGFRrs730437 and EGFRrs11506105 may decrease susceptibility to PBTs, whereas ERCC1rs3212986 may increase risk of these tumors. Moreover, stratified analyses indicated CHAF1Ars243341, CHAF1Ars2992, and XRCC1rs25487 were associated with a decreased risk of astrocytoma subtype. Furthermore, an increased risk of non-astrocytoma subtype associated with EGFRrs9642393, EME1rs12450550, ATMrs170548, and GLTSCRrs1035938 as well as a decreased risk of this subtype associated with XRCC4rs7721416 and XRCC4rs2662242 were detected.This study indicates SNPs in EGFR, ERCC1, CHAF1A, XRCC1, EME1, ATM, GLTSCR1, and XRCC4 may be associated with the risk of PBTs. Therefore, cell cycle and DNA repair pathways variations associated with susceptibility to adult brain tumors also seem to be associated with PBT risk, suggesting pediatric and adult brain tumors might share similar etiological pathways.
Collapse
Affiliation(s)
- Maral Adel Fahmideh
- Unit of Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Catharina Lavebratt
- Neurogenetics Unit, Department of Molecular Medicine and Surgery, Karolinska Institutet, and Center for Molecular Medicine, Karolinska University Hospital, L8:00, SE-171 76 Stockholm, Sweden
| | - Joachim Schüz
- Section of Environment and Radiation, International Agency for Research on Cancer (IARC), 69372 Lyon, France
| | - Martin Röösli
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, 4002 Basel, Switzerland
- University of Basel, 4003 Basel, Switzerland
| | - Tore Tynes
- The Cancer Registry of Norway, NO-0379 Oslo, Norway
- National Institute of Occupational Health, NO-0360 Oslo, Norway
| | - Michael A. Grotzer
- Department of Oncology, University Children's Hospital of Zurich, 8032 Zurich, Switzerland
| | - Christoffer Johansen
- Unit of Survivorship, The Danish Cancer Society Research Centre, DK-2100 Copenhagen, Denmark
- Oncology Department, Finsen Centre, Rigshospitalet, DK-2100 Copenhagen, Denmark
| | - Claudia E Kuehni
- Swiss Childhood Cancer Registry, Institute of Social and Preventive Medicine, University of Bern, 3012 Bern, Switzerland
| | - Birgitta Lannering
- Childrens Cancer Center, Queen Silvia Childrens Hospital, SE-416 85 Gothenburg, Sweden
| | - Michaela Prochazka
- Unit of Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Lisbeth S Schmidt
- Department of Clinical Genetics, University Hospital Rigshospitalet, DK-2100 Copenhagen, Denmark
| | - Maria Feychting
- Unit of Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| |
Collapse
|
31
|
Polymorphisms in DNA repair genes in gastrointestinal stromal tumours: susceptibility and correlation with tumour characteristics and clinical outcome. Tumour Biol 2016; 37:13413-13423. [PMID: 27460091 DOI: 10.1007/s13277-016-5276-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 07/15/2016] [Indexed: 12/13/2022] Open
Abstract
DNA repair pathways play an essential role in cancer susceptibility by maintaining genomic integrity. This led us to investigate the influence of polymorphisms in the genes coding repair pathway enzymes on gastrointestinal stromal tumours (GIST) susceptibility, tumour characteristics and clinical outcome. We investigated a panel of 20 polymorphisms in 11 genes in 81 cases and 147 controls. The XPD rs13181 wild-type allele and hOGG1 rs1052133 and XPF rs1800067 minor alleles were significantly associated with disease susceptibility. XPA rs1800975 and rs2808668 were associated with tumour size (P = 0.018), metastatic status at onset (P = 0.035) and mitotic index (P = 0.002). With regards to outcome treatment, the XPD rs50872 minor allele had a significant favourable impact on time to progression (TTP). Similarly, the XPC rs2228000 minor allele was correlated with a longer TTP (P = 0.03). On the contrary, the XPC rs2228001 and hOGG1 rs1052133 minor alleles were associated with a diminished TTP (P = 0.005 and P = 0.01, respectively). Regarding OS, we found the presence of at least one hOGG1 (rs1052133) minor allele that had a 60 % lower risk to die compared to the wild-type carriers (P = 0.04). Furthermore, the XRCC3 rs861539 variant allele is associated with a hazard of early death compared with the wild-type genotype (P = 0.04). To the best of our knowledge, this is the first study on polymorphisms in DNA repair genes, belonging to the different pathways, extensively evaluated in GIST patients. Through this multiple candidate gene approach, we report for the first time the significant associations between polymorphisms in DNA repair genes, susceptibility, clinical pathological features and clinical outcome in GIST.
