51
|
Sharafeldin N, Slattery ML, Liu Q, Franco-Villalobos C, Caan BJ, Potter JD, Yasui Y. Multiple Gene-Environment Interactions on the Angiogenesis Gene-Pathway Impact Rectal Cancer Risk and Survival. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2017; 14:ijerph14101146. [PMID: 28956832 PMCID: PMC5664647 DOI: 10.3390/ijerph14101146] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 09/06/2017] [Accepted: 09/23/2017] [Indexed: 12/25/2022]
Abstract
Characterization of gene-environment interactions (GEIs) in cancer is limited. We aimed at identifying GEIs in rectal cancer focusing on a relevant biologic process involving the angiogenesis pathway and relevant environmental exposures: cigarette smoking, alcohol consumption, and animal protein intake. We analyzed data from 747 rectal cancer cases and 956 controls from the Diet, Activity and Lifestyle as a Risk Factor for Rectal Cancer study. We applied a 3-step analysis approach: first, we searched for interactions among single nucleotide polymorphisms on the pathway genes; second, we searched for interactions among the genes, both steps using Logic regression; third, we examined the GEIs significant at the 5% level using logistic regression for cancer risk and Cox proportional hazards models for survival. Permutation-based test was used for multiple testing adjustment. We identified 8 significant GEIs associated with risk among 6 genes adjusting for multiple testing: TNF (OR = 1.85, 95% CI: 1.10, 3.11), TLR4 (OR = 2.34, 95% CI: 1.38, 3.98), and EGR2 (OR = 2.23, 95% CI: 1.04, 4.78) with smoking; IGF1R (OR = 1.69, 95% CI: 1.04, 2.72), TLR4 (OR = 2.10, 95% CI: 1.22, 3.60) and EGR2 (OR = 2.12, 95% CI: 1.01, 4.46) with alcohol; and PDGFB (OR = 1.75, 95% CI: 1.04, 2.92) and MMP1 (OR = 2.44, 95% CI: 1.24, 4.81) with protein. Five GEIs were associated with survival at the 5% significance level but not after multiple testing adjustment: CXCR1 (HR = 2.06, 95% CI: 1.13, 3.75) with smoking; and KDR (HR = 4.36, 95% CI: 1.62, 11.73), TLR2 (HR = 9.06, 95% CI: 1.14, 72.11), EGR2 (HR = 2.45, 95% CI: 1.42, 4.22), and EGFR (HR = 6.33, 95% CI: 1.95, 20.54) with protein. GEIs between angiogenesis genes and smoking, alcohol, and animal protein impact rectal cancer risk. Our results support the importance of considering the biologic hypothesis to characterize GEIs associated with cancer outcomes.
Collapse
Affiliation(s)
- Noha Sharafeldin
- School of Public Health, University of Alberta, Edmonton, AB T6G 2R3, Canada.
- Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Martha L Slattery
- Department of Internal Medicine, University of Utah Health Sciences Center, Salt Lake City, UT 84132, USA.
| | - Qi Liu
- School of Public Health, University of Alberta, Edmonton, AB T6G 2R3, Canada.
| | | | - Bette J Caan
- Division of Research, Kaiser Permanente Medical Care Program, Oakland, CA 94612, USA.
| | - John D Potter
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA 98195, USA.
- Centre for Public Health Research, Massey University, P.O. Box 756, Wellington 6140, New Zealand.
| | - Yutaka Yasui
- School of Public Health, University of Alberta, Edmonton, AB T6G 2R3, Canada.
- Department of Epidemiology & Cancer Control, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
52
|
Takahashi Y, Sugimachi K, Yamamoto K, Niida A, Shimamura T, Sato T, Watanabe M, Tanaka J, Kudo S, Sugihara K, Hase K, Kusunoki M, Yamada K, Shimada Y, Moriya Y, Suzuki Y, Miyano S, Mori M, Mimori K. Japanese genome-wide association study identifies a significant colorectal cancer susceptibility locus at chromosome 10p14. Cancer Sci 2017; 108:2239-2247. [PMID: 28869801 PMCID: PMC5665761 DOI: 10.1111/cas.13391] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 08/19/2017] [Accepted: 08/29/2017] [Indexed: 12/25/2022] Open
Abstract
Genome‐wide association studies are a powerful tool for searching for disease susceptibility loci. Several studies identifying single nucleotide polymorphisms (SNP) connected intimately to the onset of colorectal cancer (CRC) have been published, but there are few reports of genome‐wide association studies in Japan. To identify genetic variants that modify the risk of CRC oncogenesis, especially in the Japanese population, we performed a multi‐stage genome‐wide association study using a large number of samples: 1846 CRC cases and 2675 controls. We identified 4 SNP (rs7912831, rs4749812, rs7898455 and rs10905453) in chromosome region 10p14 associated with CRC; however, there are no coding or non‐coding genes within this region of fairly extensive linkage disequilibrium (a 500‐kb block) on 10p14. Our study revealed that the 10p14 locus is significantly correlated with susceptibility to CRC in the Japanese population, in accordance with the results of multiple studies in other races.
Collapse
Affiliation(s)
- Yusuke Takahashi
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan.,Department of Gastroenterological Surgery, Osaka University, Suita, Japan
| | - Keishi Sugimachi
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan.,Department of Hepatobiliary-pancreatic surgery, National Kyushu Cancer Center, Fukuoka, Japan
| | - Ken Yamamoto
- Department of Medical Biochemistry, Kurume University, Kurume, Japan
| | - Atsushi Niida
- Laboratory of DNA Information Analysis, Human Genome Center Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Teppei Shimamura
- Laboratory of DNA Information Analysis, Human Genome Center Institute of Medical Science, University of Tokyo, Tokyo, Japan.,Division of System Biology, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Tetsuya Sato
- Division of Bioinformatics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | | | - Junichi Tanaka
- Digestive Disease Center, Northern Yokohama Hospital, Showa University, Yokohama, Japan
| | - Shinei Kudo
- Digestive Disease Center, Northern Yokohama Hospital, Showa University, Yokohama, Japan
| | - Kenichi Sugihara
- Department of Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kazuo Hase
- Department of Surgery, National Defense University, Tokorozawa, Japan
| | | | | | - Yasuhiro Shimada
- Department of Surgery and Digestive Tract Medicine, National Cancer Center, Tokyo, Japan
| | - Yoshihiro Moriya
- Department of Surgery and Digestive Tract Medicine, National Cancer Center, Tokyo, Japan
| | - Yutaka Suzuki
- Department of Medical Genome Sciences, Graduate School of Frontier Sciences, University of Tokyo, Kashiwa, Japan
| | - Satoru Miyano
- Laboratory of DNA Information Analysis, Human Genome Center Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Masaki Mori
- Department of Gastroenterological Surgery, Osaka University, Suita, Japan
| | - Koshi Mimori
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| |
Collapse
|
53
|
Grosso G, Micek A, Godos J, Pajak A, Sciacca S, Galvano F, Boffetta P. Health risk factors associated with meat, fruit and vegetable consumption in cohort studies: A comprehensive meta-analysis. PLoS One 2017; 12:e0183787. [PMID: 28850610 PMCID: PMC5574618 DOI: 10.1371/journal.pone.0183787] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Accepted: 08/11/2017] [Indexed: 12/17/2022] Open
Abstract
The aim of this study was to perform a meta-analysis to test the association between red, processed, and total meat, as well as fruit and vegetable consumption, and selected health risk factors, including body weight status, smoking habit, physical activity level, level of education, and alcohol drinking in cohort studies on non-communicable disease. A systematic search of electronic databases was performed to identify relevant articles published up to March 2017. In a two-stage approach, frequency-weighted linear regression coefficients were first calculated for each variable, and then combined across studies through meta-regression. Ninety-eight studies including 20 on red meat, 6 on processed meat, 12 on total meat, 37 on fruit and vegetable combined, 21 on fruit and 24 on vegetable consumption were analyzed. Intake of red meat was positively associated with BMI, percentage of overweight and obese, low physical activity, and current and ever smoking and inversely associated with percentage of non-smokers and high physically active individuals. Similar associations were found for red meat were found, although based on fewer data. Intake of fruits and vegetables was positively associated with prevalence of non-smokers, high education and high physical activity, and similar results were found when examining fruit and vegetable consumption separately. Stratification by geographical area revealed that some associations were stronger in US rather than European or Asian cohorts. In conclusions, the distribution of health risk factors associated with high meat and fruit/vegetable consumption may differ from those of low-consumers. Some of these differences may mediate, confound, or modify the relation between diet and non-communicable disease risk.
