1
|
Hamilton KR, McGill LS, Campbell CM, Lanzkron SM, Carroll CP, Latremoliere A, Haythornthwaite JA, Korczeniewska OA. Genetic contributions to pain modulation in sickle cell: A focus on single nucleotide polymorphisms. GENE REPORTS 2024; 36:101983. [PMID: 39219841 PMCID: PMC11361162 DOI: 10.1016/j.genrep.2024.101983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Background Despite recent advances in our knowledge of genetic contributions to the highly variable sickle cell disease (SCD) phenotype, our understanding of genetic factors associated with pain sensitivity in SCD remains limited. Previous studies investigated specific variants in single candidate genes and their association with SCD pain variability. The primary aim of the current study was to expand the genes and polymorphisms tested to discover new risk genes (polymorphisms) associated with central sensitization for individuals with SCD. Methods Adults with sickle cell disease (n = 59, Mage = 36.8 ± 11.5, 65.8 % female) underwent quantitative sensory testing to examine central sensitization and general pain sensitivity. Participants reported average crisis and non-crisis pain intensities weekly using a 0-100 scale, and provided salivary samples for genotyping. The Hardy-Weinberg equilibrium was verified for controls, and allele distributions were tested with chi-square and odds ratio tests. The Benjamini-Hochberg procedure was used to control for false discovery rate. Regression analyses and Wilcoxon tests were used to test associations for normally distributed and skewed data, respectively. Results Central sensitization and general pain sensitivity were not associated with hemoglobin genotype (Ps > 0.05). Of 4145 SNPs tested, following false discovery rate adjustments, 11 SNPs (rs11575839, rs12185625, rs12289836, rs1493383, rs2233976, rs3131787, rs3739693, rs4292454, rs4364, rs4678, rs6773307) were significantly associated with central sensitization, and one SNP (rs7778077) was significantly associated with average weekly non-crisis pain. No SNPs were associated with general pain sensitivity. Conclusions These findings provide insights into genetic variants association with average non-crisis pain and central sensitization for individuals with SCD, and may provide support for genetic predictors of heightened pain experience within SCD.
Collapse
Affiliation(s)
- Katrina R. Hamilton
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Lakeya S. McGill
- Department of Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Claudia M. Campbell
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Sophie M. Lanzkron
- Department of Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - C. Patrick Carroll
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Alban Latremoliere
- Department of Neurosurgery, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Jennifer A. Haythornthwaite
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Olga A. Korczeniewska
- Center for Orofacial Pain and Temporomandibular Disorders, Department of Diagnostic Sciences, Rutgers School of Dental Medicine, Newark, NJ, USA
| |
Collapse
|
2
|
Loomis D, Dzhambov AM, Momen NC, Chartres N, Descatha A, Guha N, Kang SK, Modenese A, Morgan RL, Ahn S, Martínez-Silveira MS, Zhang S, Pega F. The effect of occupational exposure to welding fumes on trachea, bronchus and lung cancer: A systematic review and meta-analysis from the WHO/ILO Joint Estimates of the Work-related Burden of Disease and Injury. ENVIRONMENT INTERNATIONAL 2022; 170:107565. [PMID: 36402034 DOI: 10.1016/j.envint.2022.107565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/27/2022] [Accepted: 10/03/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND The World Health Organization (WHO) and the International Labour Organization (ILO) are the producers of the WHO/ILO Joint Estimates of the Work-related Burden of Disease and Injury (WHO/ILO Joint Estimates). Welding fumes have been classified as carcinogenic to humans (Group 1) by the WHO International Agency for Research on Cancer (IARC) in IARC Monograph 118; this assessment found sufficient evidence from studies in humans that welding fumes are a cause of lung cancer. In this article, we present a systematic review and meta-analysis of parameters for estimating the number of deaths and disability-adjusted life years from trachea, bronchus, and lung cancer attributable to occupational exposure to welding fumes, to inform the development of WHO/ILO Joint Estimates on this burden of disease (if considered feasible). OBJECTIVES We aimed to systematically review and meta-analyse estimates of the effect of any (or high) occupational exposure to welding fumes, compared with no (or low) occupational exposure to welding fumes, on trachea, bronchus, and lung cancer (three outcomes: prevalence, incidence, and mortality). DATA SOURCES We developed and published a protocol, applying the Navigation Guide as an organizing systematic review framework where feasible. We searched electronic databases for potentially relevant records from published and unpublished studies, including Medline, EMBASE, Web of Science, CENTRAL and CISDOC. We also searched grey literature databases, Internet search engines, and organizational websites; hand-searched reference lists of previous systematic reviews; and consulted additional experts. STUDY ELIGIBILITY AND CRITERIA We included working-age (≥15 years) workers in the formal and informal economy in any Member State of WHO and/or ILO but excluded children (<15 years) and unpaid domestic workers. We included randomized controlled trials, cohort studies, case-control studies, and other non-randomized intervention studies with an estimate of the effect of any (or high) occupational exposure to welding fumes, compared with occupational exposure to no (or low) welding fumes, on trachea, bronchus, and lung cancer (prevalence, incidence, and mortality). STUDY APPRAISAL AND SYNTHESIS METHODS At least two review authors independently screened titles and abstracts against the eligibility criteria at a first review stage and full texts of potentially eligible records at a second stage, followed by extraction of data from qualifying studies. If studies reported odds ratios, these were converted to risk ratios (RRs). We combined all RRs using random-effects meta-analysis. Two or more review authors assessed the risk of bias, quality of evidence, and strength of evidence, using the Navigation Guide tools and approaches adapted to this project. Subgroup (e.g., by WHO region and sex) and sensitivity analyses (e.g., studies judged to be of "high"/"probably high" risk of bias compared with "low"/"probably low" risk of bias) were conducted. RESULTS Forty-one records from 40 studies (29 case control studies and 11 cohort studies) met the inclusion criteria, comprising over 1,265,512 participants (≥22,761 females) in 21 countries in three WHO regions (Region of the Americas, European Region, and Western Pacific Region). The exposure and outcome were generally assessed by job title or self-report, and medical or administrative records, respectively. Across included studies, risk of bias was overall generally probably low/low, with risk judged high or probably high for several studies in the domains for misclassification bias and confounding. Our search identified no evidence on the outcome of having trachea, bronchus, and lung cancer (prevalence). Compared with no (or low) occupational exposure to welding fumes, any (or high) occupational exposure to welding fumes increased the risk of acquiring trachea, bronchus, and lung cancer (incidence) by an estimated 48 % (RR 1.48, 95 % confidence interval [CI] 1.29-1.70, 23 studies, 57,931 participants, I2 24 %; moderate quality of evidence). Compared with no (or low) occupational exposure to welding fumes, any (or high) occupational exposure to welding fumes increased the risk dying from trachea, bronchus, and lung cancer (mortality) by an estimated 27 % (RR 1.27, 95 % CI 1.04-1.56, 3 studies, 8,686 participants, I2 0 %; low quality of evidence). Our subgroup analyses found no evidence for difference by WHO region and sex. Sensitivity analyses supported the main analyses. CONCLUSIONS Overall, for incidence and mortality of trachea, bronchus, and lung cancer, we judged the existing body of evidence for human data as "sufficient evidence of harmfulness" and "limited evidence of harmfulness", respectively. Occupational exposure to welding fumes increased the risk of acquiring and dying from trachea, bronchus, and lung cancer. Producing estimates for the burden of trachea, bronchus, and lung cancer attributable to any (or high) occupational exposure to welding fumes appears evidence-based, and the pooled effect estimates presented in this systematic review could be used as input data for the WHO/ILO Joint Estimates. PROTOCOL IDENTIFIER: https://doi.org/10.1016/j.envint.2020.106089.
Collapse
Affiliation(s)
- Dana Loomis
- School of Community Health Sciences, University of Nevada, Reno, Reno, NV, the United States of America; Plumas County Public Health Agency, Plumas County, CA, the United States of America.
| | - Angel M Dzhambov
- Department of Hygiene, Faculty of Public Health, Medical University of Plovdiv, Plovdiv, Bulgaria; Institute for Highway Engineering and Transport Planning, Graz University of Technology, Graz, Austria.
| | - Natalie C Momen
- Department of Environment, Climate Change and Health, World Health Organization, Geneva, Switzerland.
| | - Nicholas Chartres
- Program on Reproductive Health and the Environment, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Francisco, San Francisco, the United States of America.
| | - Alexis Descatha
- AP-HP (Paris Hospital "Assistance Publique Hôpitaux de Paris"), Occupational Health Unit, University Hospital of West Suburb of Paris, Poincaré Site, Garches, France /Versailles St-Quentin Univ - Paris Saclay Univ (UVSQ), UMS 011, UMR-S 1168, France; Univ Angers, CHU Angers, Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S1085, SFR ICAT, CAPTV CDC, Angers, France.
| | - Neela Guha
- Office of Environmental Health Hazard Assessment, California Environmental Protection Agency, Oakland, CA, the United States of America.
| | - Seong-Kyu Kang
- Department of Occupational and Environmental Medicine, Gil Medical Center, Gachon University College of Medicine, Incheon, Republic of Korea.
| | - Alberto Modenese
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena & Reggio Emilia, Modena, Italy.
| | - Rebecca L Morgan
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada.
| | - Seoyeon Ahn
- National Pension Research Institute, Jeonju-si, Republic of Korea.
| | | | - Siyu Zhang
- National Institute for Occupational Health and Poison Control, Center for Disease Control and Prevention, Beijing, People's Republic of China.
| | - Frank Pega
- Department of Environment, Climate Change and Health, World Health Organization, Geneva, Switzerland.
| |
Collapse
|
3
|
Rare POLN mutations confer risk for familial nasopharyngeal carcinoma through weakened Epstein-Barr virus lytic replication. EBioMedicine 2022; 84:104267. [PMID: 36116213 PMCID: PMC9486052 DOI: 10.1016/j.ebiom.2022.104267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 08/29/2022] [Accepted: 08/29/2022] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Nasopharyngeal carcinoma (NPC) exhibits significant familial aggregation; however, its susceptibility genes are largely unknown. Thus, this study aimed to identify germline mutations that might contribute to the risk of familial NPC, and explore their biological functions. METHODS Whole-exome sequencing was performed in 13 NPC pedigrees with multiple cases. Mutations co-segregated with disease status were further validated in a cohort composed of 563 probands from independent families, 2,953 sporadic cases, and 3,175 healthy controls. Experimental studies were used to explore the functions of susceptibility genes and their disease-related mutations. FINDINGS The three rare missense mutations in POLN (DNA polymerase nu) gene, P577L, R303Q, and F545C, were associated with familial NPC risk (5/576 [0·87%] in cases vs. 2/3374 [0·059%] in healthy controls with an adjusted OR of 44·84 [95% CI:3·91-514·34, p = 2·25 × 10-3]). POLN was involved in Epstein-Barr virus (EBV) lytic replication in NPC cells in vitro. POLN promoted viral DNA replication, immediate-early and late lytic gene expression, and progeny viral particle production, ultimately affecting the proliferation of host cells. The three mutations were located in two pivotal functional domains and were predicted to alter the protein stability of POLN in silico. Further assays demonstrated that POLN carrying any of the three mutations displayed reduced protein stability and decreased expression levels, thereby impairing its ability to promote complete EBV lytic replication and facilitate cell survival. INTERPRETATION We identified a susceptibility gene POLN for familial NPC and elucidated its function. FUNDING This study was funded by the National Key Research and Development Program of China (2021YFC2500400); the National Key Research and Development Program of China (2020YFC1316902); the Basic and Applied Basic Research Foundation of Guangdong Province, China (2021B1515420007); the National Natural Science Foundation of China (81973131); the National Natural Science Foundation of China (82003520); the National Natural Science Foundation of China (81903395).
Collapse
|
4
|
Olsson A, Guha N, Bouaoun L, Kromhout H, Peters S, Siemiatycki J, Ho V, Gustavsson P, Boffetta P, Vermeulen R, Behrens T, Brüning T, Kendzia B, Guénel P, Luce D, Karrasch S, Wichmann HE, Consonni D, Landi MT, Caporaso NE, Merletti F, Mirabelli D, Richiardi L, Jöckel KH, Ahrens W, Pohlabeln H, Tardón A, Zaridze D, Field JK, Lissowska J, Świątkowska B, McLaughlin JR, Demers PA, Bencko V, Foretova L, Janout V, Pándics T, Fabianova E, Mates D, Forastiere F, Bueno-de-Mesquita B, Schüz J, Straif K. Occupational Exposure to Polycyclic Aromatic Hydrocarbons and Lung Cancer Risk: Results from a Pooled Analysis of Case-Control Studies (SYNERGY). Cancer Epidemiol Biomarkers Prev 2022; 31:1433-1441. [PMID: 35437574 PMCID: PMC9377765 DOI: 10.1158/1055-9965.epi-21-1428] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/22/2022] [Accepted: 04/11/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Exposure to polycyclic aromatic hydrocarbons (PAH) occurs widely in occupational settings. We investigated the association between occupational exposure to PAH and lung cancer risk and joint effects with smoking within the SYNERGY project. METHODS We pooled 14 case-control studies with information on lifetime occupational and smoking histories conducted between 1985 and 2010 in Europe and Canada. Exposure to benzo[a]pyrene (BaP) was used as a proxy of PAH and estimated from a quantitative general population job-exposure matrix. Multivariable unconditional logistic regression models, adjusted for smoking and exposure to other occupational lung carcinogens, estimated ORs, and 95% confidence intervals (CI). RESULTS We included 16,901 lung cancer cases and 20,965 frequency-matched controls. Adjusted OR for PAH exposure (ever) was 1.08 (CI, 1.02-1.15) in men and 1.20 (CI, 1.04-1.38) in women. When stratified by smoking status and histologic subtype, the OR for cumulative exposure ≥0.24 BaP μg/m3-years in men was higher in never smokers overall [1.31 (CI, 0.98-1.75)], for small cell [2.53 (CI, 1.28-4.99)] and squamous cell cancers [1.33 (CI, 0.80-2.21)]. Joint effects between PAH and smoking were observed. Restricting analysis to the most recent studies showed no increased risk. CONCLUSIONS Elevated lung cancer risk associated with PAH exposure was observed in both sexes, particularly for small cell and squamous cell cancers, after accounting for cigarette smoking and exposure to other occupational lung carcinogens. IMPACT The lack of association between PAH and lung cancer in more recent studies merits further research under today's exposure conditions and worker protection measures.