Collapse
|
32
|
Qi L, Yu HQ, Zhang Y, Ding LJ, Zhao DH, Lv P, Wang WY, Xu Y. A Comprehensive Meta-analysis of Genetic Associations Between Key Polymorphic Loci in DNA Repair Genes and Glioma Risk. Mol Neurobiol 2016; 54:1314-1325. [PMID: 26843108 DOI: 10.1007/s12035-016-9725-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 01/13/2016] [Indexed: 02/07/2023]
Abstract
Genetic variants found in DNA repair genes (ERCC1, rs3212986; ERCC2, rs13181; ERCC4, rs1800067; ERCC5, rs17655; XRCC1, rs1799782, rs25487, rs25489; XRCC3, rs861539) have been reported to have an ambivalent association with the development of glioma. In the present study, a meta-analysis was conducted to confirm the relationship, taking heterogeneity of population into consideration. We analyzed 21 articles of 6 genes along with 8 single nucleotide polymorphisms (SNPs) (24,078 cases and 30,926 healthy individuals), which assessed the relationship between nucleotide excision, base excision, double-strand break repair gene, and the development of glioma under five models. All statistical analysis was implemented by the software of R 3.2.1, and the relationships between key polymorphic loci in DNA repair genes and glioma were quantified by the pooled odds ratio (OR) and 95 % confidential intervals. Overall, the synthesized evidence demonstrated that the SNP of rs13181 and rs1799782 significantly increased the risk of glioma whereas SNP of rs1800067 were significantly associated with a decrease in the risk of glioma. Additionally, subgroup analyses of 8 SNPs by ethnicity indicated that the mutation of rs13181, rs1800067 were apparently protective factors of glioma among Asians, while the mutation of rs13181 was a risk factors of glioma in Caucasians. Furthermore, the mutation of rs1799782 significantly raises the risk of glioma for Asian. Our study suggested that rs13181*C and rs1799782*A are risk alleles for glioma; rs1800067*A are beneficial alleles for decreased susceptibility to glioma. Future studies with large sample size and other races are strongly recommended to confirm the results from this study.
Collapse
Affiliation(s)
- Ling Qi
- Department of Pathology, Jilin Medical University, Jilin, 132013, China
| | - Hong-Quan Yu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, 130021, China
| | - Yu Zhang
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, 130021, China
| | - Li-Juan Ding
- Department of Radiation Oncology, First Hospital of Jilin University, Changchun, 130021, China
| | - Dong-Hai Zhao
- Department of Pathology, Jilin Medical University, Jilin, 132013, China
| | - Peng Lv
- Department of Pathology, Jilin Medical University, Jilin, 132013, China
| | - Wei-Yao Wang
- Department of Pathology, Jilin Medical University, Jilin, 132013, China
| | - Ye Xu
- Medical Research Laboratory, Jilin Medical University, No.5 Jilin District, Jilin, 132013, China.
| |
Collapse
|
33
|
Henríquez-Hernández LA, Valenciano A, Foro-Arnalot P, Álvarez-Cubero MJ, Cozar JM, Suárez-Novo JF, Castells-Esteve M, Fernández-Gonzalo P, De-Paula-Carranza B, Ferrer M, Guedea F, Sancho-Pardo G, Craven-Bartle J, Ortiz-Gordillo MJ, Cabrera-Roldán P, Rodríguez-Melcón JI, Herrera-Ramos E, Rodríguez-Gallego C, Lara PC. Association between single-nucleotide polymorphisms in DNA double-strand break repair genes and prostate cancer aggressiveness in the Spanish population. Prostate Cancer Prostatic Dis 2016; 19:28-34. [PMID: 26754263 DOI: 10.1038/pcan.2015.63] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 10/27/2015] [Accepted: 11/24/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND Novel predictors of prognosis and treatment response for prostate cancer (PCa) are required to better individualize treatment. Single-nucleotide polymorphisms (SNPs) in four genes directly (XRCC5 (X-ray repair complementing defective repair in Chinese hamster cells 5) and XRCC6 (X-ray repair complementing defective repair in Chinese hamster cells 6)) or indirectly (PARP1 and major vault protein (MVP)) involved in non-homologous end joining were examined in 494 Spanish PCa patients. METHODS A total of 22 SNPs were genotyped in a Biotrove OpenArray NT Cycler. Clinical tumor stage, diagnostic PSA serum levels and Gleason score at diagnosis were obtained for all participants. Genotypic and allelic frequencies were determined using the web-based environment SNPator. RESULTS (XRCC6) rs2267437 appeared as a risk factor for developing more aggressive PCa tumors. Those patients carrying the GG genotype were at higher risk of developing bigger tumors (odds ratio (OR)=2.04, 95% confidence interval (CI) 1.26-3.29, P=0.004), present higher diagnostic PSA levels (OR=2.12, 95% CI 1.19-3.78, P=0.011), higher Gleason score (OR=1.65, 95% CI 1.01-2.68, P=0.044) and D'Amico higher risk tumors (OR=2.38, 95% CI 1.24-4.58, P=0.009) than those patients carrying the CC/CG genotypes. Those patients carrying the (MVP) rs3815824 TT genotype were at higher risk of presenting higher diagnostic PSA levels (OR=4.74, 95% CI 1.40-16.07, P=0.013) than those patients carrying the CC genotype. When both SNPs were analyzed in combination, those patients carrying the risk genotypes were at higher risk of developing D'Amico higher risk tumors (OR=3.33, 95% CI 1.56-7.17, P=0.002). CONCLUSIONS We believe that for the first time, genetic variants at XRCC6 and MVP genes are associated with risk of more aggressive disease, and would be taken into account when assessing the malignancy of PCa.