Collapse
Affiliation(s)
- Giuseppe Grosso
- Integrated Cancer Registry of Catania-Messina-Siracusa-Enna, Azienda Ospedaliera Policlinico-Universitaria “Vittorio Emanuele”, Catania, Italy
- The Need for Nutrition Education/Innovation Programme (NNEdPro), University of Cambridge, Cambridge, United Kingdom
- * E-mail:
| | - Agnieszka Micek
- Department of Epidemiology and Population Studies, Jagiellonian University Medical College, Krakow, Poland
| | - Justyna Godos
- Integrated Cancer Registry of Catania-Messina-Siracusa-Enna, Azienda Ospedaliera Policlinico-Universitaria “Vittorio Emanuele”, Catania, Italy
| | - Andrzej Pajak
- Department of Epidemiology and Population Studies, Jagiellonian University Medical College, Krakow, Poland
| | - Salvatore Sciacca
- Integrated Cancer Registry of Catania-Messina-Siracusa-Enna, Azienda Ospedaliera Policlinico-Universitaria “Vittorio Emanuele”, Catania, Italy
| | - Fabio Galvano
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Paolo Boffetta
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| |
Collapse
|
54
|
Qin N, Wang C, Lu Q, Huang T, Zhu M, Wang L, Yu F, Huang M, Jiang Y, Dai J, Ma H, Jin G, Wu C, Lin D, Shen H, Hu Z. A cis-eQTL genetic variant of the cancer-testis gene CCDC116 is associated with risk of multiple cancers. Hum Genet 2017; 136:987-997. [PMID: 28653172 DOI: 10.1007/s00439-017-1827-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 06/19/2017] [Indexed: 11/29/2022]
Abstract
Recent studies have found that cancer-testis (CT) genes, which are expressed predominantly in germ and cancer cells, may be candidate cancer drivers. Because of their crucial roles, genetic variants in these genes may contribute to the development of cancer. Here, we systematically evaluated associations of common variants in CT genes and their promoters for the risk of lung cancer in our initial GWAS (2331 cases and 3077 controls), followed by in silico replication using additional 10,512 lung cancer cases and 9562 controls. We found a significant association between rs3747093 located in the CCDC116 promoter and lung cancer risk (OR = 0.91, P meta = 7.81 × 10-6). Although CCDC116 was expressed at lower levels in somatic tissues compared to the testis, the protective allele A of rs3747093 was associated with decreased CCDC116 expression in many normal tissues, including the lung (P = 8.1 × 10-13). We subsequently genotyped this variant in another four commonly diagnosed cancers (gastric, esophageal, colorectal, and breast cancers), as we found expression quantitative trait locus (eQTL) signals for rs3747093 and CCDC116 in their corresponding normal tissues. Interestingly, we observed consistent associations between rs3747093 and multiple cancers (gastric cancer: OR = 0.85, P = 2.21 × 10-4; esophageal cancer: OR = 0.91, P = 2.57 × 10-2; colorectal cancer: OR = 0.80, P = 1.85 × 10-6; and breast cancer: OR = 0.87, P = 1.55 × 10-3). Taken together, the A allele of rs3747093 showed significant protective effects on cancer risk (OR = 0.88, P pool = 6.52 × 10-13) in an Asian population. Moreover, our findings suggest that low abundance expression of CT genes in normal tissues may also contribute to tumorigenesis, providing a new mechanism of CT genes in the development of cancer.
Collapse
Affiliation(s)
- Na Qin
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 210029, China.,Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Cheng Wang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 210029, China.,Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Qun Lu
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Tongtong Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Meng Zhu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 210029, China.,Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Lihua Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Fei Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Mingtao Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Yue Jiang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 210029, China.,Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Juncheng Dai
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 210029, China.,Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Hongxia Ma
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 210029, China.,Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Guangfu Jin
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 210029, China.,Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Chen Wu
- State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Dongxin Lin
- State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Hongbing Shen
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 210029, China.,Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Zhibin Hu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 210029, China. .,Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China. .,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
55
|
Huang B, Yang H, Cheng X, Wang D, Fu S, Shen W, Zhang Q, Zhang L, Xue Z, Li Y, Da Y, Yang Q, Li Z, Liu L, Qiao L, Kong Y, Yao Z, Zhao P, Li M, Zhang R. tRF/miR-1280 Suppresses Stem Cell-like Cells and Metastasis in Colorectal Cancer. Cancer Res 2017; 77:3194-3206. [PMID: 28446464 DOI: 10.1158/0008-5472.can-16-3146] [Citation(s) in RCA: 184] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 04/05/2017] [Accepted: 04/18/2017] [Indexed: 12/27/2022]
Abstract
Several studies have shown that tRNAs can be enzymatically cleaved to generate distinct classes of tRNA-derived fragments (tRF). Here, we report that tRF/miR-1280, a 17-bp fragment derived from tRNALeu and pre-miRNA, influences Notch signaling pathways that support the function of cancer stem-like cells (CSC) in colorectal cancer progression. tRF/miR-1280 expression was decreased in human specimens of colorectal cancer. Ectopic expression of tRF/miR-1280 reduced cell proliferation and colony formation, whereas its suppression reversed these effects. Mechanistic investigations implicated the Notch ligand JAG2 as a direct target of tRF/miR-1280 binding through which it reduced tumor formation and metastasis. Notably, tRF/miR-1280-mediated inactivation of Notch signaling suppressed CSC phenotypes, including by direct transcriptional repression of the Gata1/3 and miR-200b genes. These results were consistent with findings of decreased levels of miR-200b and elevated levels of JAG2, Gata1, Gata3, Zeb1, and Suz12 in colorectal cancer tissue specimens. Taken together, our results established that tRF/miR-1280 suppresses colorectal cancer growth and metastasis by repressing Notch signaling pathways that support CSC phenotypes. Furthermore, they provide evidence that functionally active miRNA can be derived from tRNA, offering potential biomarker and therapeutic uses. Cancer Res; 77(12); 3194-206. ©2017 AACR.
