1
|
Fu S, Wheeler W, Wang X, Hua X, Godbole D, Duan J, Zhu B, Deng L, Qin F, Zhang H, Shi J, Yu K. A comprehensive framework for trans-ancestry pathway analysis using GWAS summary data from diverse populations. PLoS Genet 2024; 20:e1011322. [PMID: 39441834 PMCID: PMC11534268 DOI: 10.1371/journal.pgen.1011322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 11/04/2024] [Accepted: 10/07/2024] [Indexed: 10/25/2024] Open
Abstract
As more multi-ancestry GWAS summary data become available, we have developed a comprehensive trans-ancestry pathway analysis framework that effectively utilizes this diverse genetic information. Within this framework, we evaluated various strategies for integrating genetic data at different levels-SNP, gene, and pathway-from multiple ancestry groups. Through extensive simulation studies, we have identified robust strategies that demonstrate superior performance across diverse scenarios. Applying these methods, we analyzed 6,970 pathways for their association with schizophrenia, incorporating data from African, East Asian, and European populations. Our analysis identified over 200 pathways significantly associated with schizophrenia, even after excluding genes near genome-wide significant loci. This approach substantially enhances detection efficiency compared to traditional single-ancestry pathway analysis and the conventional approach that amalgamates single-ancestry pathway analysis results across different ancestry groups. Our framework provides a flexible and effective tool for leveraging the expanding pool of multi-ancestry GWAS summary data, thereby improving our ability to identify biologically relevant pathways that contribute to disease susceptibility.
Collapse
Affiliation(s)
- Sheng Fu
- School of Statistics and Data Science, Nankai University, Tianjin, China
- Key Laboratory of Pure Mathematics and Combinatorics, Nankai University, Tianjin, China
| | - William Wheeler
- Information Management Services, Inc, Bethesda, Maryland, United States of America
| | - Xiaoyu Wang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, United States of America
- Cancer Genomics Research Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc, Rockville, Maryland, United States of America
| | - Xing Hua
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, United States of America
- Cancer Genomics Research Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc, Rockville, Maryland, United States of America
| | - Devika Godbole
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, United States of America
- Cancer Genomics Research Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc, Rockville, Maryland, United States of America
| | - Jubao Duan
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, Illinois, United States of America
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, Illinois, United States of America
| | - Bin Zhu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Lu Deng
- School of Statistics and Data Science, Nankai University, Tianjin, China
| | - Fei Qin
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Haoyu Zhang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Jianxin Shi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Kai Yu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, United States of America
| |
Collapse
|
2
|
Jackson SS, Lex M, De Wyngard VV, Cook P, Hildesheim A, Pinto LA, Jackson SH, Choi K, Minas TZ, Losada Morales HF, Araya JC, Ferreccio C, Koshiol J, Pfeiffer RM. Statin use is not associated with inflammation among Chilean women of Mapuche and non-Mapuche ancestry with gallstones. Future Sci OA 2024; 10:2340327. [PMID: 38817359 PMCID: PMC11137765 DOI: 10.2144/fsoa-2023-0032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 10/18/2023] [Indexed: 06/01/2024] Open
Abstract
Aim: Statins are associated with lower risk of gallstones due to anti-inflammatory effects. We assessed whether statins impact circulating inflammation among Chilean women with gallstones. Materials & methods: 200 Mapuche women were matched on statin use and age to 200 non-Mapuche women in the Chile Biliary Longitudinal Study. We analyzed 92 inflammatory biomarkers using multivariable-adjusted regression models, random forests and pathway analyses. Results: Statins were not significantly associated with any inflammation marker when women were analyzed jointly or stratified by ancestry. No significant associations were found through random forest methods and pathway analyses. Discussion: We did not find significant associations between statin use and inflammation markers in women with gallstones, suggesting that statins do not reduce inflammation once gallstones have formed.