Collapse
Affiliation(s)
- L A Henríquez-Hernández
- Radiation Oncology Department, Hospital Universitario de Gran Canaria Dr. Negrín, Las Palmas, Spain.,Instituto Canario de Investigación del Cáncer, Las Palmas, Spain.,Clinical Sciences Department, Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| | - A Valenciano
- Instituto Canario de Investigación del Cáncer, Las Palmas, Spain
| | - P Foro-Arnalot
- Radiation Oncology Department, Institud d'Oncologia Radioteràpica, Hospital de la Esperanza, Parc de Salut Mar, Barcelona, Spain
| | - M J Álvarez-Cubero
- Laboratory of Genetic Identification, Legal Medicine and Toxicology Department, Facultad de Medicina, Universidad de Granada, Granada, Spain.,GENYO, Pfizer-University of Granada-Andalusian Government Centre for Genomics and Oncological Research, Granada, Spain
| | - J M Cozar
- Department of Urology, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - J F Suárez-Novo
- Department of Urology, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
| | - M Castells-Esteve
- Department of Urology, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
| | | | | | - M Ferrer
- Health Services Research Group, Institut de Recerca Hospital del Mar (IMIM), Barcelona, Spain
| | - F Guedea
- Department of Radiation Oncology, Institut Català d'Oncologia (ICO), L'Hospitalet de Llobregat, Barcelona, Spain
| | - G Sancho-Pardo
- Radiation Oncology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - J Craven-Bartle
- Radiation Oncology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - M J Ortiz-Gordillo
- Radiation Oncology Department, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - P Cabrera-Roldán
- Radiation Oncology Department, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - J I Rodríguez-Melcón
- Radiation Oncology Department, Hospital Universitario de Gran Canaria Dr. Negrín, Las Palmas, Spain.,Instituto Canario de Investigación del Cáncer, Las Palmas, Spain
| | - E Herrera-Ramos
- Department of Immunology, Hospital Universitario de Gran Canaria Dr. Negrín, Las Palmas, Spain.,Department of Medical and Surgical Sciences, Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| | - C Rodríguez-Gallego
- Department of Immunology, Hospital Universitario de Gran Canaria Dr. Negrín, Las Palmas, Spain
| | - P C Lara
- Radiation Oncology Department, Hospital Universitario de Gran Canaria Dr. Negrín, Las Palmas, Spain.,Instituto Canario de Investigación del Cáncer, Las Palmas, Spain.,Clinical Sciences Department, Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| |
Collapse
|
34
|
Li Y, Li Y, Ge P, Ma C. MiR-126 Regulates the ERK Pathway via Targeting KRAS to Inhibit the Glioma Cell Proliferation and Invasion. Mol Neurobiol 2016; 54:137-145. [PMID: 26732596 DOI: 10.1007/s12035-015-9654-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 12/17/2015] [Indexed: 11/30/2022]
Abstract
The activity of some constitutive contained in the extracellular signal-regulated kinase (ERK) pathway plays crucial roles in glioma cell growth and proliferation. Emerging studies have reported that microRNA (miRNA) could regulate the ERK signal pathway by directly targeting various oncogenes. This study enabled us to discover that the average miR-126 expression was significantly decreased in glioblastoma tissues, and this significant decrease was related to high histopathological grades. Our experiment also demonstrated that the over-expression of miR-126 suppressed glioma cell proliferation and invasion in vitro. Kirsten rat sarcoma viral oncogene (KRAS) which is involved in ERK pathway was directly targeted by miR-126 in glioma through binding to two sites in the 3' untranslated region (3'-UTR) of KRAS mRNA. Notably, the expression level of KRAS was positively correlated to the activity of ERK pathway and its downstream regulators (phosphorylation level of ERK (pERK) and c-Fos). Furthermore, the over-expression of KRAS expression vector without the 3'-UTR partially reverses the tumor-suppressive effects of miR-126. Moreover, the up-regulation of miR-126 contributes to the aberrant activation of the ERK signaling and inhibits cell proliferation and invasion through targeting KRAS. Therefore, it was suspected that miR-126 may be a potential therapeutic target for high-grade glioma.
Collapse
Affiliation(s)
- Yang Li
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, 130021, Jilin Province, China
| | - Yunqian Li
- Department of Neurosurgery, The First Hospital of Jilin University, Xinmin Street 71, Changchun, 130021, Jilin Province, China
| | - Pengfei Ge
- Department of Neurosurgery, The First Hospital of Jilin University, Xinmin Street 71, Changchun, 130021, Jilin Province, China
| | - Chengyuan Ma
- Department of Neurosurgery, The First Hospital of Jilin University, Xinmin Street 71, Changchun, 130021, Jilin Province, China.
| |
Collapse
|
35
|
Abstract
To systematically assess the relationship between smoking and glioma risk.A dose-response meta-analysis of case-control and cohort studies was performed. Pertinent studies were identified by searching database and reference lists. Random-effects model was employed to pool the estimates of the relative risks (RRs) with corresponding 95% confidence intervals (CIs).A total of 19 case-control and 6 cohort studies were included. Overall, compared with those who never smoked, the pooled RR and 95% CI was 0.98 (0.92-1.05) for ever smoker. The subgroups were not significantly different regarding risk of glioma except the group of age at start smoking (RR = 1.17, 95% CI: 0.93-1.48 for age < 20; RR = 1.25, 95% CI: 1.02-1.52 for age ≥ 20). Dose-response analysis also suggested no significant association between smoking and the risk of glioma, although some evidence for a linear relationship between smoking and glioma risk was observed.In conclusion, this meta-analysis provides little support for a causal relationship between smoking and risk of glioma.