Collapse
Affiliation(s)
- Bingqing Huang
- Department of Immunology, Research Center of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Medical University, Tianjin, China.,Department of Pathology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Huipeng Yang
- Department of Immunology, Research Center of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Medical University, Tianjin, China
| | - Xixi Cheng
- Department of Immunology, Research Center of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Medical University, Tianjin, China
| | - Dan Wang
- Department of Immunology, Research Center of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Medical University, Tianjin, China
| | - Shuyu Fu
- Department of Immunology, Research Center of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Medical University, Tianjin, China
| | - Wencui Shen
- Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Vision Science, Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Qi Zhang
- Department of Immunology, Research Center of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Medical University, Tianjin, China
| | - Lijuan Zhang
- Department of Immunology, Research Center of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Medical University, Tianjin, China
| | - Zhenyi Xue
- Department of Immunology, Research Center of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Medical University, Tianjin, China
| | - Yan Li
- Department of Immunology, Research Center of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Medical University, Tianjin, China
| | - Yurong Da
- Department of Immunology, Research Center of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Medical University, Tianjin, China
| | - Qing Yang
- Department of Colorectal Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Department of Genitourinary Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Zesong Li
- Shenzhen Key Laboratory of Genitourinary Tumor, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Li Liu
- Department of Radiology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Liang Qiao
- Storr Liver Centre, Westmead Millennium Institute for Medical Research, The University of Sydney at Westmead Hospital, Westmead, New South Wales, Australia
| | - Ying Kong
- Department of Biochemistry and Molecular Biology, Liaoning Key Lab of Glycobiology and Glycoengn, Dalian Medical University, Dalian, China
| | - Zhi Yao
- Department of Immunology, Research Center of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Medical University, Tianjin, China
| | - Peng Zhao
- Department of Colorectal Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China. .,Department of Genitourinary Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Min Li
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma.,Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Rongxin Zhang
- Department of Immunology, Research Center of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Medical University, Tianjin, China. .,Laboratory of Immunology and Inflammation, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
56
|
Dietary intake alters gene expression in colon tissue: possible underlying mechanism for the influence of diet on disease. Pharmacogenet Genomics 2017; 26:294-306. [PMID: 26959716 PMCID: PMC4853256 DOI: 10.1097/fpc.0000000000000217] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Supplemental Digital Content is available in the text. Background Although the association between diet and disease is well documented, the biologic mechanisms involved have not been entirely elucidated. In this study, we evaluate how dietary intake influences gene expression to better understand the underlying mechanisms through which diet operates. Methods We used data from 144 individuals who had comprehensive dietary intake and gene expression data from RNAseq using normal colonic mucosa. Using the DESeq2 statistical package, we identified genes that showed statistically significant differences in expression between individuals in high-intake and low-intake categories for several dietary variables of interest adjusting for age and sex. We examined total calories, total fats, vegetable protein, animal protein, carbohydrates, trans-fatty acids, mutagen index, red meat, processed meat, whole grains, vegetables, fruits, fiber, folate, dairy products, calcium, and prudent and western dietary patterns. Results Using a false discovery rate of less than 0.1, meat-related foods were statistically associated with 68 dysregulated genes, calcium with three dysregulated genes, folate with four dysregulated genes, and nonmeat-related foods with 65 dysregulated genes. With a more stringent false discovery rate of less than 0.05, there were nine meat-related dysregulated genes and 23 nonmeat-related genes. Ingenuity pathway analysis identified three major networks among genes identified as dysregulated with respect to meat-related dietary variables and three networks among genes identified as dysregulated with respect to nonmeat-related variables. The top networks (Ingenuity Pathway Analysis network score >30) associated with meat-related genes were (i) cancer, organismal injury, and abnormalities, tumor morphology, and (ii) cellular function and maintenance, cellular movement, cell death, and survival. Among genes related to nonmeat consumption variables, the top networks were (i) hematological system development and function, nervous system development and function, tissue morphology and (ii) connective tissue disorders, organismal injury, and abnormalities. Conclusion Several dietary factors were associated with gene expression in our data. These findings provide insight into the possible mechanisms by which diet may influence disease processes.
Collapse
|
57
|
Microbial carcinogenic toxins and dietary anti-cancer protectants. Cell Mol Life Sci 2017; 74:2627-2643. [PMID: 28238104 PMCID: PMC5487888 DOI: 10.1007/s00018-017-2487-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: 11/03/2016] [Revised: 02/03/2017] [Accepted: 02/06/2017] [Indexed: 12/16/2022]
Abstract
Several toxins are known which account for the ability of some bacteria to initiate or promote carcinogenesis. These ideas are summarised and evidence is discussed for more specific mechanisms involving chymotrypsin and the bacterial chymotryptic enzyme subtilisin. Subtilisin and Bacillus subtilis are present in the gut and environment and both are used commercially in agriculture, livestock rearing and meat processing. The enzymes deplete cells of tumour suppressors such as deleted in colorectal cancer (DCC) and neogenin, so their potential presence in the food chain might represent an important link between diet and cancer. Over-eating increases secretion of chymotrypsin which is absorbed from the gut and could contribute to several forms of cancer linked to obesity. Inhibition of these serine proteases by Bowman–Birk inhibitors in fruit and vegetables could account for some of the protective effects of a plant-rich diet. These interactions represent previously unknown non-genetic mechanisms for the modification of tumour suppressor proteins and provide a plausible explanation contributing to both the pro-oncogenic effects of meat products and the protective activity of a plant-rich diet. The data suggest that changes to farming husbandry and food processing methods to remove these sources of extrinsic proteases might significantly reduce the incidence of several cancers.
Collapse
|
58
|
Cohen AJ, Saiakhova A, Corradin O, Luppino JM, Lovrenert K, Bartels CF, Morrow JJ, Mack SC, Dhillon G, Beard L, Myeroff L, Kalady MF, Willis J, Bradner JE, Keri RA, Berger NA, Pruett-Miller SM, Markowitz SD, Scacheri PC. Hotspots of aberrant enhancer activity punctuate the colorectal cancer epigenome. Nat Commun 2017; 8:14400. [PMID: 28169291 PMCID: PMC5309719 DOI: 10.1038/ncomms14400] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 12/22/2016] [Indexed: 12/12/2022] Open
Abstract
In addition to mutations in genes, aberrant enhancer element activity at non-coding regions of the genome is a key driver of tumorigenesis. Here, we perform epigenomic enhancer profiling of a cohort of more than forty genetically diverse human colorectal cancer (CRC) specimens. Using normal colonic crypt epithelium as a comparator, we identify enhancers with recurrently gained or lost activity across CRC specimens. Of the enhancers highly recurrently activated in CRC, most are constituents of super enhancers, are occupied by AP-1 and cohesin complex members, and originate from primed chromatin. Many activate known oncogenes, and CRC growth can be mitigated through pharmacologic inhibition or genome editing of these loci. Nearly half of all GWAS CRC risk loci co-localize to recurrently activated enhancers. These findings indicate that the CRC epigenome is defined by highly recurrent epigenetic alterations at enhancers which activate a common, aberrant transcriptional programme critical for CRC growth and survival.