Collapse
Affiliation(s)
- Ann Olsson
- International Agency for Research on Cancer (IARC/WHO), Lyon, France
| | - Neela Guha
- Office of Environmental Health Hazard Assessment, California Environmental Protection Agency, Oakland, California
| | - Liacine Bouaoun
- International Agency for Research on Cancer (IARC/WHO), Lyon, France
| | - Hans Kromhout
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, the Netherlands
| | - Susan Peters
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, the Netherlands
| | - Jack Siemiatycki
- Department of Social and Preventive Medicine, University of Montreal, Montreal, Canada
| | - Vikki Ho
- Department of Social and Preventive Medicine, University of Montreal, Montreal, Canada
| | - Per Gustavsson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Paolo Boffetta
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, New York
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Roel Vermeulen
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, the Netherlands
| | - Thomas Behrens
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr University (IPA), Bochum, Germany
| | - Thomas Brüning
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr University (IPA), Bochum, Germany
| | - Benjamin Kendzia
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr University (IPA), Bochum, Germany
| | - Pascal Guénel
- Center for research in Epidemiology and Population Health (CESP), Exposome and Heredity team, Inserm U1018, University Paris-Saclay, Villejuif, France
| | - Danièle Luce
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Pointe-à-Pitre, France
| | - Stefan Karrasch
- Institute and Clinic for Occupational, Social and Environmental Medicine, University Hospital, LMU Munich, Munich, Germany
- Comprehensive Pneumology Center Munich (CPC-M), Member of the German Center for Lung Research (DZL), Munich, Germany
- Institute of Epidemiology, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
| | - Heinz-Erich Wichmann
- Institute of Epidemiology, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
- Institut für Medizinische Informatik Biometrie Epidemiologie, Ludwig Maximilians University, Munich, Germany
| | - Dario Consonni
- Epidemiology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Maria Teresa Landi
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland
| | - Neil E. Caporaso
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland
| | - Franco Merletti
- Cancer Epidemiology Unit, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Dario Mirabelli
- Cancer Epidemiology Unit, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Lorenzo Richiardi
- Cancer Epidemiology Unit, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Karl-Heinz Jöckel
- Institute for Medical Informatics, Biometry and Epidemiology (IMIBE), University Hospital Essen, Essen, Germany
| | - Wolfgang Ahrens
- Leibniz Institute for Prevention Research and Epidemiology - BIPS, Bremen, Germany
- Faculty of Mathematics and Computer Science, Institute of Statistics, University of Bremen, Bremen, Germany
| | - Hermann Pohlabeln
- Leibniz Institute for Prevention Research and Epidemiology - BIPS, Bremen, Germany
| | - Adonina Tardón
- Department of Public Health, University of Oviedo. ISPA and CIBERESP, Oviedo, Spain
| | - David Zaridze
- Department of cancer epidemiology and Prevention, N.N. Blokhin National Research Centre of oncology, Moscow, Russia
| | - John K. Field
- Roy Castle Lung Cancer Research Programme, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Jolanta Lissowska
- Epidemiology Unit, Department of Cancer Epidemiology and Prevention, M. Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Beata Świątkowska
- Department of Environmental Epidemiology, The Nofer Institute of Occupational Medicine, Lodz, Poland
| | - John R. McLaughlin
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - Paul A. Demers
- Occupational Cancer Research Centre, Ontario Health, Toronto, Canada
| | - Vladimir Bencko
- Institute of Hygiene and Epidemiology, First Faculty of Medicine, Charles University, Prague, Czechia
| | | | - Vladimir Janout
- Faculty of Health Sciences, Palacky University, Olomouc, Czechia
| | | | - Eleonora Fabianova
- Regional Authority of Public Health, Banská Bystrica, Slovakia
- Faculty of Health, Catholic University, Ružomberok, Slovakia
| | - Dana Mates
- National Institute of Public Health, Bucharest, Romania
| | | | - Bas Bueno-de-Mesquita
- Former senior scientist, Centre for Nutrition, Prevention and Health Services, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Joachim Schüz
- International Agency for Research on Cancer (IARC/WHO), Lyon, France
| | - Kurt Straif
- ISGlobal, Barcelona, Spain
- Boston College, Massachusetts
| |
Collapse
|
5
|
Chien HT, Yeh CC, Young CK, Chen TP, Liao CT, Wang HM, Cho KL, Huang SF. Polygenic Panels Predicting the Susceptibility of Multiple Upper Aerodigestive Tract Cancer in Oral Cancer Patients. J Pers Med 2021; 11:jpm11050425. [PMID: 34070222 PMCID: PMC8158753 DOI: 10.3390/jpm11050425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/08/2021] [Accepted: 05/14/2021] [Indexed: 11/16/2022] Open
Abstract
Head and neck cancer was closely related with habitual use of cigarette and alcohol. Those cancer patients are susceptible to develop multiple primary tumors (MPTs). In this study, we utilized the single nucleotide polymorphisms (SNPs) array (Affymetrix Axion Genome-Wide TWB 2.0 Array Plate) to investigate patients' risks of developing multiple primary cancers. We recruited 712 male head and neck cancer patients between Mar 1996 and Feb 2017. Two hundred and eighty-six patients (40.2%) had MPTs and 426 (59.8%) had single cancer. Four hundred and twelve normal controls were also recruited. A list of seventeen factors was extracted and ten factors were demonstrated to increase the risks of multiple primary cancers (alcohol drinking, rs118169127, rs149089400, rs76367287, rs61401220, rs141057871, rs7129229, older age, rs3760265, rs9554264; all were p value < 0.05). Polygenic scoring model was built and the area under curve to predict the risk developing MPTs is 0.906. Alcohol drinking, among the seventeen factors, was the most important risk factor to develop MPT in upper aerodigestive tract (OR: 7.071, 95% C.I.: 2.134-23.434). For those with high score in polygenic model, routine screening of upper digestive tract including laryngoscope and esophagoscope is suggested to detect new primaries early.
Collapse
Affiliation(s)
- Huei-Tzu Chien
- Department of Nutrition and Health Sciences, Chang Gung University of Science and Technology, Tao-Yuan 33302, Taiwan;
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Tao-Yuan 33302, Taiwan
| | - Chi-Chin Yeh
- Master Program in Applied Molecular Epidemiology, College of Public Health, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Public Health, College of Public Health, China Medical University, Taichung 40402, Taiwan
- Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
| | - Chi-Kuang Young
- Department of Otolaryngology, Chang Gung Memorial Hospital, Keelung 20401, Taiwan;
| | - Tzu-Ping Chen
- Department of Thoracic Surgery, Chang Gung Memorial Hospital, Keelung 20401, Taiwan;
| | - Chun-Ta Liao
- Department of Otolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital, Linkou 33342, Taiwan; (C.-T.L.); (K.-L.C.)
- Medical College, Chang Gung University, Tao-Yuan 33302, Taiwan;
| | - Hung-Ming Wang
- Medical College, Chang Gung University, Tao-Yuan 33302, Taiwan;
- Division of Hematology/Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Tao-Yuan 33342, Taiwan
| | - Kai-Lun Cho
- Department of Otolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital, Linkou 33342, Taiwan; (C.-T.L.); (K.-L.C.)
| | - Shiang-Fu Huang
- Department of Otolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital, Linkou 33342, Taiwan; (C.-T.L.); (K.-L.C.)
- Medical College, Chang Gung University, Tao-Yuan 33302, Taiwan;
- Graduate Institute of Clinical Medical Science, Chang Gung University, Tao-Yuan 33302, Taiwan
- Correspondence: ; Tel.: +88-633-281-200 (ext. 3968); Fax: +88-633-979-361
| |
Collapse
|
6
|
DNA Mismatch Repair Gene Variants in Sporadic Solid Cancers. Int J Mol Sci 2020; 21:ijms21155561. [PMID: 32756484 PMCID: PMC7432688 DOI: 10.3390/ijms21155561] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 07/29/2020] [Accepted: 07/30/2020] [Indexed: 12/18/2022] Open
Abstract
The phenotypic effects of single nucleotide polymorphisms (SNPs) in the development of sporadic solid cancers are still scarce. The aim of this review was to summarise and analyse published data on the associations between SNPs in mismatch repair genes and various cancers. The mismatch repair system plays a unique role in the control of the genetic integrity and it is often inactivated (germline and somatic mutations and hypermethylation) in cancer patients. Here, we focused on germline variants in mismatch repair genes and found the outcomes rather controversial: some SNPs are sometimes ascribed as protective, while other studies reported their pathological effects. Regarding the complexity of cancer as one disease, we attempted to ascertain if particular polymorphisms exert the effect in the same direction in the development and treatment of different malignancies, although it is still not straightforward to conclude whether polymorphisms always play a clear positive role or a negative one. Most recent and robust genome-wide studies suggest that risk of cancer is modulated by variants in mismatch repair genes, for example in colorectal cancer. Our study shows that rs1800734 in MLH1 or rs2303428 in MSH2 may influence the development of different malignancies. The lack of functional studies on many DNA mismatch repair SNPs as well as their interactions are not explored yet. Notably, the concerted action of more variants in one individual may be protective or harmful. Further, complex interactions of DNA mismatch repair variations with both the environment and microenvironment in the cancer pathogenesis will deserve further attention.
Collapse
|
7
|
Rosenberger A, Hung RJ, Christiani DC, Caporaso NE, Liu G, Bojesen SE, Le Marchand L, Haiman CA, Albanes D, Aldrich MC, Tardon A, Fernández-Tardón G, Rennert G, Field JK, Kiemeney B, Lazarus P, Haugen A, Zienolddiny S, Lam S, Schabath MB, Andrew AS, Brunnsstöm H, Goodman GE, Doherty JA, Chen C, Teare MD, Wichmann HE, Manz J, Risch A, Muley TR, Johansson M, Brennan P, Landi MT, Amos CI, Pesch B, Johnen G, Brüning T, Bickeböller H, Gomolka M. Genetic modifiers of radon-induced lung cancer risk: a genome-wide interaction study in former uranium miners. Int Arch Occup Environ Health 2018; 91:937-950. [PMID: 29971594 PMCID: PMC6375683 DOI: 10.1007/s00420-018-1334-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 06/28/2018] [Indexed: 01/10/2023]
Abstract
PURPOSE Radon is a risk factor for lung cancer and uranium miners are more exposed than the general population. A genome-wide interaction analysis was carried out to identify genomic loci, genes or gene sets that modify the susceptibility to lung cancer given occupational exposure to the radioactive gas radon. METHODS Samples from 28 studies provided by the International Lung Cancer Consortium were pooled with samples of former uranium miners collected by the German Federal Office of Radiation Protection. In total, 15,077 cases and 13,522 controls, all of European ancestries, comprising 463 uranium miners were compared. The DNA of all participants was genotyped with the OncoArray. We fitted single-marker and in multi-marker models and performed an exploratory gene-set analysis to detect cumulative enrichment of significance in sets of genes. RESULTS We discovered a genome-wide significant interaction of the marker rs12440014 within the gene CHRNB4 (OR = 0.26, 95% CI 0.11-0.60, p = 0.0386 corrected for multiple testing). At least suggestive significant interaction of linkage disequilibrium blocks was observed at the chromosomal regions 18q21.23 (p = 1.2 × 10-6), 5q23.2 (p = 2.5 × 10-6), 1q21.3 (p = 3.2 × 10-6), 10p13 (p = 1.3 × 10-5) and 12p12.1 (p = 7.1 × 10-5). Genes belonging to the Gene Ontology term "DNA dealkylation involved in DNA repair" (GO:0006307; p = 0.0139) or the gene family HGNC:476 "microRNAs" (p = 0.0159) were enriched with LD-blockwise significance. CONCLUSION The well-established association of the genomic region 15q25 to lung cancer might be influenced by exposure to radon among uranium miners. Furthermore, lung cancer susceptibility is related to the functional capability of DNA damage signaling via ubiquitination processes and repair of radiation-induced double-strand breaks by the single-strand annealing mechanism.
Collapse
Affiliation(s)
- Albert Rosenberger
- Department of Genetic Epidemiology, University Medical Center, Georg August University Göttingen, Humboldtallee 32, 37073, Göttingen, Germany.