Collapse
Affiliation(s)
- Chuan Shao
- From the Department of Neurosurgery, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, Sichuan, China (CS, WZ, JH); and Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China (ZQ)
| | | | | | | |
Collapse
|
36
|
Pouchieu C, Baldi I, Gruber A, Berteaud E, Carles C, Loiseau H. Descriptive epidemiology and risk factors of primary central nervous system tumors: Current knowledge. Rev Neurol (Paris) 2015; 172:46-55. [PMID: 26708326 DOI: 10.1016/j.neurol.2015.10.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 10/08/2015] [Accepted: 10/09/2015] [Indexed: 11/16/2022]
Abstract
Although comparisons are difficult due to differences in methodologies, the annual incidence rates of central nervous system (CNS) tumors range from 8.5 to 21.4/100,000 population according to cancer registries, with a predominance of neuroepithelial tumors in men and meningiomas in women. An increase in the incidence of CNS tumors has been observed during the past decades in several countries. It has been suggested that this trend could be due to aging of the population, and improvements in diagnostic imaging and healthcare access, but these factors do not explain differences in incidence by gender and histological subtypes. Several etiological hypotheses related to intrinsic (sociodemographic, anthropometric, hormonal, immunological, genetic) and exogenous (ionizing radiation, electromagnetic fields, diet, infections, pesticides, drugs) risk factors have led to analytical epidemiological studies to establish relationships with CNS tumors. The only established environmental risk factor for CNS tumors is ionizing radiation exposure. However, for other risk factors, studies have been inconsistent and inconclusive due to systematic differences in study design and difficulties in accurately measuring exposures. Thus, the etiology of CNS tumors is complex and may involve several genetic and/or environmental factors that may act differently according to histological subtype.
Collapse
Affiliation(s)
- C Pouchieu
- ISPED, Équipe Santé Travail Environnement, Université de Bordeaux, 33000 Bordeaux, France; Inserm, ISPED, Centre Inserm U1219, Bordeaux Population Health Center, 33000 Bordeaux, France
| | - I Baldi
- ISPED, Équipe Santé Travail Environnement, Université de Bordeaux, 33000 Bordeaux, France; Inserm, ISPED, Centre Inserm U1219, Bordeaux Population Health Center, 33000 Bordeaux, France; Service de médecine du travail, CHU de Bordeaux, 33000 Bordeaux, France.
| | - A Gruber
- ISPED, Équipe Santé Travail Environnement, Université de Bordeaux, 33000 Bordeaux, France; Inserm, ISPED, Centre Inserm U1219, Bordeaux Population Health Center, 33000 Bordeaux, France
| | - E Berteaud
- ISPED, Équipe Santé Travail Environnement, Université de Bordeaux, 33000 Bordeaux, France; Inserm, ISPED, Centre Inserm U1219, Bordeaux Population Health Center, 33000 Bordeaux, France; Service de médecine du travail, CHU de Bordeaux, 33000 Bordeaux, France
| | - C Carles
- ISPED, Équipe Santé Travail Environnement, Université de Bordeaux, 33000 Bordeaux, France; Inserm, ISPED, Centre Inserm U1219, Bordeaux Population Health Center, 33000 Bordeaux, France; Service de médecine du travail, CHU de Bordeaux, 33000 Bordeaux, France
| | - H Loiseau
- Service de neurochirurgie, CHU de Bordeaux, 33000 Bordeaux, France
| |
Collapse
|
37
|
Geng P, Ou J, Li J, Liao Y, Wang N, Xie G, Sa R, Liu C, Xiang L, Liang H. A Comprehensive Analysis of Influence ERCC Polymorphisms Confer on the Development of Brain Tumors. Mol Neurobiol 2015; 53:2705-14. [PMID: 26264164 DOI: 10.1007/s12035-015-9371-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 07/21/2015] [Indexed: 12/21/2022]
Abstract
Within DNA repair genes, there lie a number of single nucleotide polymorphisms that may impair protein function and attenuate DNA repair capability, resulting in genomic instability and individual predisposition to malignancies. The purpose of this study was to assess the previously reported inconsistent association of polymorphisms in ERCC1 (rs11615, rs3212986), ERCC2 (rs13181, rs1799793, rs238406), and ERCC5 (rs17655) with the development of brain tumors. In the present work, we carried out a comprehensive meta-analysis of results from all published data (5 data sets for rs11615, 7 for rs3212986, 11 for rs13181, 5 for rs1799793, 3 for rs238406, and 4 for rs17655) to evaluate risk of brain tumors contributed by the polymorphisms being investigated. Either the analytic method described by Mantel and Haenszel or that proposed by DerSimonian and Laird was properly used to summarize the risk estimates (OR and 95% CI). Data analyses were done with Stata version 12.0. Meta-analyses were performed for all polymorphisms, and only rs3212986 in the ERCC1 gene showed a significant association with glioma incidence. In the homozygote comparison, we found 1.26-fold elevated risk of glioma in relation to presence of the AA genotype (OR = 1.26, 95% CI = 1.05-1.52, P OR = 0.013, P heterogeneity = 0.849, I(2) = 0.0%). We also noted that individuals with the rs3212986-AA as compared to those with rs3212986-CC/CA had a 28% higher risk to develop glioma (OR = 1.28, 95% CI = 1.06-1.53, P OR = 0.008, Pheterogeneity = 0.808, I(2) = 0.0%). No major effects were observed for Caucasians or Asians in subgroup analysis by ethnicity. ERCC1 rs3212986 is a common single nucleotide polymorphism and may contribute toward individual susceptibility for glioma. Further research in this filed is required to verify the association obtained based on a relatively small number.