Collapse
Affiliation(s)
- Andrea J. Cohen
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, 10900 Euclid Ave, Cleveland, Ohio 44106, USA
| | - Alina Saiakhova
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, 10900 Euclid Ave, Cleveland, Ohio 44106, USA
| | - Olivia Corradin
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, 10900 Euclid Ave, Cleveland, Ohio 44106, USA
- Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge, Massachusetts 02142, USA
| | - Jennifer M. Luppino
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, 10900 Euclid Ave, Cleveland, Ohio 44106, USA
| | - Katreya Lovrenert
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, 10900 Euclid Ave, Cleveland, Ohio 44106, USA
| | - Cynthia F. Bartels
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, 10900 Euclid Ave, Cleveland, Ohio 44106, USA
| | - James J. Morrow
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, 10900 Euclid Ave, Cleveland, Ohio 44106, USA
- Department of Pathology, Case Western Reserve University School of Medicine, 10900 Euclid Ave, Cleveland, Ohio 44106, USA
| | - Stephen C. Mack
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, Ohio 44195, USA
| | - Gursimran Dhillon
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, 10900 Euclid Ave, Cleveland, Ohio 44106, USA
| | - Lydia Beard
- Case Comprehensive Cancer Center, Case Western Reserve University, 10900 Euclid Ave, Cleveland, Ohio 44106, USA
| | - Lois Myeroff
- Case Comprehensive Cancer Center, Case Western Reserve University, 10900 Euclid Ave, Cleveland, Ohio 44106, USA
| | - Matthew F. Kalady
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, Ohio 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, 10900 Euclid Ave, Cleveland, Ohio 44106, USA
- Department of Colorectal Surgery, Cleveland Clinic Foundation, 9500 Euclid Ave, Cleveland, Ohio 44195, USA
| | - Joseph Willis
- Department of Pathology, Case Western Reserve University School of Medicine, 10900 Euclid Ave, Cleveland, Ohio 44106, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, 10900 Euclid Ave, Cleveland, Ohio 44106, USA
- Department of Medicine, University Hospitals Cleveland Medical Center, 11100 Euclid Ave, Cleveland, Ohio 44106, USA
| | - James E. Bradner
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, Massachusetts 02215, USA
- Department of Medicine, Harvard Medical School, 25 Shattuck St, Boston, Massachusetts 02115, USA
| | - Ruth A. Keri
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, 10900 Euclid Ave, Cleveland, Ohio 44106, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, 10900 Euclid Ave, Cleveland, Ohio 44106, USA
- Department of Pharmacology, Case Western Reserve University School of Medicine, 10900 Euclid Ave, Cleveland, Ohio 44106, USA
| | - Nathan A. Berger
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, 10900 Euclid Ave, Cleveland, Ohio 44106, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, 10900 Euclid Ave, Cleveland, Ohio 44106, USA
- Department of Medicine, University Hospitals Cleveland Medical Center, 11100 Euclid Ave, Cleveland, Ohio 44106, USA
| | - Shondra M. Pruett-Miller
- Genome Engineering and iPSC Center, Department of Genetics, Washington University, 4515 McKinley Building, St. Louis, Missouri 63110, USA
| | - Sanford D. Markowitz
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, 10900 Euclid Ave, Cleveland, Ohio 44106, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, 10900 Euclid Ave, Cleveland, Ohio 44106, USA
- Department of Medicine, University Hospitals Cleveland Medical Center, 11100 Euclid Ave, Cleveland, Ohio 44106, USA
| | - Peter C. Scacheri
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, 10900 Euclid Ave, Cleveland, Ohio 44106, USA
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, Ohio 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, 10900 Euclid Ave, Cleveland, Ohio 44106, USA
| |
Collapse
|
59
|
Hahn MM, de Voer RM, Hoogerbrugge N, Ligtenberg MJL, Kuiper RP, van Kessel AG. The genetic heterogeneity of colorectal cancer predisposition - guidelines for gene discovery. Cell Oncol (Dordr) 2016; 39:491-510. [PMID: 27279102 PMCID: PMC5121185 DOI: 10.1007/s13402-016-0284-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2016] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is a cumulative term applied to a clinically and genetically heterogeneous group of neoplasms that occur in the bowel. Based on twin studies, up to 45 % of the CRC cases may involve a heritable component. Yet, only in 5-10 % of these cases high-penetrant germline mutations are found (e.g. mutations in APC and DNA mismatch repair genes) that result in a familial aggregation and/or an early onset of the disease. Genome-wide association studies have revealed that another ~5 % of the CRC cases may be explained by a cumulative effect of low-penetrant risk factors. Recent attempts to identify novel genetic factors using whole exome and whole genome sequencing has proven to be difficult since the remaining, yet to be discovered, high penetrant CRC predisposing genes appear to be rare. In addition, most of the moderately penetrant candidate genes identified so far have not been confirmed in independent cohorts. Based on literature examples, we here discuss how careful patient and cohort selection, candidate gene and variant selection, and corroborative evidence may be employed to facilitate the discovery of novel CRC predisposing genes. CONCLUSIONS The picture emerges that the genetic predisposition to CRC is heterogeneous, involving complex interplays between common and rare (inter)genic variants with different penetrances. It is anticipated, however, that the use of large clinically well-defined patient and control datasets, together with improved functional and technical possibilities, will yield enough power to unravel this complex interplay and to generate accurate individualized estimates for the risk to develop CRC.
Collapse
Affiliation(s)
- M M Hahn
- Department of Human Genetics, Radboud Institute of Molecular Life Sciences, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - R M de Voer
- Department of Human Genetics, Radboud Institute of Molecular Life Sciences, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - N Hoogerbrugge
- Department of Human Genetics, Radboud Institute of Molecular Life Sciences, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - M J L Ligtenberg
- Department of Human Genetics, Radboud Institute of Molecular Life Sciences, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - R P Kuiper
- Department of Human Genetics, Radboud Institute of Molecular Life Sciences, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.
| | - A Geurts van Kessel
- Department of Human Genetics, Radboud Institute of Molecular Life Sciences, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| |
Collapse
|
60
|
Abstract
Colorectal cancer is one of the so-called westernized diseases and the second leading cause of cancer death worldwide. On the basis of global epidemiological and scientific studies, evidence suggests that the risk of colorectal cancer is increased by processed and unprocessed meat consumption but suppressed by fibre, and that food composition affects colonic health and cancer risk via its effects on colonic microbial metabolism. The gut microbiota can ferment complex dietary residues that are resistant to digestion by enteric enzymes. This process provides energy for the microbiota but culminates in the release of short-chain fatty acids including butyrate, which are utilized for the metabolic needs of the colon and the body. Butyrate has a remarkable array of colonic health-promoting and antineoplastic properties: it is the preferred energy source for colonocytes, it maintains mucosal integrity and it suppresses inflammation and carcinogenesis through effects on immunity, gene expression and epigenetic modulation. Protein residues and fat-stimulated bile acids are also metabolized by the microbiota to inflammatory and/or carcinogenic metabolites, which increase the risk of neoplastic progression. This Review will discuss the mechanisms behind these microbial metabolite effects, which could be modified by diet to achieve the objective of preventing colorectal cancer in Western societies.