| | - Rayjean J Hung
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, University of Toronto, Toronto, ON, Canada
| | - David C Christiani
- Department of Environmental Health, Harvard T.H. Chan School of Public Health and Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Neil E Caporaso
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, Bethesda, MD, USA
| | - Geoffrey Liu
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, University of Toronto, Toronto, ON, Canada
| | - Stig E Bojesen
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen, Denmark
| | - Loic Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Ch A Haiman
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Demetrios Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, Bethesda, MD, USA
| | - Melinda C Aldrich
- Division of Epidemiology, Department of Thoracic Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Adonina Tardon
- Faculty of Medicine, University of Oviedo and CIBERESP, Oviedo, Spain
| | | | - Gad Rennert
- Clalit National Cancer Control Center at Carmel Medical Center and Technion Faculty of Medicine, Haifa, Israel
| | - John K Field
- Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - B Kiemeney
- Departments of Health Evidence and Urology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Philip Lazarus
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, WA, USA
| | - Aage Haugen
- National Institute of Occupational Health, Oslo, Norway
| | | | - Stephen Lam
- British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Matthew B Schabath
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Angeline S Andrew
- Department of Epidemiology, Geisel School of Medicine, Hanover, NH, USA
| | - Hans Brunnsstöm
- Laboratory Medicine Region Skåne, Department of Clinical Sciences and Pathology, Lund University, Lund, Sweden
| | | | - Jennifer A Doherty
- Department of Epidemiology, Geisel School of Medicine, Hanover, NH, USA
- Program in Epidemiology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Population Health Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Chu Chen
- Program in Epidemiology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - M Dawn Teare
- School of Health and Related Research, University of Sheffield, Sheffield, UK
| | - H-Erich Wichmann
- Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Ludwig Maximilians University, Munich, Germany
- Institute of Medical Statistics and Epidemiology, Technical University of Munich, Munich, Germany
| | - Judith Manz
- Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Angela Risch
- Thoraxklinik at University Hospital Heidelberg, Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC-H), Heidelberg, Germany
- University of Salzburg and Cancer Cluster Salzburg, Salzburg, Austria
| | - Thomas R Muley
- Thoraxklinik at University Hospital Heidelberg, Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC-H), Heidelberg, Germany
| | | | - Paul Brennan
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Maria Teresa Landi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, Bethesda, MD, USA
| | - Christopher I Amos
- Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Beate Pesch
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr-Universität Bochum (IPA), Bochum, Germany
| | - Georg Johnen
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr-Universität Bochum (IPA), Bochum, Germany
| | - Thomas Brüning
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr-Universität Bochum (IPA), Bochum, Germany
| | - Heike Bickeböller
- Department of Genetic Epidemiology, University Medical Center, Georg August University Göttingen, Humboldtallee 32, 37073, Göttingen, Germany
| | - Maria Gomolka
- Unit Biological Radiation Effects, Biological Dosimetry, Department of Radiation Protection and Health, Federal Office for Radiation Protection, BfS, Neuherberg, Germany
| |
Collapse
|
8
|
Carrera-Lasfuentes P, Lanas A, Bujanda L, Strunk M, Quintero E, Santolaria S, Benito R, Sopeña F, Piazuelo E, Thomson C, Pérez-Aisa A, Nicolás-Pérez D, Hijona E, Espinel J, Campo R, Manzano M, Geijo F, Pellise M, Zaballa M, González-Huix F, Espinós J, Titó L, Barranco L, D'Amato M, García-González MA. Relevance of DNA repair gene polymorphisms to gastric cancer risk and phenotype. Oncotarget 2018; 8:35848-35862. [PMID: 28415781 PMCID: PMC5482622 DOI: 10.18632/oncotarget.16261] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 02/27/2017] [Indexed: 12/21/2022] Open
Abstract
Variations in DNA repair genes have been reported as key factors in gastric cancer (GC) susceptibility but results among studies are inconsistent. We aimed to assess the relevance of DNA repair gene polymorphisms and environmental factors to GC risk and phenotype in a Caucasian population in Spain. Genomic DNA from 603 patients with primary GC and 603 healthy controls was typed for 123 single nucleotide polymorphisms in DNA repair genes using the Illumina platform. Helicobacter pylori infection with CagA strains (odds ratio (OR): 1.99; 95% confidence interval (CI): 1.55–2.54), tobacco smoking (OR: 1.77; 95% CI: 1.22–2.57), and family history of GC (OR: 2.87; 95% CI: 1.85–4.45) were identified as independent risk factors for GC. By contrast, the TP53 rs9894946A (OR: 0.73; 95% CI: 0.56–0.96), TP53 rs1042522C (OR: 0.76; 95% CI: 0.56–0.96), and BRIP1 rs4986764T (OR: 0.55; 95% CI: 0.38–0.78) variants were associated with lower GC risk. Significant associations with specific anatomopathological GC subtypes were also observed, most notably in the ERCC4 gene with the rs1799801C, rs2238463G, and rs3136038T variants being inversely associated with cardia GC risk. Moreover, the XRCC3 rs861528 allele A was significantly increased in the patient subgroup with diffuse GC (OR: 1.75; 95% CI: 1.30–2.37). Our data show that specific TP53, BRIP1, ERCC4, and XRCC3 polymorphisms are relevant in susceptibility to GC risk and specific subtypes in Caucasians.
Collapse
Affiliation(s)
| | - Angel Lanas
- CIBER de Enfermedades Hepáticas y Digestivas (CIBERehd), Spain.,Instituto de Investigación Sanitaria Aragón (IIS Aragón), Zaragoza, Spain.,Department of Gastroenterology, Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain.,Faculty of Medicine, Universidad de Zaragoza, Zaragoza, Spain
| | - Luis Bujanda
- CIBER de Enfermedades Hepáticas y Digestivas (CIBERehd), Spain.,Department of Gastroenterology, Hospital Donostia/Instituto Biodonostia, Universidad del País Vasco (UPV/EHU), San Sebastián, Spain
| | - Mark Strunk
- CIBER de Enfermedades Hepáticas y Digestivas (CIBERehd), Spain.,Instituto Aragonés de Ciencias de la Salud (IACS), Zaragoza, Spain
| | - Enrique Quintero
- Department of Gastroenterology, Hospital Universitario de Canarias, Instituto Universitario de Tecnologías Biomédicas (ITB), Centro de Investigación Biomédica de Canarias (CIBICAN), Tenerife, Spain
| | | | - Rafael Benito
- CIBER de Enfermedades Hepáticas y Digestivas (CIBERehd), Spain.,Instituto de Investigación Sanitaria Aragón (IIS Aragón), Zaragoza, Spain.,Faculty of Medicine and Department of Microbiology, Hospital Clínico Universitario, Zaragoza, Spain
| | - Federico Sopeña
- CIBER de Enfermedades Hepáticas y Digestivas (CIBERehd), Spain.,Instituto de Investigación Sanitaria Aragón (IIS Aragón), Zaragoza, Spain.,Department of Gastroenterology, Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain
| | - Elena Piazuelo
- CIBER de Enfermedades Hepáticas y Digestivas (CIBERehd), Spain.,Instituto de Investigación Sanitaria Aragón (IIS Aragón), Zaragoza, Spain.,Instituto Aragonés de Ciencias de la Salud (IACS), Zaragoza, Spain
| | - Concha Thomson
- Department of Gastroenterology, Hospital Obispo Polanco, Teruel, Spain
| | | | - David Nicolás-Pérez
- Department of Gastroenterology, Hospital Universitario de Canarias, Instituto Universitario de Tecnologías Biomédicas (ITB), Centro de Investigación Biomédica de Canarias (CIBICAN), Tenerife, Spain
| | - Elizabeth Hijona
- CIBER de Enfermedades Hepáticas y Digestivas (CIBERehd), Spain.,Department of Gastroenterology, Hospital Donostia/Instituto Biodonostia, Universidad del País Vasco (UPV/EHU), San Sebastián, Spain
| | - Jesús Espinel
- Department of Gastroenterology, Complejo Hospitalario, León, Spain
| | - Rafael Campo
- Department of Gastroenterology, Hospital Parc Tauli, Sabadell, Spain
| | - Marisa Manzano
- Department of Gastroenterology, Hospital 12 de Octubre, Madrid, Spain
| | - Fernando Geijo
- Department of Gastroenterology, Hospital Clínico Universitario, Salamanca, Spain
| | - María Pellise
- CIBER de Enfermedades Hepáticas y Digestivas (CIBERehd), Spain.,Department of Gastroenterology, Hospital Clinic I Provincial, Institut d Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universidad de Barcelona, Barcelona, Spain
| | - Manuel Zaballa
- Department of Gastroenterology, Hospital de Cruces, Barakaldo, Spain
| | | | - Jorge Espinós
- Department of Gastroenterology, Mutua de Tarrasa, Spain
| | - Llúcia Titó
- Department of Gastroenterology, Hospital de Mataró, Mataró, Spain
| | - Luis Barranco
- Department of Gastroenterology, Hospital del Mar, Barcelona, Spain
| | - Mauro D'Amato
- BioDonostia Health Research Institute, IKERBASQUE, Basque Foundation for Science, San Sebastián, Spain
| | - María Asunción García-González
- CIBER de Enfermedades Hepáticas y Digestivas (CIBERehd), Spain.,Instituto de Investigación Sanitaria Aragón (IIS Aragón), Zaragoza, Spain.,Instituto Aragonés de Ciencias de la Salud (IACS), Zaragoza, Spain
| |
Collapse
|
9
|
Xiao X, Yang Y, Ren Y, Zou D, Zhang K, Wu Y. rs1760944 Polymorphism in the APE1 Region is Associated with Risk and Prognosis of Osteosarcoma in the Chinese Han Population. Sci Rep 2017; 7:9331. [PMID: 28839218 PMCID: PMC5570937 DOI: 10.1038/s41598-017-09750-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 06/16/2017] [Indexed: 01/16/2023] Open
Abstract
The effects of single nucleotide polymorphisms (SNPs) at APE1 have been investigated in several types of cancer. However, no reports of the association of APE1 polymorphisms with osteosarcoma (OS) have been published. The present study was designed to determine whether APE1 polymorphisms (rs1130409, rs1760944, rs1760941, rs2275008, rs17111750) are associated with OS. A 2-stage case-control study was performed in a total of 378 OS patients and 616 normal controls. Individuals carrying TG and GG genotypes had significantly lower risk of developing OS than those with the WT genotype TT at rs1760944 (OR = 0.65, 95%CI 0.49–0.86; OR = 0.50, 95%CI 0.34–0.74, respectively). OS patients with allele G at rs1760944 were less susceptible to low differentiation tumor and metastasis (OR = 0.73, 95%CI 0.54–0.98; OR = 0.63, 95%CI 0.43–0.92, respectively). Kaplan-Meier curves and log-rank results revealed that OS patients harboring genotype GG and G allele at rs1760944 had better survival (P < 0.001 for both). In addition, the APE1 protein was underexpressed in individuals who carried G allele at rs1760944. This study suggested that APE1 rs1760944 polymorphism is associated with decreased risk of developing OS and better survival of OS patients.
Collapse
Affiliation(s)
- Xing Xiao
- Department of Spine Surgery, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Yun Yang
- School of Medicine, Shandong University, Jinan, Shandong, China
| | - Yanjun Ren
- Department of Spine Surgery, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Debo Zou
- Department of Spine Surgery, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Kaining Zhang
- Department of Spine Surgery, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Yingguang Wu
- Department of Spine Surgery, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China.
| |
Collapse
|
10
|
A miR-SNP biomarker linked to an increased lung cancer survival by miRNA-mediated down-regulation of FZD4 expression and Wnt signaling. Sci Rep 2017; 7:9029. [PMID: 28831115 PMCID: PMC5567228 DOI: 10.1038/s41598-017-09604-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 07/21/2017] [Indexed: 12/18/2022] Open
Abstract
Through a new hypothesis-driven and microRNA-pathway-based SNP (miR-SNP) association study we identified a novel miR-SNP (rs713065) in the 3'UTR region of FZD4 gene linked with decreased risk of death in early stage NSCLC patients. We determined biological function and mechanism of action of this FZD4-miR-SNP biomarker in a cellular platform. Our data suggest that FZD4-miR-SNP loci may significantly influence overall survival in NSCLC patients by specifically interacting with miR-204 and modulating FZD4 expression and cellular function in the Wnt-signaling-driven tumor progression. Our findings are bridging the gap between the discovery of epidemiological SNP biomarkers and their biological function and will enable us to develop novel therapeutic strategies that specifically target epigenetic markers in the oncogenic Wnt/FZD signaling pathways in NSCLC.
Collapse
|
11
|
Rosenberger A, Sohns M, Friedrichs S, Hung RJ, Fehringer G, McLaughlin J, Amos CI, Brennan P, Risch A, Brüske I, Caporaso NE, Landi MT, Christiani DC, Wei Y, Bickeböller H. Gene-set meta-analysis of lung cancer identifies pathway related to systemic lupus erythematosus. PLoS One 2017; 12:e0173339. [PMID: 28273134 PMCID: PMC5342225 DOI: 10.1371/journal.pone.0173339] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 02/20/2017] [Indexed: 02/03/2023] Open
Abstract
INTRODUCTION Gene-set analysis (GSA) is an approach using the results of single-marker genome-wide association studies when investigating pathways as a whole with respect to the genetic basis of a disease. METHODS We performed a meta-analysis of seven GSAs for lung cancer, applying the method META-GSA. Overall, the information taken from 11,365 cases and 22,505 controls from within the TRICL/ILCCO consortia was used to investigate a total of 234 pathways from the Kyoto Encyclopedia of Genes and Genomes (KEGG) database. RESULTS META-GSA reveals the systemic lupus erythematosus KEGG pathway hsa05322, driven by the gene region 6p21-22, as also implicated in lung cancer (p = 0.0306). This gene region is known to be associated with squamous cell lung carcinoma. The most important genes driving the significance of this pathway belong to the genomic areas HIST1-H4L, -1BN, -2BN, -H2AK, -H4K and C2/C4A/C4B. Within these areas, the markers most significantly associated with LC are rs13194781 (located within HIST12BN) and rs1270942 (located between C2 and C4A). CONCLUSIONS We have discovered a pathway currently marked as specific to systemic lupus erythematosus as being significantly implicated in lung cancer. The gene region 6p21-22 in this pathway appears to be more extensively associated with lung cancer than previously assumed. Given wide-stretched linkage disequilibrium to the area APOM/BAG6/MSH5, there is currently simply not enough information or evidence to conclude whether the potential pleiotropy of lung cancer and systemic lupus erythematosus is spurious, biological, or mediated. Further research into this pathway and gene region will be necessary.
Collapse
Affiliation(s)
- Albert Rosenberger
- Department of Genetic Epidemiology, University Medical Center, Georg-August-University Göttingen, Göttingen, Germany
| | - Melanie Sohns
- Department of Genetic Epidemiology, University Medical Center, Georg-August-University Göttingen, Göttingen, Germany
| | - Stefanie Friedrichs
- Department of Genetic Epidemiology, University Medical Center, Georg-August-University Göttingen, Göttingen, Germany
| | - Rayjean J. Hung
- Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - Gord Fehringer
- Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, Canada
| | | | - Christopher I. Amos
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Paul Brennan
- International Agency for Research on Cancer, Lyon, France
| | - Angela Risch
- Division of Molecular Biology, University Salzburg, Salzburg, Austria
| | - Irene Brüske
- Institute of Epidemiology I, Helmholtz Center Munich, Munich, Germany
| | - Neil E. Caporaso
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Maria Teresa Landi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, United States of America
| | - David C. Christiani
- Harvard University School of Public Health, Boston, Massachusetts, United States of America
| | - Yongyue Wei
- Harvard University School of Public Health, Boston, Massachusetts, United States of America
| | - Heike Bickeböller
- Department of Genetic Epidemiology, University Medical Center, Georg-August-University Göttingen, Göttingen, Germany
| |
Collapse
|
12
|
Couto PG, Bastos-Rodrigues L, Carneiro JG, Guieiro F, Bicalho MA, Leidenz FB, Bicalho AJ, Friedman E, De Marco L. DNA Base-Excision Repair Genes OGG1 and NTH1 in Brazilian Lung Cancer Patients. Mol Diagn Ther 2016; 19:389-95. [PMID: 26400813 DOI: 10.1007/s40291-015-0164-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Lung cancer is the leading global cause of cancer-related mortality and is associated with poor prognosis. To improve survival rates of lung cancer patients, better understanding of tumorigenic mechanisms is necessary, which may lead to development of new therapeutic strategies. The hOGG1 and NTH1 genes act in the DNA BER repair pathway and their involvement in lung cancer pathogenesis has been analyzed in several populations. METHODS We analyzed targeted regions of the hOGG1 and NTH1 genes in 96 Brazilian patients with non-small-cell lung cancer (NSCLC) and 89 cancer-free, ethnically matched controls. RESULTS The NTH1 c.98G>T polymorphism rs2302172 (p = 0.02 and p = 0.02 for allele and genotype frequency between cases and controls, respectively) and the 140-17C> T variant (rs2233518) (p = 0.02 and p = 0.02 for allele and genotype frequency between cases and controls, respectively) were detected in four lung cancer cases (4 %) while the NTH1 Q131K (C391A) polymorphism was found in seven lung cancer cases (7 %) (p = 0.001 and p = 0.008, for allele and genotype frequency between cases and controls, respectively). None of these sequence variants were detected in controls. The Ser326Cys (C1245G, rs1052133) polymorphism in the OGG1 gene was detected in 42 % of analyzed NSCLC patients and in 34 % of the controls (p = 0.11 and p = 0.25 for allele and genotype frequency between cases and controls, respectively). CONCLUSIONS Our study provides preliminary evidence that polymorphisms in OGG1 do not contribute to development of NSCLC in Brazilian patients and that NTH1 polymorphisms may be associated with NSCLC pathogenesis.