Collapse
Affiliation(s)
- Peiliang Geng
- Department of Oncology and Southwest Cancer Center, Southwest Hospital Third Military Medical University, 29 Gaotanyan Main Street, Chongqing, 400038, China
| | - Juanjuan Ou
- Department of Oncology and Southwest Cancer Center, Southwest Hospital Third Military Medical University, 29 Gaotanyan Main Street, Chongqing, 400038, China
| | - Jianjun Li
- Department of Oncology and Southwest Cancer Center, Southwest Hospital Third Military Medical University, 29 Gaotanyan Main Street, Chongqing, 400038, China
| | - Yunmei Liao
- Department of Oncology and Southwest Cancer Center, Southwest Hospital Third Military Medical University, 29 Gaotanyan Main Street, Chongqing, 400038, China
| | - Ning Wang
- Department of Oncology and Southwest Cancer Center, Southwest Hospital Third Military Medical University, 29 Gaotanyan Main Street, Chongqing, 400038, China
| | - Ganfeng Xie
- Department of Oncology and Southwest Cancer Center, Southwest Hospital Third Military Medical University, 29 Gaotanyan Main Street, Chongqing, 400038, China
| | - Rina Sa
- Department of Oncology and Southwest Cancer Center, Southwest Hospital Third Military Medical University, 29 Gaotanyan Main Street, Chongqing, 400038, China
| | - Chen Liu
- Department of Oncology and Southwest Cancer Center, Southwest Hospital Third Military Medical University, 29 Gaotanyan Main Street, Chongqing, 400038, China
| | - Lisha Xiang
- Department of Oncology and Southwest Cancer Center, Southwest Hospital Third Military Medical University, 29 Gaotanyan Main Street, Chongqing, 400038, China
| | - Houjie Liang
- Department of Oncology and Southwest Cancer Center, Southwest Hospital Third Military Medical University, 29 Gaotanyan Main Street, Chongqing, 400038, China.
| |
Collapse
|
38
|
Adel Fahmideh M, Lavebratt C, Schüz J, Röösli M, Tynes T, Grotzer MA, Johansen C, Kuehni CE, Lannering B, Prochazka M, Schmidt LS, Feychting M. CCDC26, CDKN2BAS, RTEL1 and TERT Polymorphisms in pediatric brain tumor susceptibility. Carcinogenesis 2015; 36:876-82. [PMID: 26014354 DOI: 10.1093/carcin/bgv074] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 05/20/2015] [Indexed: 03/25/2024] Open
Abstract
The role of genetic polymorphisms in pediatric brain tumor (PBT) etiology is poorly understood. We hypothesized that single nucleotide polymorphisms (SNPs) identified in genome-wide association studies (GWAS) on adult glioma would also be associated with PBT risk. The study is based on the Cefalo study, a population-based multicenter case-control study. Saliva DNA from 245 cases and 489 controls, aged 7-19 years at diagnosis/reference date, was extracted and genotyped for 29 SNPs reported by GWAS to be significantly associated with risk of adult glioma. Data were analyzed using unconditional logistic regression. Stratified analyses were performed for two histological subtypes: astrocytoma alone and the other tumor types combined. The results indicated that four SNPs, CDKN2BAS rs4977756 (p = 0.036), rs1412829 (p = 0.037), rs2157719 (p = 0.018) and rs1063192 (p = 0.021), were associated with an increased susceptibility to PBTs, whereas the TERT rs2736100 was associated with a decreased risk (p = 0.018). Moreover, the stratified analyses showed a decreased risk of astrocytoma associated with RTEL1 rs6089953, rs6010620 and rs2297440 (p trend = 0.022, p trend = 0.042, p trend = 0.029, respectively) as well as an increased risk of this subtype associated with RTEL1 rs4809324 (p trend = 0.033). In addition, SNPs rs10464870 and rs891835 in CCDC26 were associated with an increased risk of non-astrocytoma tumor subtypes (p trend = 0.009, p trend = 0.007, respectively). Our findings indicate that SNPs in CDKN2BAS, TERT, RTEL1 and CCDC26 may be associated with the risk of PBTs. Therefore, we suggest that pediatric and adult brain tumors might share common genetic risk factors and similar etiological pathways.