Collapse
|
61
|
Abstract
Colorectal cancer (CRC) is one of the leading causes of cancer-related morbidity and mortality worldwide. There are well-established screening protocols involving fecal testing, radiographic, and endoscopic evaluations that have led to decreased incidence and mortality of CRC in the United States. In addition to screening for CRC, there is interest in preventing colorectal neoplasia by targeting the signaling pathways that have been identified in the pathway of dysplasia progressing to carcinoma. This review will detail the efficacy of multiple potential preventative strategies including lifestyle changes (physical activity, alcohol use, smoking cessation, and obesity); dietary factors (dietary patterns, calcium, vitamin D, fiber, folate, and antioxidants and micronutrients); and chemopreventive agents (nonsteroidal anti-inflammatory drugs, statins, metformin, bisphosphonates, and postmenopausal hormonal therapy).
Collapse
Affiliation(s)
- Scott C Dolejs
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Benjamin Gayed
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Alyssa Fajardo
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
62
|
Lee DH, Keum N, Giovannucci EL. Colorectal Cancer Epidemiology in the Nurses' Health Study. Am J Public Health 2016; 106:1599-607. [PMID: 27459444 DOI: 10.2105/ajph.2016.303320] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES To review the contribution of the Nurses' Health Study (NHS) to identifying risk and protective factors for colorectal adenomas and colorectal cancer (CRC). METHODS We performed a narrative review of the publications using the NHS between 1976 and 2016. RESULTS Existing epidemiological studies using the NHS have reported that red and processed meat, alcohol, smoking, and obesity were associated with an increased risk of CRC, whereas folate, calcium, vitamin D, aspirin, and physical activity were associated with decreased risk of CRC. Moreover, modifiable factors, such as physical activity, vitamin D, folate, insulin and insulin-like growth factor binding protein-1, and diet quality, were identified to be associated with survival among CRC patients. In recent years, molecular pathological epidemiological studies have been actively conducted and have shown refined results by molecular subtypes of CRC. CONCLUSIONS The NHS has provided new insights into colorectal adenomas, CRC etiology, and pathogenic mechanisms. With its unique strengths, the NHS should continue to contribute to the field of CRC epidemiology and play a major role in public health.
Collapse
Affiliation(s)
- Dong Hoon Lee
- All authors are with the Departments of Nutrition and Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA. Edward L. Giovannucci is also with the Department of Medicine, Harvard Medical School, Boston
| | - NaNa Keum
- All authors are with the Departments of Nutrition and Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA. Edward L. Giovannucci is also with the Department of Medicine, Harvard Medical School, Boston
| | - Edward L Giovannucci
- All authors are with the Departments of Nutrition and Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA. Edward L. Giovannucci is also with the Department of Medicine, Harvard Medical School, Boston
| |
Collapse
|
63
|
Rao TJ, Province MA. A Framework for Interpreting Type I Error Rates from a Product-Term Model of Interaction Applied to Quantitative Traits. Genet Epidemiol 2015; 40:144-53. [PMID: 26659945 PMCID: PMC4738444 DOI: 10.1002/gepi.21944] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 10/05/2015] [Accepted: 10/26/2015] [Indexed: 11/11/2022]
Abstract
Adequate control of type I error rates will be necessary in the increasing genome-wide search for interactive effects on complex traits. After observing unexpected variability in type I error rates from SNP-by-genome interaction scans, we sought to characterize this variability and test the ability of heteroskedasticity-consistent standard errors to correct it. We performed 81 SNP-by-genome interaction scans using a product-term model on quantitative traits in a sample of 1,053 unrelated European Americans from the NHLBI Family Heart Study, and additional scans on five simulated datasets. We found that the interaction-term genomic inflation factor (lambda) showed inflation and deflation that varied with sample size and allele frequency; that similar lambda variation occurred in the absence of population substructure; and that lambda was strongly related to heteroskedasticity but not to minor non-normality of phenotypes. Heteroskedasticity-consistent standard errors narrowed the range of lambda, with HC3 outperforming HC0, but in individual scans tended to create new P-value outliers related to sparse two-locus genotype classes. We explain the lambda variation as a result of non-independence of test statistics coupled with stochastic biases in test statistics due to a failure of the test to reach asymptotic properties. We propose that one way to interpret lambda is by comparison to an empirical distribution generated from data simulated under the null hypothesis and without population substructure. We further conclude that the interaction-term lambda should not be used to adjust test statistics and that heteroskedasticity-consistent standard errors come with limitations that may outweigh their benefits in this setting.
Collapse
Affiliation(s)
- Tara J Rao
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Michael A Province
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| |
Collapse
|
64
|
Fagny M, Patin E, MacIsaac JL, Rotival M, Flutre T, Jones MJ, Siddle KJ, Quach H, Harmant C, McEwen LM, Froment A, Heyer E, Gessain A, Betsem E, Mouguiama-Daouda P, Hombert JM, Perry GH, Barreiro LB, Kobor MS, Quintana-Murci L. The epigenomic landscape of African rainforest hunter-gatherers and farmers. Nat Commun 2015; 6:10047. [PMID: 26616214 PMCID: PMC4674682 DOI: 10.1038/ncomms10047] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 10/28/2015] [Indexed: 12/23/2022] Open
Abstract
The genetic history of African populations is increasingly well documented, yet their patterns of epigenomic variation remain uncharacterized. Moreover, the relative impacts of DNA sequence variation and temporal changes in lifestyle and habitat on the human epigenome remain unknown. Here we generate genome-wide genotype and DNA methylation profiles for 362 rainforest hunter-gatherers and sedentary farmers. We find that the current habitat and historical lifestyle of a population have similarly critical impacts on the methylome, but the biological functions affected strongly differ. Specifically, methylation variation associated with recent changes in habitat mostly concerns immune and cellular functions, whereas that associated with historical lifestyle affects developmental processes. Furthermore, methylation variation—particularly that correlated with historical lifestyle—shows strong associations with nearby genetic variants that, moreover, are enriched in signals of natural selection. Our work provides new insight into the genetic and environmental factors affecting the epigenomic landscape of human populations over time. Genetic and environmental factors affect genome-wide patterns of epigenetic variation. Here, the authors show that while current habitat and historical lifestyle impact the methylome of rainforest hunter-gatherers and sedentary farmers, the biological functions affected and the degree of genetic control differ.