Collapse
Affiliation(s)
- Patricia G Couto
- Department of Surgery, Universidade Federal de Minas Gerais, Av. Alfredo Balena 190, Room 114, Belo Horizonte, 30130-100, Brazil
| | - Luciana Bastos-Rodrigues
- Universidade Federal de Juiz de Fora, Campus Governador Valadares, Governador Valadares, Minas Gerais, Brazil
| | - Juliana G Carneiro
- Faculty of Medical Science, Centro de Ensino Superior e Desenvolvimento, Campina Grande, Brazil
| | - Fernanda Guieiro
- Department of Surgery, Universidade Federal de Minas Gerais, Av. Alfredo Balena 190, Room 114, Belo Horizonte, 30130-100, Brazil
| | | | - Franciele B Leidenz
- Department of Surgery, Universidade Federal de Minas Gerais, Av. Alfredo Balena 190, Room 114, Belo Horizonte, 30130-100, Brazil
| | - Ana J Bicalho
- Department of Surgery, Universidade Federal de Minas Gerais, Av. Alfredo Balena 190, Room 114, Belo Horizonte, 30130-100, Brazil
| | - Eitan Friedman
- The Susanne Levy Gertner Oncogenetics Unit, Chaim Sheba Medical Center, Tel-Hashomer, Israel
| | - Luiz De Marco
- Department of Surgery, Universidade Federal de Minas Gerais, Av. Alfredo Balena 190, Room 114, Belo Horizonte, 30130-100, Brazil.
| |
Collapse
|
13
|
Wang M, Liu H, Liu Z, Yi X, Bickeboller H, Hung RJ, Brennan P, Landi MT, Caporaso N, Christiani DC, Doherty JA, Amos CI, Wei Q. Genetic variant in DNA repair gene GTF2H4 is associated with lung cancer risk: a large-scale analysis of six published GWAS datasets in the TRICL consortium. Carcinogenesis 2016; 37:888-896. [PMID: 27288692 PMCID: PMC5008248 DOI: 10.1093/carcin/bgw070] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 05/27/2016] [Accepted: 06/07/2016] [Indexed: 12/15/2022] Open
Abstract
DNA repair pathways maintain genomic integrity and stability, and dysfunction of DNA repair leads to cancer. We hypothesize that functional genetic variants in DNA repair genes are associated with risk of lung cancer. We performed a large-scale meta-analysis of 123,371 single nucleotide polymorphisms (SNPs) in 169 DNA repair genes obtained from six previously published genome-wide association studies (GWASs) of 12160 lung cancer cases and 16838 controls. We calculated odds ratios (ORs) with 95% confidence intervals (CIs) using the logistic regression model and used the false discovery rate (FDR) method for correction of multiple testing. As a result, 14 SNPs had a significant odds ratio (OR) for lung cancer risk with P FDR < 0.05, of which rs3115672 in MSH5 (OR = 1.20, 95% CI = 1.14-1.27) and rs114596632 in GTF2H4 (OR = 1.19, 95% CI = 1.12-1.25) at 6q21.33 were the most statistically significant (P combined = 3.99×10(-11) and P combined = 5.40×10(-10), respectively). The MSH5 rs3115672, but not GTF2H4 rs114596632, was strongly correlated with MSH5 rs3131379 in that region (r (2) = 1.000 and r (2) = 0.539, respectively) as reported in a previous GWAS. Importantly, however, the GTF2H4 rs114596632 T, but not MSH5 rs3115672 T, allele was significantly associated with both decreased DNA repair capacity phenotype and decreased mRNA expression levels. These provided evidence that functional genetic variants of GTF2H4 confer susceptibility to lung cancer.
Collapse
Affiliation(s)
- Meilin Wang
- Duke Cancer Institute, Duke University Medical Center, 905 Lasalle Street, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 21116, China
| | - Hongliang Liu
- Duke Cancer Institute, Duke University Medical Center, 905 Lasalle Street, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Zhensheng Liu
- Duke Cancer Institute, Duke University Medical Center, 905 Lasalle Street, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Xiaohua Yi
- Duke Cancer Institute, Duke University Medical Center, 905 Lasalle Street, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Heike Bickeboller
- Department of Genetic Epidemiology, University Medical Center, Georg-August-University Göttingen, 37073 Göttingen, Germany
| | - Rayjean J. Hung
- Lunenfeld-Tanenbaum Research Institute of Mount Sinai hospital, Toronto, Ontario, Canada
| | - Paul Brennan
- Genetic Epidemiology Group, International Agency for Research on Cancer, 69372 Lyon, France
| | - Maria Teresa Landi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA
| | - Neil Caporaso
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA
| | - David C. Christiani
- Department of Environmental Health and
- Department of Epidemiology, Harvard University School of Public Health, Boston, MA 02115, USA and
| | - Jennifer Anne Doherty
- Community and Family Medicine, Geisel School of Medicine, Dartmouth College, Hanover, NH 03756, USA
| | - The TRICL Research Team
- Duke Cancer Institute, Duke University Medical Center, 905 Lasalle Street, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 21116, China
- Department of Genetic Epidemiology, University Medical Center, Georg-August-University Göttingen, 37073 Göttingen, Germany
- Lunenfeld-Tanenbaum Research Institute of Mount Sinai hospital, Toronto, Ontario, Canada
- Genetic Epidemiology Group, International Agency for Research on Cancer, 69372 Lyon, France
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA
- Department of Environmental Health and
- Department of Epidemiology, Harvard University School of Public Health, Boston, MA 02115, USA and
- Community and Family Medicine, Geisel School of Medicine, Dartmouth College, Hanover, NH 03756, USA
| | - Christopher I. Amos
- Community and Family Medicine, Geisel School of Medicine, Dartmouth College, Hanover, NH 03756, USA
| | - Qingyi Wei
- Duke Cancer Institute, Duke University Medical Center, 905 Lasalle Street, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
14
|
Xie H, Tao W, Wu X, Gu J. Genetic variations in apoptosis pathway and the risk of ovarian cancer. Oncotarget 2016; 7:56737-56745. [PMID: 27462919 PMCID: PMC5302949 DOI: 10.18632/oncotarget.10772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 06/09/2016] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Apoptosis is a highly conserved form of cell death and aberrant regulation of apoptotic cell death mechanisms leads to variety of major human diseases, especially tumor formation. Genetic variations in apoptosis genes may increase susceptibility to ovarian cancer. RESULTS In individual SNP analysis, 12 SNPs in 5 apoptosis pathway genes were significantly associated with ovarian cancer risk after adjustment for multiple comparisons at q-value <0.05. The most significant SNP was rs11152377 in the Bcl-2 gene. The homozygous variant TT genotype was associated with a significantly decreased risk of ovarian cancer (odds ratio [OR] =0.53; 95% confidence interval [CI], 0.37-0.77, P<0.001). Cumulative effect analysis showed joint effects of increased risk of ovarian cancer with increasing number of unfavorable genotypes in patients. Classification and regression tree (CART) analysis further revealed high-order gene-gene interactions and categorized the study subjects into low-, medium-, and high-risk groups. Compared with the low-risk group, medium-risk group and high-risk group conferred 1.76-fold (95% CI: 1.06-2.90) and 3.64-fold (95% CI: 2.37-5.59) increased risk of ovarian cancer (P for trend <0.001)Materials and Methods: In a case-control study of 417 ovarian cancer patients and 417 matched controls, we evaluated the associations of 587 single nucleotide polymorphisms (SNPs) from 65 genes of the apoptosis pathway with the risk of ovarian cancer. CONCLUSIONS Our results suggest that genetic variations in apoptosis pathway genes modulate the risk of ovarian cancer individually and jointly.
Collapse
Affiliation(s)
- Hui Xie
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Wade Tao
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Xifeng Wu
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jian Gu
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
15
|
Wang C, Nie H, Li Y, Liu G, Wang X, Xing S, Zhang L, Chen X, Chen Y, Li Y. The study of the relation of DNA repair pathway genes SNPs and the sensitivity to radiotherapy and chemotherapy of NSCLC. Sci Rep 2016; 6:26526. [PMID: 27246533 PMCID: PMC4887885 DOI: 10.1038/srep26526] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 05/04/2016] [Indexed: 12/18/2022] Open
Abstract
To analyze the relation between SNPs in DNA repair pathway-related genes and sensitivity of tumor radio-chemotherapy, 26 SNPs in 20 DNA repair genes were genotyped on 176 patients of NSCLC undertaking radio-chemotherapy treatment. In squamous cell carcinoma (SCC), as the rs2228000, rs2228001 (XPC), rs2273953 (TP73), rs2279744 (MDM2), rs2299939 (PTEN) and rs8178085, rs12334811 (DNA-PKcs) affected the sensitivity to chemotherapy, so did the rs8178085, rs12334811 to radiotherapy. Moreover rs344781, rs2273953 and rs12334811 were related with the survival time of SCC. In general, the “good” genotype GG (rs12334811) showed greater efficacy of radio-chemotherapy and MSF (24 months) on SCC. In adenocarcinoma, as the rs2699887 (PIK3), rs12334811 (DNA-PKcs) influenced the sensitivity to chemotherapy, so did the rs2299939, rs2735343 (PTEN) to radiotherapy. And rs402710, rs80270, rs2279744 and rs2909430 impacted the survival time of the adenocarcinoma patients. Both GG (rs2279744) and AG (rs2909430) showed a shorter survival time (MFS = 6). Additionally, some SNPs such as rs2228000, rs2228001 and rs344781 were found to regulate the expression of DNA repair pathway genes through eQTLs dataset analysis. These results indicate that SNPs in DNA repair pathway genes might regulate the expression and affect the DNA damage repair, and thereby impact the efficacy of radio-chemotherapy and the survival time of NSCLC.
Collapse
Affiliation(s)
- Chunbo Wang
- School of Life Science and Technology, Harbin Institute of Technology, Heilongjiang, China.,Department of Radiotherapy, Affiliated Tumour Hospital of Harbin Medical University
| | - Huan Nie
- School of Life Science and Technology, Harbin Institute of Technology, Heilongjiang, China
| | - Yiqun Li
- School of Life Science and Technology, Harbin Institute of Technology, Heilongjiang, China
| | - Guiyou Liu
- School of Life Science and Technology, Harbin Institute of Technology, Heilongjiang, China.,Genome Analysis Laboratory, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, China
| | - Xu Wang
- Department of Radiotherapy, Beijing Miyun County Hospital
| | - Shijie Xing
- School of Life Science and Technology, Harbin Institute of Technology, Heilongjiang, China
| | - Liping Zhang
- Department of Radiotherapy, Affiliated Tumour Hospital of Harbin Medical University
| | - Xin Chen
- Department of Radiotherapy, Affiliated Tumour Hospital of Harbin Medical University
| | - Yue Chen
- School of Life Science and Technology, Harbin Institute of Technology, Heilongjiang, China
| | - Yu Li
- School of Life Science and Technology, Harbin Institute of Technology, Heilongjiang, China
| |
Collapse
|
16
|
Saunders EJ, Dadaev T, Leongamornlert DA, Olama AAA, Benlloch S, Giles GG, Wiklund F, Grönberg H, Haiman CA, Schleutker J, Nordestgaard BG, Travis RC, Neal D, Pasayan N, Khaw KT, Stanford JL, Blot WJ, Thibodeau SN, Maier C, Kibel AS, Cybulski C, Cannon-Albright L, Brenner H, Park JY, Kaneva R, Batra J, Teixeira MR, Pandha H, Govindasami K, Muir K, Easton DF, Eeles RA, Kote-Jarai Z. Gene and pathway level analyses of germline DNA-repair gene variants and prostate cancer susceptibility using the iCOGS-genotyping array. Br J Cancer 2016; 114:945-52. [PMID: 26964030 PMCID: PMC5379914 DOI: 10.1038/bjc.2016.50] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 02/05/2016] [Accepted: 02/09/2016] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Germline mutations within DNA-repair genes are implicated in susceptibility to multiple forms of cancer. For prostate cancer (PrCa), rare mutations in BRCA2 and BRCA1 give rise to moderately elevated risk, whereas two of B100 common, low-penetrance PrCa susceptibility variants identified so far by genome-wide association studies implicate RAD51B and RAD23B. METHODS Genotype data from the iCOGS array were imputed to the 1000 genomes phase 3 reference panel for 21 780 PrCa cases and 21 727 controls from the Prostate Cancer Association Group to Investigate Cancer Associated Alterations in the Genome (PRACTICAL) consortium. We subsequently performed single variant, gene and pathway-level analyses using 81 303 SNPs within 20 Kb of a panel of 179 DNA-repair genes. RESULTS Single SNP analyses identified only the previously reported association with RAD51B. Gene-level analyses using the SKAT-C test from the SNP-set (Sequence) Kernel Association Test (SKAT) identified a significant association with PrCa for MSH5. Pathway-level analyses suggested a possible role for the translesion synthesis pathway in PrCa risk and Homologous recombination/Fanconi Anaemia pathway for PrCa aggressiveness, even though after adjustment for multiple testing these did not remain significant. CONCLUSIONS MSH5 is a novel candidate gene warranting additional follow-up as a prospective PrCa-risk locus. MSH5 has previously been reported as a pleiotropic susceptibility locus for lung, colorectal and serous ovarian cancers.