Collapse
Affiliation(s)
- Maral Adel Fahmideh
- Unit of Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Box 210, Stockholm SE-171 77, Sweden, Neurogenetics Unit, Department of Molecular Medicine and Surgery, Karolinska Institutet, and Center for Molecular Medicine, Karolinska University Hospital, Stockholm SE-171 76, Sweden, Section of Environment and Radiation, International Agency for Research on Cancer (IARC), 69372 Lyon CEDEX 08, France, Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel 4002, Switzerland, University of Basel, Basel 4003, Switzerland, The Cancer Registry of Norway, Oslo N-0304, Norway, National Institute of Occupational Health, Oslo NO-0033, Norway, Department of Oncology, University Children's Hospital of Zurich, Zurich 8091, Switzerland, Unit of Survivorship, The Danish Cancer Society Research Centre, Copenhagen DK-2100, Denmark, 5073 Oncology Clinic, Finsen Centre Rigshospitalet, University of Copenhagen, Copenhagen DK-2100, Denmark, Swiss Childhood Cancer Registry, Institute of Social and Preventive Medicine, University of Bern, Bern 3012, Switzerland, Department of Clinical Sciences, Pediatric Oncology, University of Gothenburg, SE 416 85 Gothenburg, Sweden and Department of Pediatric Oncology, University Hospital Rigshospitalet, Copenhagen DK-2100, Denmark
| | - Catharina Lavebratt
- Neurogenetics Unit, Department of Molecular Medicine and Surgery, Karolinska Institutet, and Center for Molecular Medicine, Karolinska University Hospital, Stockholm SE-171 76, Sweden
| | - Joachim Schüz
- Section of Environment and Radiation, International Agency for Research on Cancer (IARC), 69372 Lyon CEDEX 08, France
| | - Martin Röösli
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel 4002, Switzerland, University of Basel, Basel 4003, Switzerland
| | - Tore Tynes
- The Cancer Registry of Norway, Oslo N-0304, Norway, National Institute of Occupational Health, Oslo NO-0033, Norway
| | - Michael A Grotzer
- Department of Oncology, University Children's Hospital of Zurich, Zurich 8091, Switzerland
| | - Christoffer Johansen
- Unit of Survivorship, The Danish Cancer Society Research Centre, Copenhagen DK-2100, Denmark, 5073 Oncology Clinic, Finsen Centre Rigshospitalet, University of Copenhagen, Copenhagen DK-2100, Denmark
| | - Claudia E Kuehni
- Swiss Childhood Cancer Registry, Institute of Social and Preventive Medicine, University of Bern, Bern 3012, Switzerland
| | - Birgitta Lannering
- Department of Clinical Sciences, Pediatric Oncology, University of Gothenburg, SE 416 85 Gothenburg, Sweden and
| | - Michaela Prochazka
- Unit of Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Box 210, Stockholm SE-171 77, Sweden, Neurogenetics Unit, Department of Molecular Medicine and Surgery, Karolinska Institutet, and Center for Molecular Medicine, Karolinska University Hospital, Stockholm SE-171 76, Sweden, Section of Environment and Radiation, International Agency for Research on Cancer (IARC), 69372 Lyon CEDEX 08, France, Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel 4002, Switzerland, University of Basel, Basel 4003, Switzerland, The Cancer Registry of Norway, Oslo N-0304, Norway, National Institute of Occupational Health, Oslo NO-0033, Norway, Department of Oncology, University Children's Hospital of Zurich, Zurich 8091, Switzerland, Unit of Survivorship, The Danish Cancer Society Research Centre, Copenhagen DK-2100, Denmark, 5073 Oncology Clinic, Finsen Centre Rigshospitalet, University of Copenhagen, Copenhagen DK-2100, Denmark, Swiss Childhood Cancer Registry, Institute of Social and Preventive Medicine, University of Bern, Bern 3012, Switzerland, Department of Clinical Sciences, Pediatric Oncology, University of Gothenburg, SE 416 85 Gothenburg, Sweden and Department of Pediatric Oncology, University Hospital Rigshospitalet, Copenhagen DK-2100, Denmark
| | - Lisbeth S Schmidt
- Department of Pediatric Oncology, University Hospital Rigshospitalet, Copenhagen DK-2100, Denmark
| | - Maria Feychting
- Unit of Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Box 210, Stockholm SE-171 77, Sweden, Neurogenetics Unit, Department of Molecular Medicine and Surgery, Karolinska Institutet, and Center for Molecular Medicine, Karolinska University Hospital, Stockholm SE-171 76, Sweden, Section of Environment and Radiation, International Agency for Research on Cancer (IARC), 69372 Lyon CEDEX 08, France, Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel 4002, Switzerland, University of Basel, Basel 4003, Switzerland, The Cancer Registry of Norway, Oslo N-0304, Norway, National Institute of Occupational Health, Oslo NO-0033, Norway, Department of Oncology, University Children's Hospital of Zurich, Zurich 8091, Switzerland, Unit of Survivorship, The Danish Cancer Society Research Centre, Copenhagen DK-2100, Denmark, 5073 Oncology Clinic, Finsen Centre Rigshospitalet, University of Copenhagen, Copenhagen DK-2100, Denmark, Swiss Childhood Cancer Registry, Institute of Social and Preventive Medicine, University of Bern, Bern 3012, Switzerland, Department of Clinical Sciences, Pediatric Oncology, University of Gothenburg, SE 416 85 Gothenburg, Sweden and Department of Pediatric Oncology, University Hospital Rigshospitalet, Copenhagen DK-2100, Denmark