Collapse
Affiliation(s)
- Maud Fagny
- Institut Pasteur, Unit of Human Evolutionary Genetics, Paris 75015, France.,Centre National de la Recherche Scientifique, URA3012, Paris 75015, France.,Université Pierre et Marie Curie, Cellule Pasteur UPMC, Paris 75015, France
| | - Etienne Patin
- Institut Pasteur, Unit of Human Evolutionary Genetics, Paris 75015, France.,Centre National de la Recherche Scientifique, URA3012, Paris 75015, France
| | - Julia L MacIsaac
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute and Department of Medical Genetics, University of British Columbia, Vancouver, Canada BC V5Z 4H4
| | - Maxime Rotival
- Institut Pasteur, Unit of Human Evolutionary Genetics, Paris 75015, France.,Centre National de la Recherche Scientifique, URA3012, Paris 75015, France
| | | | - Meaghan J Jones
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute and Department of Medical Genetics, University of British Columbia, Vancouver, Canada BC V5Z 4H4
| | - Katherine J Siddle
- Institut Pasteur, Unit of Human Evolutionary Genetics, Paris 75015, France.,Centre National de la Recherche Scientifique, URA3012, Paris 75015, France
| | - Hélène Quach
- Institut Pasteur, Unit of Human Evolutionary Genetics, Paris 75015, France.,Centre National de la Recherche Scientifique, URA3012, Paris 75015, France
| | - Christine Harmant
- Institut Pasteur, Unit of Human Evolutionary Genetics, Paris 75015, France.,Centre National de la Recherche Scientifique, URA3012, Paris 75015, France
| | - Lisa M McEwen
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute and Department of Medical Genetics, University of British Columbia, Vancouver, Canada BC V5Z 4H4
| | - Alain Froment
- IRD-MNHN, Sorbonne Universités, UMR208, Paris 75005, France
| | - Evelyne Heyer
- CNRS, MNHN, Université Paris Diderot, Sorbonne Paris Cité, Sorbonne Université, UMR7206, Paris 75005, France
| | - Antoine Gessain
- Institut Pasteur, Unité d'Epidémiologie et Physiopathologie des Virus Oncogènes, Paris 75015, France
| | - Edouard Betsem
- Institut Pasteur, Unité d'Epidémiologie et Physiopathologie des Virus Oncogènes, Paris 75015, France.,Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, BP1364 Yaoundé, Cameroon
| | - Patrick Mouguiama-Daouda
- Laboratoire Langue, Culture et Cognition (LCC), Université Omar Bongo, BP 13131 Libreville, Gabon
| | | | - George H Perry
- Departments of Anthropology and Biology, Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - Luis B Barreiro
- Université de Montréal, Centre de Recherche CHU Sainte-Justine, Montréal, Canada H3T 1C5
| | - Michael S Kobor
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute and Department of Medical Genetics, University of British Columbia, Vancouver, Canada BC V5Z 4H4
| | - Lluis Quintana-Murci
- Institut Pasteur, Unit of Human Evolutionary Genetics, Paris 75015, France.,Centre National de la Recherche Scientifique, URA3012, Paris 75015, France
| |
Collapse
|
65
|
Yoshida N, Kinugasa T, Miyoshi H, Sato K, Yuge K, Ohchi T, Fujino S, Shiraiwa S, Katagiri M, Akagi Y, Ohshima K. A High RORγT/CD3 Ratio is a Strong Prognostic Factor for Postoperative Survival in Advanced Colorectal Cancer: Analysis of Helper T Cell Lymphocytes (Th1, Th2, Th17 and Regulatory T Cells). Ann Surg Oncol 2015; 23:919-27. [PMID: 26564244 DOI: 10.1245/s10434-015-4923-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Indexed: 12/19/2022]
Abstract
BACKGROUND Tumor-infiltrating lymphocytes (TILs), part of the host immune response, have been widely reported as influential factors in the tumor microenvironment for the clinical outcome of colorectal cancer (CRC). However, the network of helper T cells is very complex, and which T-cell subtypes affect the progression of CRC and postoperative prognosis remains unclear. This study investigated the expression of several subtypes of TILs including T helper type 1 (Th1), Th2, Th17, and regulatory T (Treg) cells to determine their correlation with clinicopathologic features and postoperative prognosis. METHODS The study investigated the expression of TILs using immunohistochemistry of tissue microarray samples for 199 CRC patients. The number of each T-cell subtype infiltrating tumors was counted using ImageJ software. The relationship between TIL marker expression, clinicopathologic features, and prognosis was analyzed. RESULTS A high RORγT/CD3 ratio (Th17 ratio) was significantly correlated with lymph node metastasis (p = 0.002), and a high of Foxp3/CD3 ratio (Treg ratio) was correlated with tumor location in the colon (p = 0.04), as shown by the Chi square test. In multivariate analysis, a high RORγT/CD3 ratio was the only independent prognostic factor for overall survival (p = 0.04; hazard ratio [HR], 1.84; 95% confidence interval [CI] 1.02-3.45). CONCLUSIONS This study confirmed a high RORγT/CD3 ratio as a strong prognostic marker for postoperative survival. The immunohistochemistry results suggest that Th17 may affect lymph node metastasis in CRC. If new immunotherapies reducing Th17 expression are established, they may improve the efficiency of cancer treatment and prolong the survival of patients with CRC.
Collapse
Affiliation(s)
- Naohiro Yoshida
- Department of Surgery, Kurume University School of Medicine, Kurume-shi, Fukuoka, Japan
| | - Tetsushi Kinugasa
- Department of Surgery, Kurume University School of Medicine, Kurume-shi, Fukuoka, Japan.
| | - Hiroaki Miyoshi
- Department of Pathology, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Kensaku Sato
- Department of Pathology, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Kotaro Yuge
- Department of Surgery, Kurume University School of Medicine, Kurume-shi, Fukuoka, Japan
| | - Takafumi Ohchi
- Department of Surgery, Kurume University School of Medicine, Kurume-shi, Fukuoka, Japan
| | - Shinya Fujino
- Department of Surgery, Kurume University School of Medicine, Kurume-shi, Fukuoka, Japan
| | - Sachiko Shiraiwa
- Department of Surgery, Kurume University School of Medicine, Kurume-shi, Fukuoka, Japan
| | - Mitsuhiro Katagiri
- Department of Surgery, Kurume University School of Medicine, Kurume-shi, Fukuoka, Japan
| | - Yoshito Akagi
- Department of Surgery, Kurume University School of Medicine, Kurume-shi, Fukuoka, Japan
| | - Koichi Ohshima
- Department of Pathology, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| |
Collapse
|
66
|
Abstract
Much of cancer genetics research has focused on the identification of the most-important somatic mutations ('major drivers') that cause tumour growth. However, many mutations found in cancer might not be major drivers or 'passenger' mutations, but instead might have relatively weak tumour-promoting effects. Our aim is to highlight the existence of these mutations (termed 'mini drivers' herein), as multiple mini-driver mutations might substitute for a major-driver change, especially in the presence of genomic instability or high mutagen exposure. The mini-driver model has clinical implications: for example, the effects of therapeutically targeting such genes may be limited. However, the main importance of the model lies in helping to provide a complete understanding of tumorigenesis, especially as we anticipate that an increasing number of mini-driver mutations will be found by cancer genome sequencing.