Collapse
Affiliation(s)
- Edward J Saunders
- The Institute of Cancer Research
& Royal Marsden NHS Foundation Trust, 123 Old Brompton
Rd, London
SW7 3RP, UK
| | - Tokhir Dadaev
- The Institute of Cancer Research
& Royal Marsden NHS Foundation Trust, 123 Old Brompton
Rd, London
SW7 3RP, UK
| | - Daniel A Leongamornlert
- The Institute of Cancer Research
& Royal Marsden NHS Foundation Trust, 123 Old Brompton
Rd, London
SW7 3RP, UK
| | - Ali Amin Al Olama
- Centre for Cancer Genetic
Epidemiology, Department of Public Health and Primary Care, University of
Cambridge, Strangeways Laboratory, Worts Causeway,
Cambridge
CB1 8RN, UK
| | - Sara Benlloch
- Centre for Cancer Genetic
Epidemiology, Department of Public Health and Primary Care, University of
Cambridge, Strangeways Laboratory, Worts Causeway,
Cambridge
CB1 8RN, UK
| | - Graham G Giles
- Cancer Epidemiology Centre, The
Cancer Council Victoria, 1 Rathdowne Street,
Carlton Victoria, Australia
- Centre for Molecular, Environmental,
Genetic and Analytic Epidemiology, The University of Melbourne
3053, Victoria, Australia
| | - Fredrik Wiklund
- Department of Medical Epidemiology
and Biostatistics, Karolinska Institute, Stockholm
17177, Sweden
| | - Henrik Grönberg
- Department of Medical Epidemiology
and Biostatistics, Karolinska Institute, Stockholm
17177, Sweden
| | - Christopher A Haiman
- Department of Preventive Medicine,
Keck School of Medicine, University of Southern California & Norris
Comprehensive Cancer Center, Los Angeles,
CA
90089, USA
| | - Johanna Schleutker
- Department of Medical Biochemistry
and Genetics, University of Turku, Turku,
Finland
- Institute of Biomedical Technology
and BioMediTech, University of Tampere and FimLab Laboratories,
Tampere
33520, Finland
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry,
Herlev and Gentofte Hospital, Copenhagen University Hospital,
Herlev Ringvej 75
DK-2730, Herlev, Denmark
| | - Ruth C Travis
- Cancer Epidemiology Unit, Nuffield
Department of Population Health, University of Oxford,
Oxford
OX3 7LF, UK
| | - David Neal
- Surgical Oncology (Uro-Oncology:
S4), University of Cambridge, Addenbrooke's Hospital, Hills Road,
Cambridge & Cancer Research UK Cambridge Research Institute, Li Ka
Shing Centre, Cambridge
CB2 2QQ, UK
| | - Nora Pasayan
- University College London,
Department of Applied Health Research, 1-19 Torrington
Place, London
WC1E 7HB, UK
| | - Kay-Tee Khaw
- Cambridge Institute of Public
Health, University of Cambridge, Forvie Site, Robinson
Way, Cambridge
CB2 0SR, UK
| | - Janet L Stanford
- Department of Epidemiology, School
of Public Health, University of Washington & Division of Public
Health Sciences, Fred Hutchinson Cancer Research Center,
Seattle, WA, USA
| | - William J Blot
- International Epidemiology
Institute, 1455 Research Blvd., Suite 550,
Rockville
MD 20850, USA
| | | | - Christiane Maier
- Institute of Human Genetics,
University Hospital Ulm, Ulm
89075, Germany
| | - Adam S Kibel
- Division of Urologic Surgery,
Brigham and Women's Hospital, Dana-Farber Cancer Institute,
45 Francis Street- ASB II-3
Boston, MA, 02245,
USA
| | - Cezary Cybulski
- International Hereditary Cancer
Center, Department of Genetics and Pathology, Pomeranian Medical
University, Szczecin
70-115, Poland
| | - Lisa Cannon-Albright
- Division of Genetic Epidemiology,
Department of Medicine, University of Utah School of Medicine &
George E. Wahlen Department of Veterans Affairs Medical Center,
Salt Lake City, UT
84132, USA
| | - Hermann Brenner
- Division of Clinical Epidemiology
and Aging Research, German Cancer Research Center (DKFZ), Heidelberg
& Division of Preventive Oncology, German Cancer Research Center
(DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg &
German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ),
Heidelberg, Germany
| | - Jong Y Park
- Department of Cancer Epidemiology,
H. Lee Moffitt Cancer Center, 12902 Magnolia Drive,
Tampa, FL
33612, USA
| | - Radka Kaneva
- Molecular Medicine Center and
Department of Medical Chemistry and Biochemistry, Medical University -
Sofia, 2 Zdrave Street, Sofia
1431, Bulgaria
| | - Jyotsna Batra
- Australian Prostate Cancer Research
Centre-Qld, Institute of Health and Biomedical Innovation & School
of Biomedical Science, Queensland University of Technology,
Brisbane
4102, Australia
| | - Manuel R Teixeira
- Biomedical Sciences Institute
(ICBAS), Porto University, Porto, Portugal
- Department of Genetics, Portuguese
Oncology Institute, Porto, Portugal
4200-072, Portugal
| | - Hardev Pandha
- The University of Surrey,
Guildford, Surrey
GU2 7XH, UK
| | - Koveela Govindasami
- The Institute of Cancer Research
& Royal Marsden NHS Foundation Trust, 123 Old Brompton
Rd, London
SW7 3RP, UK
| | - Ken Muir
- Warwick Medical School, University
of Warwick, Coventry
CV4 7AL, UK
| | - Douglas F Easton
- Centre for Cancer Genetic
Epidemiology, Department of Public Health and Primary Care, University of
Cambridge, Strangeways Laboratory, Worts Causeway,
Cambridge
CB1 8RN, UK
| | - Rosalind A Eeles
- The Institute of Cancer Research
& Royal Marsden NHS Foundation Trust, 123 Old Brompton
Rd, London
SW7 3RP, UK
| | - Zsofia Kote-Jarai
- The Institute of Cancer Research
& Royal Marsden NHS Foundation Trust, 123 Old Brompton
Rd, London
SW7 3RP, UK
| |
Collapse
|
17
|
Mamdani H, Jalal SI. DNA repair in lung cancer: potential not yet reached. Lung Cancer Manag 2016; 5:5-8. [PMID: 30643544 DOI: 10.2217/lmt-2016-0004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 03/04/2016] [Indexed: 11/21/2022] Open
Affiliation(s)
- Hirva Mamdani
- Division of Hematology/Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.,Indiana University Melvin & Bren Simon Cancer Center, 980 W Walnut Street, Indianapolis, IN 46202, IN, USA.,Division of Hematology/Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.,Indiana University Melvin & Bren Simon Cancer Center, 980 W Walnut Street, Indianapolis, IN 46202, IN, USA
| | - Shadia I Jalal
- Division of Hematology/Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.,Indiana University Melvin & Bren Simon Cancer Center, 980 W Walnut Street, Indianapolis, IN 46202, IN, USA.,Division of Hematology/Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.,Indiana University Melvin & Bren Simon Cancer Center, 980 W Walnut Street, Indianapolis, IN 46202, IN, USA
| |
Collapse
|
18
|
Catana A, Pop M, Marginean DH, Blaga IC, Porojan MD, Popp RA, Pop IV. XRCC3 Thr241Met Polymorphism is not Associated with Lung Cancer Risk in a Romanian Population. ACTA ACUST UNITED AC 2016; 89:89-93. [PMID: 27004030 PMCID: PMC4777474 DOI: 10.15386/cjmed-523] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 09/28/2015] [Accepted: 10/04/2015] [Indexed: 11/23/2022]
Abstract
BACKGROUND AND AIMS Deoxyribonucleic Acid (DNA) repair mechanisms play a critical role in protecting the cellular genome against carcinogens. X-ray cross-complementing gene 3 (XRCC3) is involved in DNA repair and therefore certain genetic polymorphisms that occur in DNA repair genes may affect the ability to repair DNA defects and may represent a risk factor in carcinogenesis. The purpose of our study was to investigate the association between XRCC3 gene substitution of Threonine with Methionine in codon 241 of XRCC3 gene (Thr241Met) polymorphism and the risk of lung cancer, in a Romanian population. METHODS We recruited 93 healthy controls and 85 patients with lung cancer, all smokers. Thr241Met, XRCC3 gene genotyping was determined by multiplex Polymerase Chain Reaction-Restriction Fragment Length Polymorphism (PCR-RFLP). RESULTS Statistical analysis (OR, recessive model), did not revealed an increased risk for lung cancer, for the variant 241Met allele and Thr241Met genotypes (p=0.138, OR=0.634, CI=0.348-1.157; p=0.023, OR=0.257, CI=0.085-6.824). Also, there were no positive statistical associations between Thr241Met polymorphism of XRCC3 gene, gender, tobacco and various histopathological tumor type of lung cancer. CONCLUSION In conclusion, the results of the study suggest that the XRCC3 gene Thr241Met polymorphism is not associated with an increased risk for the development of lung cancer in this Romanian group.
Collapse
Affiliation(s)
- Andreea Catana
- Department of Molecular Science, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Monica Pop
- Department of Pneumology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Dragos Horea Marginean
- Department of Molecular Science, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ioana Cristina Blaga
- Department of Molecular Science, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Mihai Dumitru Porojan
- Internal Medicine Department, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Radu Anghel Popp
- Department of Molecular Science, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ioan Victor Pop
- Department of Molecular Science, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
19
|
Scarbrough PM, Weber RP, Iversen ES, Brhane Y, Amos CI, Kraft P, Hung RJ, Sellers TA, Witte JS, Pharoah P, Henderson BE, Gruber SB, Hunter DJ, Garber JE, Joshi AD, McDonnell K, Easton DF, Eeles R, Kote-Jarai Z, Muir K, Doherty JA, Schildkraut JM. A Cross-Cancer Genetic Association Analysis of the DNA Repair and DNA Damage Signaling Pathways for Lung, Ovary, Prostate, Breast, and Colorectal Cancer. Cancer Epidemiol Biomarkers Prev 2016; 25:193-200. [PMID: 26637267 PMCID: PMC4713268 DOI: 10.1158/1055-9965.epi-15-0649] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 10/05/2015] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND DNA damage is an established mediator of carcinogenesis, although genome-wide association studies (GWAS) have identified few significant loci. This cross-cancer site, pooled analysis was performed to increase the power to detect common variants of DNA repair genes associated with cancer susceptibility. METHODS We conducted a cross-cancer analysis of 60,297 single nucleotide polymorphisms, at 229 DNA repair gene regions, using data from the NCI Genetic Associations and Mechanisms in Oncology (GAME-ON) Network. Our analysis included data from 32 GWAS and 48,734 controls and 51,537 cases across five cancer sites (breast, colon, lung, ovary, and prostate). Because of the unavailability of individual data, data were analyzed at the aggregate level. Meta-analysis was performed using the Association analysis for SubSETs (ASSET) software. To test for genetic associations that might escape individual variant testing due to small effect sizes, pathway analysis of eight DNA repair pathways was performed using hierarchical modeling. RESULTS We identified three susceptibility DNA repair genes, RAD51B (P < 5.09 × 10(-6)), MSH5 (P < 5.09 × 10(-6)), and BRCA2 (P = 5.70 × 10(-6)). Hierarchical modeling identified several pleiotropic associations with cancer risk in the base excision repair, nucleotide excision repair, mismatch repair, and homologous recombination pathways. CONCLUSIONS Only three susceptibility loci were identified, which had all been previously reported. In contrast, hierarchical modeling identified several pleiotropic cancer risk associations in key DNA repair pathways. IMPACT Results suggest that many common variants in DNA repair genes are likely associated with cancer susceptibility through small effect sizes that do not meet stringent significance testing criteria.
Collapse
Affiliation(s)
- Peter M Scarbrough
- Department of Community and Family Medicine, Duke University Medical Center, Durham, North Carolina. Cancer Prevention, Detection, and Control Research Program, Duke Cancer Institute, Durham, North Carolina. Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - Rachel Palmieri Weber
- Department of Community and Family Medicine, Duke University Medical Center, Durham, North Carolina. Cancer Prevention, Detection, and Control Research Program, Duke Cancer Institute, Durham, North Carolina
| | - Edwin S Iversen
- Department of Statistical Science, Duke University, Durham, North Carolina
| | - Yonathan Brhane
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | | | - Peter Kraft
- Program in Genetic Epidemiology and Statistical Genetics, Harvard School of Public Health, Boston, Massachusetts
| | - Rayjean J Hung
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Thomas A Sellers
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, Florida
| | - John S Witte
- Institute for Human Genetics, University of California, San Francisco, San Francisco, California. Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California
| | - Paul Pharoah
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom. Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Brian E Henderson
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California. Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California
| | - Stephen B Gruber
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California. Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California
| | - David J Hunter
- Program in Genetic Epidemiology and Statistical Genetics, Harvard School of Public Health, Boston, Massachusetts
| | - Judy E Garber
- Cancer Risk and Prevention Clinic, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Amit D Joshi
- Program in Genetic Epidemiology and Statistical Genetics, Harvard School of Public Health, Boston, Massachusetts
| | - Kevin McDonnell
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California. Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California
| | - Doug F Easton
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Ros Eeles
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, United Kingdom. Royal Marsden NHS Foundation Trust, London and Sutton, United Kingdom
| | - Zsofia Kote-Jarai
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, United Kingdom. Royal Marsden NHS Foundation Trust, London and Sutton, United Kingdom
| | - Kenneth Muir
- Institute of Population Health, University of Manchester, Manchester, United Kingdom. Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | | | - Joellen M Schildkraut
- Department of Community and Family Medicine, Duke University Medical Center, Durham, North Carolina. Cancer Prevention, Detection, and Control Research Program, Duke Cancer Institute, Durham, North Carolina. Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia.
| |
Collapse
|
20
|
Verde Z, Reinoso L, Chicharro LM, Resano P, Sánchez-Hernández I, Rodríguez González-Moro JM, Bandrés F, Gómez-Gallego F, Santiago C. Are SNP-Smoking Association Studies Needed in Controls? DNA Repair Gene Polymorphisms and Smoking Intensity. PLoS One 2015; 10:e0129374. [PMID: 26017978 PMCID: PMC4446361 DOI: 10.1371/journal.pone.0129374] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 05/07/2015] [Indexed: 02/01/2023] Open
Abstract
Variations in tobacco-related cancers, incidence and prevalence reflect differences in tobacco consumption in addition to genetic factors. Besides, genes related to lung cancer risk could be related to smoking behavior. Polymorphisms altering DNA repair capacity may lead to synergistic effects with tobacco carcinogen-induced lung cancer risk. Common problems in genetic association studies, such as presence of gene-by-environment (G x E) correlation in the population, may reduce the validity of these designs. The main purpose of this study was to evaluate the independence assumption for selected SNPs and smoking behaviour in a cohort of 320 healthy Spanish smokers. We found an association between the wild type alleles of XRCC3 Thr241Met or KLC3 Lys751Gln and greater smoking intensity (OR = 12.98, 95% CI = 2.86–58.82 and OR=16.90, 95% CI=2.09-142.8; respectively). Although preliminary, the results of our study provide evidence that genetic variations in DNA-repair genes may influence both smoking habits and the development of lung cancer. Population-specific G x E studies should be carried out when genetic and environmental factors interact to cause the disease.