| |
Collapse
|
39
|
Jiang C, Shen F, Du J, Wang X, Su J, Liu Z, Huang X. DNA repair gene ERCC1 polymorphisms and glioma susceptibility among Chinese population: a meta-analysis. Int J Clin Exp Med 2015; 8:10248-10255. [PMID: 26379816 PMCID: PMC4565199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 06/10/2015] [Indexed: 06/05/2023]
Abstract
BACKGROUND Excision repair cross complementation group 1 (ERCC1) has been shown to be involved in the progression of glioma susceptibility. However, the results remain conflict. The aim of this study was to systematically review and evaluate the role of ERCC1 C118T and C8092A polymorphisms in glioma risk among Chinese population. METHODS Related case-control studies were searched in online electronic databases. Odds ratio (OR) with its 95% confidence interval (CI) were employed to calculate the extracted data. RESULTS Total seven articles were retrieved, including 4426 subjects (1926 were glioma patients and 2500 were matched controls). No significant heterogeneity was found between studies (I(2)=0%, P>0.01). Our results demonstrated that A allele and AA genotype of ERCC1 C8092A polymorphism have a positive association with increasing the risk of glioma in the fixed-effect model (A vs. C: OR=1.13, 95% CI=1.02-1.25, P=0.02; AA vs. CC: OR=1.29, 95% CI=1.04-1.61, P=0.02; AA vs. CA+CC: OR=1.25, 95% CI=1.01-1.55, P=0.04). However, no significant relationship was found between C118T variant and glioma susceptibility. CONCLUSIONS Our results indicated that ERCC1 C8092A, not C118T polymorphism might be a biomarker for patients with glioma among Chinese population. Future studies with more ethnicities are needed to explore the precise association.
Collapse
Affiliation(s)
- Chunming Jiang
- Department of Pediatrics, Affiliated Hangzhou Hospital of Nanjing Medical University, Hangzhou First People’s HospitalHangzhou 301103, Zhejiang, P. R. China
| | - Fang Shen
- Department of psychiatry, Tongde Hospital of Zhejiang ProvinceHangzhou 301103, Zhejiang, P. R. China
| | - Jianmin Du
- Department of Pediatrics, Affiliated Hangzhou Hospital of Nanjing Medical University, Hangzhou First People’s HospitalHangzhou 301103, Zhejiang, P. R. China
| | - Xiaohua Wang
- Nanjing First Hospital, Nanjing Medical UniversityNanjing 210029, Jiangsu, P. R. China
| | - Jin Su
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical UniversityNanjing 210029, Jiangsu, P. R. China
| | - Zhanli Liu
- Department of Pediatrics, Affiliated Hangzhou Hospital of Nanjing Medical University, Hangzhou First People’s HospitalHangzhou 301103, Zhejiang, P. R. China
| | - Xianmei Huang
- Department of Pediatrics, Affiliated Hangzhou Hospital of Nanjing Medical University, Hangzhou First People’s HospitalHangzhou 301103, Zhejiang, P. R. China
| |
Collapse
|
40
|
Song YZ, Han FJ, Liu M, Xia CC, Shi WY, Dong LH. Association between Single Nucleotide Polymorphisms in XRCC3 and Radiation-Induced Adverse Effects on Normal Tissue: A Meta-Analysis. PLoS One 2015; 10:e0130388. [PMID: 26091483 PMCID: PMC4474802 DOI: 10.1371/journal.pone.0130388] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 05/20/2015] [Indexed: 01/26/2023] Open
Abstract
The X-ray repair cross-complementing group 3 (XRCC3) protein plays an important role in the repair of DNA double-strand breaks. The relationship between XRCC3 polymorphisms and the risk of radiation-induced adverse effects on normal tissue remains inconclusive. Thus, we performed a meta-analysis to elucidate the association between XRCC3 polymorphisms and radiation-induced adverse effects on normal tissue. All eligible studies up to December 2014 were identified through a search of the PubMed, Embase and Web of Science databases. Seventeen studies involving 656 cases and 2193 controls were ultimately included in this meta-analysis. The pooled odds ratios (ORs) with corresponding 95% confidence intervals (CIs) were calculated to evaluate the association between XRCC3 polymorphisms and the risk of radiation-induced normal tissue adverse effects. We found that the XRCC3 p.Thr241Met (rs861539) polymorphism was significantly associated with early adverse effects induced by radiotherapy (OR = 1.99, 95%CI: 1.31-3.01, P = 0.001). A positive association lacking statistical significance with late adverse effects was also identified (OR = 1.28, 95%CI: 0.97-1.68, P = 0.08). In addition, the rs861539 polymorphism was significantly correlated with a higher risk of adverse effects induced by head and neck area irradiation (OR = 2.41, 95%CI: 1.49-3.89, p = 0.0003) and breast irradiation (OR = 1.41, 95%CI: 1.02-1.95, p = 0.04), whereas the correlation was not significant for lung irradiation or pelvic irradiation. Furthermore, XRCC3 rs1799794 polymorphism may have a protective effect against late adverse effects induced by radiotherapy (OR = 0.47, 95%CI: 0.26-0.86, P = 0.01). Well-designed large-scale clinical studies are required to further validate our results.