Collapse
Affiliation(s)
- Francesc Castro-Giner
- Molecular and Population Genetics Laboratory, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Peter Ratcliffe
- Henry Wellcome Building for Molecular Physiology, Nuffield Department of Clinical Medicine, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Ian Tomlinson
- Molecular and Population Genetics Laboratory, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| |
Collapse
|
67
|
Abstract
Colorectal cancer (CRC) is a complex disease that develops as a consequence of both genetic and environmental risk factors. A small proportion (3-5%) of cases arise from hereditary syndromes predisposing to early onset CRC as a result of mutations in over a dozen well defined genes. In contrast, CRC is predominantly a late onset 'sporadic' disease, developing in individuals with no obvious hereditary syndrome. In recent years, genome wide association studies have discovered that over 40 genetic regions are associated with weak effects on sporadic CRC, and it has been estimated that increasingly large genome wide scans will identify many additional novel genetic regions. Subsequent experimental validations have identified the causally related variant(s) in a limited number of these genetic regions. Further biological insight could be obtained through ethnically diverse study populations, larger genetic sequencing studies and development of higher throughput functional experiments. Along with inherited variation, integration of the tumour genome may shed light on the carcinogenic processes in CRC. In addition to summarising the genetic architecture of CRC, this review discusses genetic factors that modify environmental predictors of CRC, as well as examples of how genetic insight has improved clinical surveillance, prevention and treatment strategies. In summary, substantial progress has been made in uncovering the genetic architecture of CRC, and continued research efforts are expected to identify additional genetic risk factors that further our biological understanding of this disease. Subsequently these new insights will lead to improved treatment and prevention of colorectal cancer.
Collapse
Affiliation(s)
- Ulrike Peters
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Epidemiology, University of Washington School of Public Health, Seattle, WA, USA
| | - Stephanie Bien
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Niha Zubair
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
68
|
Li W, Espinal-Enríquez J, Simpfendorfer KR, Hernández-Lemus E. A survey of disease connections for CD4+ T cell master genes and their directly linked genes. Comput Biol Chem 2015; 59 Pt B:78-90. [PMID: 26411796 DOI: 10.1016/j.compbiolchem.2015.08.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 08/18/2015] [Accepted: 08/21/2015] [Indexed: 02/07/2023]
Abstract
Genome-wide association studies and other genetic analyses have identified a large number of genes and variants implicating a variety of disease etiological mechanisms. It is imperative for the study of human diseases to put these genetic findings into a coherent functional context. Here we use system biology tools to examine disease connections of five master genes for CD4+ T cell subtypes (TBX21, GATA3, RORC, BCL6, and FOXP3). We compiled a list of genes functionally interacting (protein-protein interaction, or by acting in the same pathway) with the master genes, then we surveyed the disease connections, either by experimental evidence or by genetic association. Embryonic lethal genes (also known as essential genes) are over-represented in master genes and their interacting genes (55% versus 40% in other genes). Transcription factors are significantly enriched among genes interacting with the master genes (63% versus 10% in other genes). Predicted haploinsufficiency is a feature of most these genes. Disease-connected genes are enriched in this list of genes: 42% of these genes have a disease connection according to Online Mendelian Inheritance in Man (OMIM) (versus 23% in other genes), and 74% are associated with some diseases or phenotype in a Genome Wide Association Study (GWAS) (versus 43% in other genes). Seemingly, not all of the diseases connected to genes surveyed were immune related, which may indicate pleiotropic functions of the master regulator genes and associated genes.
Collapse
Affiliation(s)
- Wentian Li
- The Robert S. Boas Center for Genomics and Human Genetics, The Feinstein Institute for Medical Research, North Shore LIJ Health System, Manhasset, NY, USA.
| | - Jesús Espinal-Enríquez
- Computational Genomics Department, National Institute of Genomic Medicine, México, D.F., Mexico; Complexity in Systems Biology, Center for Complexity Sciences, Universidad Nacional Autónoma de México, México, D.F., Mexico
| | - Kim R Simpfendorfer
- The Robert S. Boas Center for Genomics and Human Genetics, The Feinstein Institute for Medical Research, North Shore LIJ Health System, Manhasset, NY, USA
| | - Enrique Hernández-Lemus
- Computational Genomics Department, National Institute of Genomic Medicine, México, D.F., Mexico; Complexity in Systems Biology, Center for Complexity Sciences, Universidad Nacional Autónoma de México, México, D.F., Mexico
| |
Collapse
|
69
|
Li K, Huang T, Li D. Gene–Diet Interaction on Body Weight Maintenance. Curr Nutr Rep 2015. [DOI: 10.1007/s13668-015-0133-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
70
|
Jiao S, Peters U, Berndt S, Bézieau S, Brenner H, Campbell PT, Chan AT, Chang-Claude J, Lemire M, Newcomb PA, Potter JD, Slattery ML, Woods MO, Hsu L. Powerful Set-Based Gene-Environment Interaction Testing Framework for Complex Diseases. Genet Epidemiol 2015; 39:609-18. [PMID: 26095235 DOI: 10.1002/gepi.21908] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 04/20/2015] [Accepted: 05/06/2015] [Indexed: 01/15/2023]
Abstract
Identification of gene-environment interaction (G × E) is important in understanding the etiology of complex diseases. Based on our previously developed Set Based gene EnviRonment InterAction test (SBERIA), in this paper we propose a powerful framework for enhanced set-based G × E testing (eSBERIA). The major challenge of signal aggregation within a set is how to tell signals from noise. eSBERIA tackles this challenge by adaptively aggregating the interaction signals within a set weighted by the strength of the marginal and correlation screening signals. eSBERIA then combines the screening-informed aggregate test with a variance component test to account for the residual signals. Additionally, we develop a case-only extension for eSBERIA (coSBERIA) and an existing set-based method, which boosts the power not only by exploiting the G-E independence assumption but also by avoiding the need to specify main effects for a large number of variants in the set. Through extensive simulation, we show that coSBERIA and eSBERIA are considerably more powerful than existing methods within the case-only and the case-control method categories across a wide range of scenarios. We conduct a genome-wide G × E search by applying our methods to Illumina HumanExome Beadchip data of 10,446 colorectal cancer cases and 10,191 controls and identify two novel interactions between nonsteroidal anti-inflammatory drugs (NSAIDs) and MINK1 and PTCHD3.
Collapse
Affiliation(s)
- Shuo Jiao
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Ulrike Peters
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Sonja Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, United States of America
| | | | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Peter T Campbell
- Epidemiology Research Program, American Cancer Society, Atlanta, Georgia, United States of America
| | - Andrew T Chan
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | | | - Polly A Newcomb
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America.,School of Public Health, University of Washington, Seattle, Washington, United States of America
| | - John D Potter
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America.,Ontario Institute for Cancer Research, Toronto, Canada.,Centre for Public Health Research, Massey University, Wellington, New Zealand
| | - Martha L Slattery
- Department of Internal Medicine, University of Utah Health Sciences Center, Salt Lake City, Utah, United States of America
| | - Michael O Woods
- Discipline of Genetics, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Li Hsu
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| |
Collapse
|
71
|
Ben-Zvi D, Melton DA. Modeling human nutrition using human embryonic stem cells. Cell 2015; 161:12-17. [PMID: 25815980 DOI: 10.1016/j.cell.2015.02.039] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Indexed: 01/07/2023]
Abstract
Nutrition presents unanswered scientific questions of high public health importance. We envision model systems composed of interacting gastrointestinal and metabolic tissues derived from human embryonic stem cells, populated by gut microbiota. The culture will be embedded in 3D scaffolds, creating a controlled experimental system that enables tissue sampling and imaging.