Collapse
Affiliation(s)
- Sarah S Jackson
- Division of Cancer Epidemiology & Genetics, National Cancer Institute, Rockville, MD, USA
| | - Marina Lex
- Department of Mathematics, The Technical University of Munich, Munich, Germany
| | - Vanessa Van De Wyngard
- School of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), FONDAP, Santiago, Chile
| | - Paz Cook
- School of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), FONDAP, Santiago, Chile
| | - Allan Hildesheim
- Division of Cancer Epidemiology & Genetics, National Cancer Institute, Rockville, MD, USA
| | - Ligia A Pinto
- Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Sharon H Jackson
- Division of Intramural Research, National Institute on Minority Health & Health Disparities, Bethesda, MD, USA
| | - Kelvin Choi
- Division of Intramural Research, National Institute on Minority Health & Health Disparities, Bethesda, MD, USA
| | - Tsion Zewdu Minas
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Héctor Fabio Losada Morales
- Hepato-pancreatic & biliary surgery team, Surgery Department, Universidad de la Frontera, Temuco, Chile
- Hepato-pancreatic & biliary surgery team, Hospital Dr. Hernán Henriquez Aravena, Temuco, Chile
| | - Juan Carlos Araya
- Advanced Center for Chronic Diseases (ACCDiS), FONDAP, Santiago, Chile
- Laboratorio de Inmunopatología Traslacional, Facultad de Ciencias, Universidad Mayor, Chile
- Department of Pathology, Faculty of Medicine, Universidad de la Frontera, Temuco, Chile
- Center for Cancer Prevention and Control, CECAN (ANID 152220002), Santiago, Chile
| | - Catterina Ferreccio
- School of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), FONDAP, Santiago, Chile
| | - Jill Koshiol
- Division of Cancer Epidemiology & Genetics, National Cancer Institute, Rockville, MD, USA
| | - Ruth M Pfeiffer
- Division of Cancer Epidemiology & Genetics, National Cancer Institute, Rockville, MD, USA
| |
Collapse
|
3
|
Meng Z, Jiang Z. Cauchy combination omnibus test for normality. PLoS One 2023; 18:e0289498. [PMID: 37535617 PMCID: PMC10399863 DOI: 10.1371/journal.pone.0289498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 07/20/2023] [Indexed: 08/05/2023] Open
Abstract
Testing whether data are from a normal distribution is a traditional problem and is of great concern for data analyses. The normality is the premise of many statistical methods, such as t-test, Hotelling T2 test and ANOVA. There are numerous tests in the literature and the commonly used ones are Anderson-Darling test, Shapiro-Wilk test and Jarque-Bera test. Each test has its own advantageous points since they are developed for specific patterns and there is no method that consistently performs optimally in all situations. Since the data distribution of practical problems can be complex and diverse, we propose a Cauchy Combination Omnibus Test (CCOT) that is robust and valid in most data cases. We also give some theoretical results to analyze the good properties of CCOT. Two obvious advantages of CCOT are that not only does CCOT have a display expression for calculating statistical significance, but extensive simulation results show its robustness regardless of the shape of distribution the data comes from. Applications to South African Heart Disease and Neonatal Hearing Impairment data further illustrate its practicability.