Collapse
Affiliation(s)
- Zoraida Verde
- Department of Morphological Sciences and Biomedicine, Universidad Europea, Madrid, Spain
- * E-mail:
| | - Luis Reinoso
- Department of Morphological Sciences and Biomedicine, Universidad Europea, Madrid, Spain
- Department of Occupational Health, Grupo Banco Popular, Madrid, Spain
| | - Luis Miguel Chicharro
- Department of Morphological Sciences and Biomedicine, Universidad Europea, Madrid, Spain
| | - Pilar Resano
- Department of Neumology, Hospital Guadalajara, Guadalajara, Spain
| | | | | | - Fernando Bandrés
- Department of Toxicology and Health Sanitary, Universidad Complutense, Madrid, Spain
| | | | - Catalina Santiago
- School of Doctoral Studies & Research, Universidad Europea, Madrid, Spain
| |
Collapse
|
21
|
Yang HY, Yang SY, Shao FY, Wang HY, Wang YD. Updated assessment of the association of the XRCC1 Arg399Gln polymorphism with lung cancer risk in the Chinese population. Asian Pac J Cancer Prev 2015; 16:495-500. [PMID: 25684477 DOI: 10.7314/apjcp.2015.16.2.495] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Published studies have reported relationships between X-ray repair cross-complementing group 1 (XRCC1) Arg399Gln polymorphism and lung cancer risk in Chinese population. However, the epidemiological results remained controversial. The objective of this study was to clarify the association of XRCC1 Arg399Gln polymorphism with lung cancer risk in the Chinese population. MATERIALS AND METHODS Systematic searches were performed through the database of Medline/Pubmed, Web of Science, Embase, CNKI and WanFang Medical Online. Odds ratios (ORs) with 95% confidence interval (95%CI) were calculated to estimate the strength of the association. RESULTS Overall, we observed an increased lung cancer risk among subjects carrying XRCC1 codon 399 Gln/Gln genotype (OR=1.36, 95%CI: 1.09-1.71) in the Chinese population on the basis of 19 studies with 5,416 cases and 5,782 controls. We did not observe any association between XRCC1 codon 399 Arg/Gln and Arg/Gln+Gln/Gln polymorphisms and lung cancer risk (OR=1.00, 95%CI: 0.92-1.08 and OR=1.05, 95%CI: 0.97- 1.13, respectively). Limiting the analysis to studies with controls in agreement with Hardy-Weinberg equilibrium (HWE), we observed an increased lung cancer risk among subjects carrying XRCC1 codon 399 Gln/Gln genotype (OR=1.18, 95%CI: 1.01-1.38). When stratified by source of control, we observed an increased lung cancer risk among subjects carrying XRCC1 codon 399 Arg/Gln+Gln/Gln genotype on the basis of hospitalized patient-based controls (OR=1.21, 95%CI: 1.04-1.42) and among subjects carrying XRCC1 codon 399 Gln/Gln genotype on the basis of healthy subject-based controls (OR=1.22, 95%CI: 1.04-1.43). CONCLUSIONS Our findings indicated that certain XRCC1 Arg399Gln variants might affect the susceptibility of lung cancer in Chinese population. Larger sample size studies are required to confirm our findings.
Collapse
Affiliation(s)
- Hai-Yan Yang
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou, China E-mail :
| | | | | | | | | |
Collapse
|
22
|
Bigert C, Gustavsson P, Straif K, Pesch B, Brüning T, Kendzia B, Schüz J, Stücker I, Guida F, Brüske I, Wichmann HE, Pesatori AC, Landi MT, Caporaso N, Tse LA, Yu ITS, Siemiatycki J, Pintos J, Merletti F, Mirabelli D, Simonato L, Jöckel KH, Ahrens W, Pohlabeln H, Tardón A, Zaridze D, Field J, 't Mannetje A, Pearce N, McLaughlin J, Demers P, Szeszenia-Dabrowska N, Lissowska J, Rudnai P, Fabianova E, Dumitru RS, Bencko V, Foretova L, Janout V, Boffetta P, Forastiere F, Bueno-de-Mesquita B, Peters S, Vermeulen R, Kromhout H, Olsson AC. Lung cancer risk among cooks when accounting for tobacco smoking: a pooled analysis of case-control studies from Europe, Canada, New Zealand, and China. J Occup Environ Med 2015; 57:202-9. [PMID: 25654522 PMCID: PMC7508228 DOI: 10.1097/jom.0000000000000337] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
OBJECTIVES To investigate the risk of lung cancer among cooks, while controlling for smoking habits. METHODS We used data from the SYNERGY project including pooled information on lifetime work histories and smoking habits from 16 case-control studies conducted in Europe, Canada, New Zealand, and China. RESULTS Before adjustment for smoking, we observed an increased risk of lung cancer in male cooks, but not in female cooks. After adjusting, there was no increased risk and no significant exposure-response relationship. Nevertheless, subgroup analyses highlighted some possible excess risks of squamous cell carcinoma and small cell carcinoma in female cooks. CONCLUSIONS There is evidence that lung cancer risks among cooks may be confounded by smoking. After adjustment, cooks did not experience an increased risk of lung cancer overall. The subgroup analyses showing some excess risks among female cooks require cautious interpretation.
Collapse
Affiliation(s)
- Carolina Bigert
- From the Institute of Environmental Medicine (Drs Bigert and Gustavsson), Karolinska Institutet, Stockholm, Sweden; International Agency for Research on Cancer (Drs Straif, Schüz, and Olsson), Lyon, France; Institute for Prevention and Occupational Medicine of the German Social Accident Insurance-Institute of the Ruhr-Universität Bochum (IPA) (Drs Pesch and Brüning, Mr Kendzia), Germany; Inserm, Centre for Research in Epidemiology and Population Health (CESP) (Drs Stücker and Guida), U1018, Environmental Epidemiology of Cancer Team, F-94807, Villejuif, France; Université Paris-Sud (Drs Stücker and Guida), UMRS 1018, F-94807, Villejuif, France; Institut für Epidemiologie I (Drs Brüske and Wichmann), Deutsches Forschungszentrum für Gesundheit und Umwelt, Neuherberg, Germany; Department of Clinical Sciences and Community Health (Dr Pesatori), Università degli Studi di Milano, Milan, Italy; National Cancer Institute (Drs Landi and Caporaso), Bethesda, MD; Division of Occupational and Environmental Health (Drs Tse and Yu), School of Public Health and Primary Care, The Chinese University of Hong Kong, China; Research Centre of University of Montréal Hospital Centre (Drs Siemiatycki and Pintos), University of Montréal, Canada; Cancer Epidemiology Unit (Drs Merletti and Mirabelli), Department of Medical Sciences, University of Turin, Italy; Department of Environmental Medicine and Public Health (Dr Simonato), University of Padua, Italy; Institute for Medical Informatics (Dr Jöckel), Biometry and Epidemiology, University of Duisburg-Essen, Essen, Germany; Bremen Institute for Prevention Research and Social Medicine (Drs Ahrens and Pohlabeln), Bremen, Germany; CIBERESP (Dr Tardón), University of Oviedo, Spain; Russian Cancer Research Centre (Dr Zaridze), Moscow, Russia; Roy Castle Lung Cancer Research Programme, Cancer Research Centre (Dr Field), University of Liverpool, UK; Centre for Public Health Research (Drs Mannetje and Pearce), Massey University, Wellingt
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Family L, Bensen JT, Troester MA, Wu MC, Anders CK, Olshan AF. Single-nucleotide polymorphisms in DNA bypass polymerase genes and association with breast cancer and breast cancer subtypes among African Americans and Whites. Breast Cancer Res Treat 2015; 149:181-90. [PMID: 25417172 PMCID: PMC4498665 DOI: 10.1007/s10549-014-3203-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 11/09/2014] [Indexed: 01/18/2023]
Abstract
DNA damage recognition and repair is a complex system of genes focused on maintaining genomic stability. Recently, there has been a focus on how breast cancer susceptibility relates to genetic variation in the DNA bypass polymerases pathway. Race-stratified and subtype-specific logistic regression models were used to estimate odds ratios (ORs) and 95 % confidence intervals (CIs) for the association between 22 single-nucleotide polymorphisms (SNPs) in seven bypass polymerase genes and breast cancer risk in the Carolina Breast Cancer Study, a population-based, case-control study (1,972 cases and 1,776 controls). We used SNP-set kernel association test (SKAT) to evaluate the multi-gene, multi-locus (combined) SNP effects within bypass polymerase genes. We found similar ORs for breast cancer with three POLQ SNPs (rs487848 AG/AA vs. GG; OR = 1.31, 95 % CI 1.03-1.68 for Whites and OR = 1.22, 95 % CI 1.00-1.49 for African Americans), (rs532411 CT/TT vs. CC; OR = 1.31, 95 % CI 1.02-1.66 for Whites and OR = 1.22, 95 % CI 1.00-1.48 for African Americans), and (rs3218634 CG/CC vs. GG; OR = 1.29, 95 % CI 1.02-1.65 for Whites). These three SNPs are in high linkage disequilibrium in both races. Tumor subtype analysis showed the same SNPs to be associated with increased risk of Luminal breast cancer. SKAT analysis showed no significant combined SNP effects. These results suggest that variants in the POLQ gene may be associated with the risk of Luminal breast cancer.
Collapse
Affiliation(s)
- Leila Family
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA,
| | | | | | | | | | | |
Collapse
|
24
|
Du Y, He Y, Mei Z, Qian L, Shi J, Jie Z. Association between genetic polymorphisms inXPDandXRCC1genes and risks of non-small cell lung cancer in East Chinese Han population. CLINICAL RESPIRATORY JOURNAL 2014; 10:311-7. [PMID: 25308691 DOI: 10.1111/crj.12218] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 08/09/2014] [Accepted: 09/29/2014] [Indexed: 12/01/2022]
Affiliation(s)
- Yong Du
- Department of Respiratory Medicine; The Fifth People's Hospital of Shanghai; Fudan University; Shanghai China
| | - Yanchao He
- Department of Respiratory Medicine; The Fifth People's Hospital of Shanghai; Fudan University; Shanghai China
| | - Zhoufang Mei
- Department of Respiratory Medicine; The Fifth People's Hospital of Shanghai; Fudan University; Shanghai China
| | - Ling Qian
- Department of Respiratory Medicine; The Fifth People's Hospital of Shanghai; Fudan University; Shanghai China
| | - Jindong Shi
- Department of Respiratory Medicine; The Fifth People's Hospital of Shanghai; Fudan University; Shanghai China
| | - Zhijun Jie
- Department of Respiratory Medicine; The Fifth People's Hospital of Shanghai; Fudan University; Shanghai China
| |
Collapse
|
25
|
Liu Y, Zhang Z, Song T, Liang F, Xie M, Sheng H. Resistance to BH3 mimetic S1 in SCLC cells that up-regulate and phosphorylate Bcl-2 through ERK1/2. Br J Pharmacol 2014; 169:1612-23. [PMID: 23651505 DOI: 10.1111/bph.12243] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 04/09/2013] [Accepted: 04/19/2013] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE B cell lymphoma 2 (Bcl-2) is a central regulator of cell survival that is overexpressed in the majority of small-cell lung cancers (SCLC) and contributes to both malignant transformation and therapeutic resistance. The purpose of this work was to study the key factors that determine the sensitivity of SCLC cells to Bcl-2 homology domain-3 (BH3) mimetic S1 and the mechanism underlying the resistance of BH3 mimetics. EXPERIMENTAL APPROACHES Western blot was used to evaluate the contribution of Bcl-2 family members to the cellular response of SCLC cell lines to S1. Acquired resistant cells were derived from initially sensitive H1688 cells. Quantitative PCR and gene silencing were performed to investigate Bcl-2 up-regulation. KEY RESULTS A progressive increase in the relative levels of Bcl-2 and phosphorylated Bcl-2 (pBcl-2) characterized the increased de novo and acquired resistance of SCLC cell lines. Furthermore, acute treatment of S1 induced Bcl-2 expression and phosphorylation. We showed that BH3 mimetics, including S1 and ABT-737, induced endoplasmic reticulum (ER) stress and then activated MAPK/ERK pathway. The dual function of MAPK/ERK pathway in defining BH3 mimetics was illustrated; ERK1/2 activation leaded to Bcl-2 transcriptional up-regulation and sustained phosphorylation in naïve and acquired resistant SCLC cells. pBcl-2 played a key role in creating resistance of S1 and ABT-737 not only by sequestrating pro-apoptotic proteins, but also sequestrating a positive feedback to promote ERK1/2 activation. CONCLUSIONS AND IMPLICATIONS These results provide significant novel insights into the molecular mechanisms for crosstalk between ER stress and endogenously apoptotic pathways in SCLC following BH3 mimetics treatment.