Collapse
Affiliation(s)
- Danny Ben-Zvi
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge MA, USA.
| | - Douglas A Melton
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge MA, USA
| |
Collapse
|
72
|
Abstract
Several dietary approaches have been proposed to prevent the onset of chronic diseases. As yet, no single approach has emerged as having the most consistent health benefits. This arises, in part, due to the fact that diet influences health in the context of individual factors with genetic components. Therefore, the effects of diet on health may be dependent on an individual's genetic background. At this time we lack robust evidence for the effects of interactions between genes and dietary patterns on health. To understand why, I will briefly review the most methodologically strong attempts to identify gene-diet interactions, which will illuminate how the challenges facing all of genetic research apply to the search for gene-diet interactions. Then I will discuss some ways in which these challenges are being addressed that offer hope for the future in which the best diet for an individual is identified based on their genetic variation.
Collapse
Affiliation(s)
- Alexis C. Frazier-Wood
- USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, 1100 Bates Street, Houston, TX 77030 USA
| |
Collapse
|
73
|
Potter JD. Nutritional epidemiology--there's life in the old dog yet! Cancer Epidemiol Biomarkers Prev 2014; 24:323-30. [PMID: 25515549 DOI: 10.1158/1055-9965.epi-14-1327] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Consideration is given to the idea that the nutritional epidemiology of cancer is dead, as some in the media have claimed. The basis for the claim does not lie in science nor has anyone with relevant knowledge made such a statement-although that, too, has been claimed. Evidence is adduced for the importance of past achievements of nutritional epidemiology. Attention is similarly drawn to recent contributions. In particular, I note the state of play of cancer and plant foods, fat and breast cancer, meat and cancer, vegetarians, intervention studies, migrant studies, and westernization of diet and lifestyle. Some next steps and some currently important questions are outlined.
Collapse
Affiliation(s)
- John D Potter
- Centre for Public Health Research, Massey University, Wellington, New Zealand. Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington.
| |
Collapse
|
74
|
Abstract
A number of studies have evaluated the role of gene-diet interaction in the etiology of colorectal cancer (CRC). Historically, these studies focused on established dietary risk factors and genes involved in their metabolism. However, results from these candidate gene studies were inconsistent, possibly due to multiple testing and publication bias. In recent years, genome-wide association studies have identified a number of CRC susceptibility loci, and subsequent meta-analyses have observed limited evidence that diet may modify the risk associated with these susceptibility loci. Statistical techniques have been recently developed to evaluate the presence of interaction across the entire genome; results from these genome-wide studies have demonstrated limited evidence of interaction and have failed to replicate results from candidate gene studies and those using established susceptibility loci. However, larger sample sizes are likely needed to elucidate modest or weak interaction in genome-wide studies of gene-diet interaction.
Collapse
|
75
|
Du M, Zhang X, Hoffmeister M, Schoen RE, Baron JA, Berndt SI, Brenner H, Carlson CS, Casey G, Chan AT, Curtis KR, Duggan D, Gauderman WJ, Giovannucci EL, Gong J, Harrison TA, Hayes RB, Henderson BE, Hopper JL, Hsu L, Hudson TJ, Hutter CM, Jenkins MA, Jiao S, Kocarnik JM, Kolonel LN, Le Marchand L, Lin Y, Newcomb PA, Rudolph A, Seminara D, Thornquist MD, Ulrich CM, White E, Wu K, Zanke BW, Campbell PT, Slattery ML, Peters U, Chang-Claude J, Potter JD. No evidence of gene-calcium interactions from genome-wide analysis of colorectal cancer risk. Cancer Epidemiol Biomarkers Prev 2014; 23:2971-6. [PMID: 25192705 PMCID: PMC4257872 DOI: 10.1158/1055-9965.epi-14-0893] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Calcium intake may reduce risk of colorectal cancer, but the mechanisms remain unclear. Studies of interaction between calcium intake and SNPs in calcium-related pathways have yielded inconsistent results. METHODS To identify gene-calcium interactions, we tested interactions between approximately 2.7 million SNPs across the genome with self-reported calcium intake (from dietary or supplemental sources) in 9,006 colorectal cancer cases and 9,503 controls of European ancestry. To test for multiplicative interactions, we used multivariable logistic regression and defined statistical significance using the conventional genome-wide α = 5E-08. RESULTS After accounting for multiple comparisons, there were no statistically significant SNP interactions with total, dietary, or supplemental calcium intake. CONCLUSIONS We found no evidence of SNP interactions with calcium intake for colorectal cancer risk in a large population of 18,509 individuals. IMPACT These results suggest that in genome-wide analysis common genetic variants do not strongly modify the association between calcium intake and colorectal cancer in European populations.
Collapse
Affiliation(s)
- Mengmeng Du
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington. School of Public Health, University of Washington, Seattle, Washington. Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Xuehong Zhang
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Robert E Schoen
- Department of Medicine and Epidemiology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - John A Baron
- Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Sonja I Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany. German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Christopher S Carlson
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Graham Casey
- Department of Preventive Medicine, University of Southern California, Los Angeles, California
| | - Andrew T Chan
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts. Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Keith R Curtis
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - David Duggan
- Genetic Basis of Human Disease, Translational Genomics Research Institute (TGen), Phoenix, Arizona
| | - W James Gauderman
- Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Edward L Giovannucci
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts. Departments of Epidemiology and Nutrition, Harvard School of Public Health, Boston, Massachusetts
| | - Jian Gong
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Tabitha A Harrison
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Richard B Hayes
- Division of Epidemiology, Department of Population Health, New York University School of Medicine, New York, New York
| | - Brian E Henderson
- Keck School of Medicine, University of Southern California, Los Angeles, California
| | - John L Hopper
- Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Li Hsu
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Thomas J Hudson
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada. Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada. Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Carolyn M Hutter
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington. Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, Maryland
| | - Mark A Jenkins
- Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Shuo Jiao
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Jonathan M Kocarnik
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington. School of Public Health, University of Washington, Seattle, Washington
| | - Laurence N Kolonel
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Loic Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Yi Lin
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Polly A Newcomb
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington. School of Public Health, University of Washington, Seattle, Washington
| | - Anja Rudolph
- Division of Cancer Epidemiology, Unit of Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniela Seminara
- Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, Maryland
| | - Mark D Thornquist
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Cornelia M Ulrich
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington. School of Public Health, University of Washington, Seattle, Washington. Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Emily White
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington. School of Public Health, University of Washington, Seattle, Washington
| | - Kana Wu
- Departments of Epidemiology and Nutrition, Harvard School of Public Health, Boston, Massachusetts
| | - Brent W Zanke
- Division of Hematology, Faculty of Medicine, The University of Ottawa, Ottawa, Ontario, Canada
| | - Peter T Campbell
- Epidemiology Research Program, American Cancer Society, Atlanta, Georgia
| | - Martha L Slattery
- Department of Internal Medicine, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Ulrike Peters
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington. School of Public Health, University of Washington, Seattle, Washington
| | - Jenny Chang-Claude
- Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, Maryland
| | - John D Potter
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington. School of Public Health, University of Washington, Seattle, Washington. Centre for Public Health Research, Massey University, Wellington, New Zealand
| |
Collapse
|