Collapse
Affiliation(s)
- Yu-Zhe Song
- Department of Radiation Oncology, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Fu-Jun Han
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Min Liu
- Department of Radiation Oncology, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Cheng-Cheng Xia
- Department of Radiation Oncology, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Wei-Yan Shi
- Department of Radiation Oncology, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Li-Hua Dong
- Department of Radiation Oncology, the First Hospital of Jilin University, Changchun, Jilin, China
- * E-mail:
| |
Collapse
|
41
|
Liu YC, Wang YZ. Role of Yes-associated protein 1 in gliomas: pathologic and therapeutic aspects. Tumour Biol 2015; 36:2223-7. [PMID: 25750037 DOI: 10.1007/s13277-015-3297-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 02/27/2015] [Indexed: 11/26/2022] Open
Abstract
The activation of proline-rich phosphoprotein Yes-associated protein 1 (YAP1) possesses a possible link between stem/progenitor cells, organ size, and cancer. YAP1 has been indicated as an oncoprotein, and overexpression of YAP1 is reported in many human brain tumors, including infiltrating gliomas. During normal brain development, the neurofibromatosis 2 (NF2) protein suppresses YAP1 activity in neural progenitor cells to promote guidepost cell differentiation, but loss of NF2 causes elevating YAP1 activity in midline neural progenitors, which disrupts guidepost formation. Overexpression of endogenous CD44 (cancer stem cell marker) promotes phosphorylation/inactivation of NF2, and upregulates YAP1 expression and leads to cancer cell resistance in glioblastoma. The hippo pathway is also related to the YAP1 action. However, the mechanism of YAP1 action in glioma is still far from clear understanding. Advances in clinical management based on an improved understanding of the function of YAP1 may help to serve as a molecular target in glioma therapeutics. Knockdown of YAP1 by shRNA technology has been shown to reduce glioma in vitro; however, clinical implications are still under investigation. YAP1 can be used as a diagnostic marker for gliomas to monitor the disease status and may help to evaluate its treatment effects. More functional experiments are needed to support the direct roles of YAP1 on gliomas at molecular and cellular levels.
Collapse
Affiliation(s)
- Yong-Chang Liu
- Fourth Department of Neurosurgery, Cangzhou Central Hospital, Cangzhou, 061001, China,
| | | |
Collapse
|
42
|
Rao KS, Paul A, Kumar ASA, Umamaheswaran G, Dubashi B, Gunaseelan K, Dkhar SA. Allele and genotype distributions of DNA repair gene polymorphisms in South Indian healthy population. BIOMARKERS IN CANCER 2014; 6:29-35. [PMID: 25520562 PMCID: PMC4259864 DOI: 10.4137/bic.s19681] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Revised: 11/11/2014] [Accepted: 11/13/2014] [Indexed: 12/11/2022]
Abstract
Various DNA repair pathways protect the structural and chemical integrity of the human genome from environmental and endogenous threats. Polymorphisms of genes encoding the proteins involved in DNA repair have been found to be associated with cancer risk and chemotherapeutic response. In this study, we aim to establish the normative frequencies of DNA repair genes in South Indian healthy population and compare with HapMap populations. Genotyping was done on 128 healthy volunteers from South India, and the allele and genotype distributions were established. The minor allele frequency of Xeroderma pigmentosum group A (XPA) G23A, Excision repair cross-complementing 2 (ERCC2)/Xeroderma pigmentosum group D (XPD) Lys751Gln, Xeroderma pigmentosum group G (XPG) His46His, XPG Asp1104His, and X-ray repair cross-complementing group 1 (XRCC1) Arg399Gln polymorphisms were 49.2%, 36.3%, 48.0%, 23.0%, and 34.0% respectively. Ethnic variations were observed in the frequency distribution of these polymorphisms between the South Indians and other HapMap populations. The present work forms the groundwork for cancer association studies and biomarker identification for treatment response and prognosis.
Collapse
Affiliation(s)
- Katiboina Srinivasa Rao
- Department of Pharmacology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry, India
| | - Abialbon Paul
- Department of Pharmacology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry, India
| | - Annan Sudarsan Arun Kumar
- Department of Pharmacology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry, India
| | - Gurusamy Umamaheswaran
- Department of Pharmacology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry, India
| | - Biswajit Dubashi
- Department of Medical Oncology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry, India
| | - Karunanithi Gunaseelan
- Department of Radiotherapy, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry, India
| | - Steven Aibor Dkhar
- Department of Pharmacology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry, India
| |
Collapse
|