Collapse
Affiliation(s)
- Yubo Liu
- State Key Laboratory of Fine Chemicals, School of Chemistry, Dalian University of Technology, Dalian, China
| | | | | | | | | | | |
Collapse
|
26
|
Feigelson HS, Goddard KAB, Hollombe C, Tingle SR, Gillanders EM, Mechanic LE, Nelson SA. Approaches to integrating germline and tumor genomic data in cancer research. Carcinogenesis 2014; 35:2157-63. [PMID: 25115441 DOI: 10.1093/carcin/bgu165] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Cancer is characterized by a diversity of genetic and epigenetic alterations occurring in both the germline and somatic (tumor) genomes. Hundreds of germline variants associated with cancer risk have been identified, and large amounts of data identifying mutations in the tumor genome that participate in tumorigenesis have been generated. Increasingly, these two genomes are being explored jointly to better understand how cancer risk alleles contribute to carcinogenesis and whether they influence development of specific tumor types or mutation profiles. To understand how data from germline risk studies and tumor genome profiling is being integrated, we reviewed 160 articles describing research that incorporated data from both genomes, published between January 2009 and December 2012, and summarized the current state of the field. We identified three principle types of research questions being addressed using these data: (i) use of tumor data to determine the putative function of germline risk variants; (ii) identification and analysis of relationships between host genetic background and particular tumor mutations or types; and (iii) use of tumor molecular profiling data to reduce genetic heterogeneity or refine phenotypes for germline association studies. We also found descriptive studies that compared germline and tumor genomic variation in a gene or gene family, and papers describing research methods, data sources, or analytical tools. We identified a large set of tools and data resources that can be used to analyze and integrate data from both genomes. Finally, we discuss opportunities and challenges for cancer research that integrates germline and tumor genomics data.
Collapse
Affiliation(s)
- Heather Spencer Feigelson
- Institute for Health Research, Kaiser Permanente Colorado, Denver, CO, USA, Center for Health Research, Kaiser Permanente Northwest, Portland, OR, USA and Epidemiology and Genomics Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD, USA
| | - Katrina A B Goddard
- Center for Health Research, Kaiser Permanente Northwest, Portland, OR, USA and
| | - Celine Hollombe
- Center for Health Research, Kaiser Permanente Northwest, Portland, OR, USA and
| | - Sharna R Tingle
- Epidemiology and Genomics Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD, USA
| | - Elizabeth M Gillanders
- Epidemiology and Genomics Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD, USA
| | - Leah E Mechanic
- Epidemiology and Genomics Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD, USA
| | - Stefanie A Nelson
- Epidemiology and Genomics Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD, USA
| |
Collapse
|
27
|
Babron MC, Kazma R, Gaborieau V, McKay J, Brennan P, Sarasin A, Benhamou S. Genetic variants in DNA repair pathways and risk of upper aerodigestive tract cancers: combined analysis of data from two genome-wide association studies in European populations. Carcinogenesis 2014; 35:1523-7. [PMID: 24658182 DOI: 10.1093/carcin/bgu075] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
DNA repair pathways are good candidates for upper aerodigestive tract cancer susceptibility because of their critical role in maintaining genome integrity. We have selected 13 pathways involved in DNA repair representing 212 autosomal genes. To assess the role of these pathways and their associated genes, two European data sets from the International Head and Neck Cancer Epidemiology consortium were pooled, totaling 1954 cases and 3121 controls, with documented demographic, lifetime alcohol and tobacco consumption information. We applied an innovative approach that tests single nucleotide polymorphism (SNP)-sets within DNA repair pathways and then within genes belonging to the significant pathways. We showed an association between the polymerase pathway and oral cavity/pharynx cancers (P-corrected = 4.45 × 10(-) (2)), explained entirely by the association with one SNP, rs1494961 (P = 2.65 × 10(-) (4)), a missense mutation V306I in the second exon of HELQ gene. We also found an association between the cell cycle regulation pathway and esophagus cancer (P-corrected = 1.48 × 10(-) (2)), explained by three SNPs located within or near CSNK1E gene: rs1534891 (P = 1.27 × 10(-) (4)), rs7289981 (P = 3.37 × 10(-) (3)) and rs13054361 (P = 4.09 × 10(-) (3)). As a first attempt to investigate pathway-level associations, our results suggest a role of specific DNA repair genes/pathways in specific upper aerodigestive tract cancer sites.
Collapse
Affiliation(s)
- Marie-Claude Babron
- Inserm, U946, Genetic Variation and Human, Diseases and Université Paris-Diderot, Sorbonne Paris-Cité, UMRS-946, Paris, F-75010, France
| | - Rémi Kazma
- Department of Epidemiology and Biostatistics, Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| | - Valérie Gaborieau
- Department of Genetic Epidemiology, International Agency for Research on Cancer, Lyon, F-69008, France
| | - James McKay
- Department of Genetic Epidemiology, International Agency for Research on Cancer, Lyon, F-69008, France
| | - Paul Brennan
- Department of Genetic Epidemiology, International Agency for Research on Cancer, Lyon, F-69008, France
| | - Alain Sarasin
- Université Paris-Sud, Faculty of Medicine, Villejuif, F-94805, France, CNRS, UMR8200, Genomes and Cancers and Gustave Roussy, Villejuif, F-94805, France
| | - Simone Benhamou
- Inserm, U946, Genetic Variation and Human, Diseases and Université Paris-Diderot, Sorbonne Paris-Cité, UMRS-946, Paris, F-75010, France, Gustave Roussy, Villejuif, F-94805, France
| |
Collapse
|
28
|
Boja ES, Rodriguez H. Proteogenomic convergence for understanding cancer pathways and networks. Clin Proteomics 2014; 11:22. [PMID: 24994965 PMCID: PMC4067069 DOI: 10.1186/1559-0275-11-22] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 03/31/2014] [Indexed: 11/21/2022] Open
Abstract
During the past several decades, the understanding of cancer at the molecular level has been primarily focused on mechanisms on how signaling molecules transform homeostatically balanced cells into malignant ones within an individual pathway. However, it is becoming more apparent that pathways are dynamic and crosstalk at different control points of the signaling cascades, making the traditional linear signaling models inadequate to interpret complex biological systems. Recent technological advances in high throughput, deep sequencing for the human genomes and proteomic technologies to comprehensively characterize the human proteomes in conjunction with multiplexed targeted proteomic assays to measure panels of proteins involved in biologically relevant pathways have made significant progress in understanding cancer at the molecular level. It is undeniable that proteomic profiling of differentially expressed proteins under many perturbation conditions, or between normal and "diseased" states is important to capture a first glance at the overall proteomic landscape, which has been a main focus of proteomics research during the past 15-20 years. However, the research community is gradually shifting its heavy focus from that initial discovery step to protein target verification using multiplexed quantitative proteomic assays, capable of measuring changes in proteins and their interacting partners, isoforms, and post-translational modifications (PTMs) in response to stimuli in the context of signaling pathways and protein networks. With a critical link to genotypes (i.e., high throughput genomics and transcriptomics data), new and complementary information can be gleaned from multi-dimensional omics data to (1) assess the effect of genomic and transcriptomic aberrations on such complex molecular machinery in the context of cell signaling architectures associated with pathological diseases such as cancer (i.e., from genotype to proteotype to phenotype); and (2) target pathway- and network-driven changes and map the fluctuations of these functional units (proteins) responsible for cellular activities in response to perturbation in a spatiotemporal fashion to better understand cancer biology as a whole system.
Collapse
Affiliation(s)
- Emily S Boja
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, National Institutes of Health, 31 Center Drive, MSC 2580, 20892 Bethesda, MD, USA
| | - Henry Rodriguez
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, National Institutes of Health, 31 Center Drive, MSC 2580, 20892 Bethesda, MD, USA
| |
Collapse
|
29
|
Zhu J, Hua RX, Jiang J, Zhao LQ, Sun X, Luan J, Lang Y, Sun Y, Shang K, Peng S, Ma J. Association studies of ERCC1 polymorphisms with lung cancer susceptibility: a systematic review and meta-analysis. PLoS One 2014; 9:e97616. [PMID: 24841208 PMCID: PMC4026486 DOI: 10.1371/journal.pone.0097616] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 04/21/2014] [Indexed: 12/29/2022] Open
Abstract
Background Excision repair cross-complimentary group 1 (ERCC1) is an essential component of the nucleotide excision repair system that is responsible for repairing damaged DNA. Functional genetic variations in the ERCC1 gene may alter DNA repair capacity and modulate cancer risk. The putative roles of ERCC1 gene polymorphisms in lung cancer susceptibility have been widely investigated. However, the results remain controversial. Objectives An updated meta-analysis was conducted to explore whether lung cancer risk could be attributed to the following ERCC1 polymorphisms: rs11615 (T>C), rs3212986 (C>A), rs3212961 (A>C), rs3212948 (G>C), rs2298881 (C>A). Methods Several major databases (MEDLINE, EMBASE and Scopus) and the Chinese Biomedical database were searched for eligible studies. Crude odds ratios (ORs) with 95% confidence intervals (CIs) were used to measure the strength of associations. Results Sixteen studies with 10,106 cases and 13,238 controls were included in this meta-analysis. Pooled ORs from 11 eligible studies (8,215 cases vs. 11,402 controls) suggested a significant association of ERCC1 rs11615 with increased risk for lung cancer (homozygous: CC versus TT, OR = 1.24, 95% CI: 1.04–1.48, P = 0.02). However, such an association was disproportionately driven by a single study. Removal of that study led to null association. Moreover, initial analyses suggested that ERCC1 rs11615 exerts a more profound effect on the susceptibility of non-smokers to lung cancer than that of smokers. Moreover, no statistically significant association was found between remaining ERCC1 polymorphisms of interest and lung cancer risk, except for rs3212948 variation (heterozygous: CG vs.GG, OR = 0.78, 95% CI: 0.67–0.90, P = 0.001; dominant: CG/CC vs.GG, OR = 0.79, 95% CI: 0.69–0.91, P = 0.001). Conclusion Overall, this meta-analysis suggests that ERCC1 rs3212948 G>C, but not others, is a lung cancer risk-associated polymorphism. Carefully designed studies with large sample size involving different ethnicity, smoking status, and cancer types are needed to validate these findings.
Collapse
Affiliation(s)
- Jinhong Zhu
- Molecular Epidemiology Laboratory, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
- Department of Laboratory Medicine, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Rui-Xi Hua
- Department of Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jing Jiang
- Department of Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Li-Qin Zhao
- Department of Hematology, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Xiuwei Sun
- Department of Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Jinwei Luan
- Department of Radiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Yaoguo Lang
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Yanqi Sun
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Kun Shang
- Department of Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Shiyun Peng
- Department of Laboratory Medicine, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
- * E-mail: (JM); (SP)
| | - Jianqun Ma
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
- * E-mail: (JM); (SP)
| |
Collapse
|
30
|
Lim U, Kocarnik JM, Bush WS, Matise TC, Caberto C, Park SL, Carlson CS, Deelman E, Duggan D, Fesinmeyer M, Haiman CA, Henderson BE, Hindorff LA, Kolonel LN, Peters U, Stram DO, Tiirikainen M, Wilkens LR, Wu C, Kooperberg C, Le Marchand L. Pleiotropy of cancer susceptibility variants on the risk of non-Hodgkin lymphoma: the PAGE consortium. PLoS One 2014; 9:e89791. [PMID: 24598796 PMCID: PMC3943855 DOI: 10.1371/journal.pone.0089791] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2013] [Accepted: 01/27/2014] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Risk of non-Hodgkin lymphoma (NHL) is higher among individuals with a family history or a prior diagnosis of other cancers. Genome-wide association studies (GWAS) have suggested that some genetic susceptibility variants are associated with multiple complex traits (pleiotropy). OBJECTIVE We investigated whether common risk variants identified in cancer GWAS may also increase the risk of developing NHL as the first primary cancer. METHODS As part of the Population Architecture using Genomics and Epidemiology (PAGE) consortium, 113 cancer risk variants were analyzed in 1,441 NHL cases and 24,183 controls from three studies (BioVU, Multiethnic Cohort Study, Women's Health Initiative) for their association with the risk of overall NHL and common subtypes [diffuse large B-cell lymphoma (DLBCL), follicular lymphoma (FL), chronic lymphocytic leukemia or small lymphocytic lymphoma (CLL/SLL)] using an additive genetic model adjusted for age, sex and ethnicity. Study-specific results for each variant were meta-analyzed across studies. RESULTS The analysis of NHL subtype-specific GWAS SNPs and overall NHL suggested a shared genetic susceptibility between FL and DLBCL, particularly involving variants in the major histocompatibility complex region (rs6457327 in 6p21.33: FL OR=1.29, p=0.013; DLBCL OR=1.23, p=0.013; NHL OR=1.22, p=5.9 × E-05). In the pleiotropy analysis, six risk variants for other cancers were associated with NHL risk, including variants for lung (rs401681 in TERT: OR per C allele=0.89, p=3.7 × E-03; rs4975616 in TERT: OR per A allele=0.90, p=0.01; rs3131379 in MSH5: OR per T allele=1.16, p=0.03), prostate (rs7679673 in TET2: OR per C allele=0.89, p=5.7 × E-03; rs10993994 in MSMB: OR per T allele=1.09, p=0.04), and breast (rs3817198 in LSP1: OR per C allele=1.12, p=0.01) cancers, but none of these associations remained significant after multiple test correction. CONCLUSION This study does not support strong pleiotropic effects of non-NHL cancer risk variants in NHL etiology; however, larger studies are warranted.
Collapse
Affiliation(s)
- Unhee Lim
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, United States of America
| | - Jonathan M. Kocarnik
- Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - William S. Bush
- Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Tara C. Matise
- Department of Genetics, Rutgers University, Piscataway, New Jersey, United States of America
| | - Christian Caberto
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, United States of America
| | - Sungshim Lani Park
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Christopher S. Carlson
- Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Ewa Deelman
- Department of Genetics, Rutgers University, Piscataway, New Jersey, United States of America
| | - David Duggan
- Translational Genomics Research Institute, Phoenix, Arizona, United States of America
| | - Megan Fesinmeyer
- Center for Child Health, Behavior and Development, Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Christopher A. Haiman
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Brian E. Henderson
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Lucia A. Hindorff
- Division of Genomic Medicine, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Laurence N. Kolonel
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, United States of America
| | - Ulrike Peters
- Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Daniel O. Stram
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Maarit Tiirikainen
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, United States of America
| | - Lynne R. Wilkens
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, United States of America
| | - Chunyuan Wu
- Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Charles Kooperberg
- Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Loïc Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, United States of America
| |
Collapse
|
31
|
Li Y, Chang SC, Niu R, Liu L, Crabtree-Ide CR, Zhao B, Shi J, Han X, Li J, Su J, Cai L, Yu S, Zhang ZF, Mu L. TP53 genetic polymorphisms, interactions with lifestyle factors and lung cancer risk: a case control study in a Chinese population. BMC Cancer 2013; 13:607. [PMID: 24369748 PMCID: PMC3877976 DOI: 10.1186/1471-2407-13-607] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 12/18/2013] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND A pathway-based genotyping analysis suggested rs2078486 was a novel TP53 SNP, but very few studies replicate this association. TP53 rs1042522 is the most commonly studied SNP, but very few studies examined its potential interaction with environmental factors in relation to lung cancer risk. This study aims to examine associations between two TP53 single-nucleotide polymorphisms (SNPs) (rs2078486, rs1042522), their potential interaction with environmental factors and risk of lung cancer. METHODS A case-control study was conducted in Taiyuan, China. Unconditional logistic regression was used to estimate odds ratios (ORs) and 95% confidence intervals (95% CIs). Multiplicative and additive interactions between TP53 SNPs and lifestyle factors were evaluated. RESULTS Variant TP53 rs2078486 SNP was significantly associated with elevated lung cancer risk among smokers (OR: 1.70, 95% CI: 1.08 - 2.67) and individuals with high indoor air pollution exposure (OR: 1.51, 95% CI: 1.00-2.30). Significant or borderline significant multiplicative and additive interactions were found between TP53 rs2078486 polymorphism with smoking and indoor air pollution exposure. The variant genotype of TP53 SNP rs1042522 significantly increased lung cancer risk in the total population (OR: 1.57, 95% CI: 1.11-2.21), but there was no evidence of heterogeneity among individuals with different lifestyle factors. CONCLUSIONS This study confirmed that TP53 rs2078486 SNP is potentially a novel TP53 SNP that may affect lung cancer risk. Our study also suggested potential synergetic effects of TP53 rs2078486 SNP with smoking and indoor air pollution exposure on lung cancer risk.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Lina Mu
- Department of Social and Preventive Medicine, School of Public Health and Health Professions, The State University of New York (SUNY) at Buffalo, 273A Farber Hall, Buffalo, New York 14214-8001, USA.
| |
Collapse
|
32
|
Kendzia B, Behrens T, Jöckel KH, Siemiatycki J, Kromhout H, Vermeulen R, Peters S, Van Gelder R, Olsson A, Brüske I, Wichmann HE, Stücker I, Guida F, Tardón A, Merletti F, Mirabelli D, Richiardi L, Pohlabeln H, Ahrens W, Landi MT, Caporaso N, Consonni D, Zaridze D, Szeszenia-Dabrowska N, Lissowska J, Gustavsson P, Marcus M, Fabianova E, 't Mannetje A, Pearce N, Tse LA, Yu ITS, Rudnai P, Bencko V, Janout V, Mates D, Foretova L, Forastiere F, McLaughlin J, Demers P, Bueno-de-Mesquita B, Boffetta P, Schüz J, Straif K, Pesch B, Brüning T. Welding and lung cancer in a pooled analysis of case-control studies. Am J Epidemiol 2013; 178:1513-25. [PMID: 24052544 DOI: 10.1093/aje/kwt201] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Several epidemiologic studies have indicated an increased risk of lung cancer among welders. We used the SYNERGY project database to assess welding as a risk factor for developing lung cancer. The database includes data on 15,483 male lung cancer cases and 18,388 male controls from 16 studies in Europe, Canada, China, and New Zealand conducted between 1985 and 2010. Odds ratios and 95% confidence intervals between regular or occasional welding and lung cancer were estimated, with adjustment for smoking, age, study center, and employment in other occupations associated with lung cancer risk. Overall, 568 cases and 427 controls had ever worked as welders and had an odds ratio of developing lung cancer of 1.44 (95% confidence interval: 1.25, 1.67) with the odds ratio increasing for longer duration of welding. In never and light smokers, the odds ratio was 1.96 (95% confidence interval: 1.37, 2.79). The odds ratios were somewhat higher for squamous and small cell lung cancers than for adenocarcinoma. Another 1,994 cases and 1,930 controls had ever worked in occupations with occasional welding. Work in any of these occupations was associated with some elevation of risk, though not as much as observed in regular welders. Our findings lend further support to the hypothesis that welding is associated with an increased risk of lung cancer.
Collapse
|
33
|
Olsson AC, Xu Y, Schüz J, Vlaanderen J, Kromhout H, Vermeulen R, Peters S, Stücker I, Guida F, Brüske I, Wichmann HE, Consonni D, Landi MT, Caporaso N, Tse LA, Yu ITS, Siemiatycki J, Richardson L, Mirabelli D, Richiardi L, Simonato L, Gustavsson P, Plato N, Jöckel KH, Ahrens W, Pohlabeln H, Tardón A, Zaridze D, Marcus MW, 't Mannetje A, Pearce N, McLaughlin J, Demers P, Szeszenia-Dabrowska N, Lissowska J, Rudnai P, Fabianova E, Dumitru RS, Bencko V, Foretova L, Janout V, Boffetta P, Fortes C, Bueno-de-Mesquita B, Kendzia B, Behrens T, Pesch B, Brüning T, Straif K. Lung cancer risk among hairdressers: a pooled analysis of case-control studies conducted between 1985 and 2010. Am J Epidemiol 2013; 178:1355-65. [PMID: 24068200 DOI: 10.1093/aje/kwt119] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Increased lung cancer risks among hairdressers were observed in large registry-based cohort studies from Scandinavia, but these studies could not adjust for smoking. Our objective was to evaluate the lung cancer risk among hairdressers while adjusting for smoking and other confounders in a pooled database of 16 case-control studies conducted in Europe, Canada, China, and New Zealand between 1985 and 2010 (the Pooled Analysis of Case-Control Studies on the Joint Effects of Occupational Carcinogens in the Development of Lung Cancer). Lifetime occupational and smoking information was collected through interviews with 19,369 cases of lung cancer and 23,674 matched population or hospital controls. Overall, 170 cases and 167 controls had ever worked as hairdresser or barber. The odds ratios for lung cancer in women were 1.65 (95% confidence interval (CI): 1.16, 2.35) without adjustment for smoking and 1.12 (95% CI: 0.75, 1.68) with adjustment for smoking; however, women employed before 1954 also experienced an increased lung cancer risk after adjustment for smoking (odds ratio = 2.66, 95% CI: 1.09, 6.47). The odds ratios in male hairdressers/barbers were generally not elevated, except for an increased odds ratio for adenocarcinoma in long-term barbers (odds ratio = 2.20, 95% CI: 1.02, 4.77). Our results suggest that the increased lung cancer risks among hairdressers are due to their smoking behavior; single elevated risk estimates should be interpreted with caution and need replication in other studies.
Collapse
|
34
|
Ouyang FD, Yang FL, Chen HC, Khan MA, Huang FM, Wan XX, Xu AH, Huang X, Zhou MJ, Fang Q, Zhang DZ. Polymorphisms of DNA repair genes XPD, XRCC1, and OGG1, and lung adenocarcinoma susceptibility in Chinese population. Tumour Biol 2013; 34:2843-2848. [PMID: 23700156 DOI: 10.1007/s13277-013-0844-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 05/03/2013] [Indexed: 10/26/2022] Open
Abstract
Lung adenocarcinoma (ADC) is one of the major histological types of lung cancer. Genetic polymorphism in DNA repair genes and lung ADC susceptibility is well documented. In this case-control study, the association between the polymorphic sites of DNA repair genes XPD-751, XRCC1-399, and OGG1-326, and lung ADC susceptibility in ethnic Han Chinese population has been investigated. Genomic DNA was isolated from the peripheral blood of 201 healthy controls and 82 lung ADC patients from the people of Hunan Province, China. Polymorphisms of the investigated genes were analyzed by using polymerase chain reaction-restriction fragment length polymorphism. There was no significant difference between the samples from lung ADC patients and healthy controls about the genotype frequencies of XPD-751, XRCC1-399, and OGG1-326 sites. However, multifactor dimensionality reduction analysis showed that the genetic polymorphisms of the three-loci models of DNA repair genes (XPD-751/XRCC1-399/OGG1-326) are associated with lung ADC. Thus, this study reveals that a three-order interaction among the polymorphic sites of XPD-751, XRCC1-399, and OGG1-326 is associated with lung ADC risk in the studied population, although polymorphism in individual gene was not associated.
Collapse
Affiliation(s)
- Fang-dan Ouyang
- Department of Biochemistry, School of Life Sciences, Central South University, Changsha, Hunan, 410013, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Clark N, Wu X, Her C. MutS Homologues hMSH4 and hMSH5: Genetic Variations, Functions, and Implications in Human Diseases. Curr Genomics 2013; 14:81-90. [PMID: 24082819 PMCID: PMC3637681 DOI: 10.2174/1389202911314020002] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Revised: 01/19/2013] [Accepted: 01/21/2013] [Indexed: 12/01/2022] Open
Abstract
The prominence of the human mismatch repair (MMR) pathway is clearly reflected by the causal link between MMR gene mutations and the occurrence of Lynch syndrome (or HNPCC). The MMR family of proteins also carries out a plethora of diverse cellular functions beyond its primary role in MMR and homologous recombination. In fact, members of the MMR family of proteins are being increasingly recognized as critical mediators between DNA damage repair and cell survival. Thus, a better functional understanding of MMR proteins will undoubtedly aid the development of strategies to effectively enhance apoptotic signaling in response to DNA damage induced by anti-cancer therapeutics. Among the five known human MutS homologs, hMSH4 and hMSH5 form a unique heterocomplex. However, the expression profiles of the two genes are not correlated in a number of cell types, suggesting that they may function independently as well. Consistent with this, these two proteins are promiscuous and thought to play distinct roles through interacting with different binding partners. Here, we describe the gene and protein structures of eukaryotic MSH4 and MSH5 with a particular emphasis on their human homologues, and we discuss recent findings of the roles of these two genes in DNA damage response and repair. Finally, we delineate the potential links of single nucleotide polymorphism (SNP) loci of these two genes with several human diseases.
Collapse
Affiliation(s)
- Nicole Clark
- STARS Program, College of Veterinary Medicine, Washington State University, Pullman, WA 99164-7520, USA ; School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99164-7520, USA
| | | | | |
Collapse
|
36
|
Doherty JA, Sakoda LC, Loomis MM, Barnett MJ, Julianto L, Thornquist MD, Neuhouser ML, Weiss NS, Goodman GE, Chen C. DNA repair genotype and lung cancer risk in the beta-carotene and retinol efficacy trial. INTERNATIONAL JOURNAL OF MOLECULAR EPIDEMIOLOGY AND GENETICS 2013; 4:11-34. [PMID: 23565320 PMCID: PMC3612452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Accepted: 02/28/2013] [Indexed: 06/02/2023]
Abstract
Many carcinogens in tobacco smoke cause DNA damage, and some of that damage can be mitigated by the actions of DNA repair enzymes. In a case-control study nested within the Beta-Carotene and Retinol Efficacy Trial, a randomized chemoprevention trial in current and former heavy smokers, we examined whether lung cancer risk was associated with variation in 26 base excision repair, mismatch repair, and homologous recombination repair genes. Analyses were limited to Caucasians (744 cases, 1477 controls), and logistic regression was used to calculate odds ratios (ORs) and 95% confidence intervals (CIs) for individual SNPs and common haplotypes, with adjustment for matching factors. Lung cancer associations were observed (p<0.05) with SNPs in MSH5 (rs3131379, rs707938), MSH2 (rs2303428), UNG (rs246079), and PCNA (rs25406). MSH5 rs3131379 is a documented lung cancer susceptibility locus in complete linkage disequilibrium with rs3117582 in BAT3, and we observed associations similar in magnitude to those in prior studies (per A allele OR 1.37, 95% CI 1.13-1.65). UNG was associated with lung cancer risk at the gene level (p=0.02), and the A allele of rs246079 was associated with an increased risk (per A allele OR 1.15, 95% CI1.01-1.31). We observed stronger associations with UNG rs246079 among individuals who carried the risk genotypes (AG/AA) for MSH5 rs3131379 (pinteraction= 0.038). Our results provide additional evidence to suggest that the MSH5/BAT3 locus is associated with increased lung cancer risk among smokers, and that associations with other SNPs may vary depending upon MSH5/BAT3 genotype. Future studies to examine this possibility are warranted.
Collapse
Affiliation(s)
- Jennifer A Doherty
- The Geisel School of Medicine at DartmouthLebanon, NH, USA
- Division of Public Health Sciences, Fred Hutchinson Cancer Research CenterSeattle, WA, USA
| | - Lori C Sakoda
- Division of Public Health Sciences, Fred Hutchinson Cancer Research CenterSeattle, WA, USA
- Kaiser Permanente Northern California, Division of ResearchOakland, CA, USA
| | - Melissa M Loomis
- Division of Public Health Sciences, Fred Hutchinson Cancer Research CenterSeattle, WA, USA
| | - Matt J Barnett
- Division of Public Health Sciences, Fred Hutchinson Cancer Research CenterSeattle, WA, USA
| | - Liberto Julianto
- Division of Public Health Sciences, Fred Hutchinson Cancer Research CenterSeattle, WA, USA
| | - Mark D Thornquist
- Division of Public Health Sciences, Fred Hutchinson Cancer Research CenterSeattle, WA, USA
| | - Marian L Neuhouser
- Division of Public Health Sciences, Fred Hutchinson Cancer Research CenterSeattle, WA, USA
| | - Noel S Weiss
- Division of Public Health Sciences, Fred Hutchinson Cancer Research CenterSeattle, WA, USA
- Department of Epidemiology, School of Public Health, University of WashingtonSeattle, WA, USA
| | - Gary E Goodman
- Division of Public Health Sciences, Fred Hutchinson Cancer Research CenterSeattle, WA, USA
| | - Chu Chen
- Division of Public Health Sciences, Fred Hutchinson Cancer Research CenterSeattle, WA, USA
- Department of Epidemiology, School of Public Health, University of WashingtonSeattle, WA, USA
- Department of Otolaryngology: Head and Neck Surgery, School of Medicine, University of WashingtonSeattle, WA, USA
| |
Collapse
|