Collapse
Affiliation(s)
- Zhen Meng
- School of Statistics, Capital University of Economics and Business, Beijing, China
| | - Zhenzhen Jiang
- Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing, China
- School of Mathematical Sciences, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
4
|
Argirion I, Pfeiffer RM, Proietti C, Coghill AE, Yu KJ, Middeldorp JM, Sarathkumara YD, Hsu WL, Chien YC, Lou PJ, Wang CP, Rothman N, Lan Q, Chen CJ, Mbulaiteye SM, Jarrett RF, Glimelius I, Smedby KE, Hjalgrim H, Hildesheim A, Doolan DL, Liu Z. Comparative Analysis of the Humoral Immune Response to the EBV Proteome across EBV-Related Malignancies. Cancer Epidemiol Biomarkers Prev 2023; 32:687-696. [PMID: 36788424 PMCID: PMC10159936 DOI: 10.1158/1055-9965.epi-22-0452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 07/14/2022] [Accepted: 02/13/2023] [Indexed: 02/16/2023] Open
Abstract
BACKGROUND Epstein-Barr virus (EBV) is linked to multiple cancers, including classical Hodgkin lymphoma (cHL), endemic Burkitt lymphoma (eBL), nasopharyngeal carcinoma (NPC), and extranodal natural killer/T-cell lymphoma (NKTCL). METHODS Anti-EBV IgG and IgA antibody responses targeting 202 sequences from 86 EBV proteins were measured using the same EBV whole proteome array across four case-control studies investigating EBV-positive cHL, eBL, NPC, and NKTCL (407 cases/620 controls). We grouped EBV-targeted antibodies into pathways by immunoglobulin type (IgA and IgG) and life-cycle stage (latent, immediate early lytic, early lytic, late lytic, and glycoprotein) and evaluated their association with each cancer type. In an additional analysis, we focused on the subset of 46 individual antibodies representing the top candidates for each cancer and compared their associations across the four cancer types using multivariable linear regression models. RESULTS IgA antibody responses targeting all EBV life-cycle stages were associated with NPC but limited to anti-early lytic stage for cHL. NPC and eBL were associated with IgG antibodies across the viral life cycle; cHL with antibodies in the early lytic, late lytic and glycoprotein stages; and NKTCL with antibodies in the latent, immediate early lytic and early lytic phases. EBNA3A, BBLF1, BDLF4, and BLRF2 IgG antibodies were associated with all cancer types. CONCLUSIONS Our observed similarities and differences across four EBV-associated cancers may inform EBV-related oncogenesis. IMPACT Understanding the comparative humoral immune response across EBV-related cancers may aid in identifying shared etiologic roles of EBV proteins and inform unique pathogenic processes for each cancer.
Collapse
Affiliation(s)
- Ilona Argirion
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Ruth M. Pfeiffer
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Carla Proietti
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health & Medicine, James Cook University, Cairns, QLD, Australia
| | - Anna E. Coghill
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
- Cancer Epidemiology Program, Division of Population Sciences, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Kelly J. Yu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | | | - Yomani D. Sarathkumara
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health & Medicine, James Cook University, Cairns, QLD, Australia
| | - Wan-Lun Hsu
- Master Program of Big Data in Biomedicine, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
- Data Science Center, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Yin-Chu Chien
- Genomics Research Center, Academica Sinica, Taipei, Taiwan
- National Institute of Cancer Research, National Health Research Institute, Miaoli, Taiwan
| | - Pei-Jen Lou
- Department of Otolaryngology, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Cheng-Ping Wang
- Department of Otolaryngology, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Nathaniel Rothman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Qing Lan
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Chien-Jen Chen
- Genomics Research Center, Academica Sinica, Taipei, Taiwan
- Graduate Institute of Epidemiology and Prevention Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Sam M. Mbulaiteye
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Ruth F. Jarrett
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Ingrid Glimelius
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Karin E. Smedby
- Department of Medicine Solna, Division of Clinical Epidemiology, Karolinska Institutet, Stockholm, Sweden
| | - Henrik Hjalgrim
- Statens Serum Institut, Copenhagen, Denmark
- Department of Haematology, Rigshospitalet, Copenhagen, Denmark
| | - Allan Hildesheim
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Denise L. Doolan
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health & Medicine, James Cook University, Cairns, QLD, Australia
| | - Zhiwei Liu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| |
Collapse
|
5
|
Defo J, Awany D, Ramesar R. From SNP to pathway-based GWAS meta-analysis: do current meta-analysis approaches resolve power and replication in genetic association studies? Brief Bioinform 2023; 24:6972298. [PMID: 36611240 DOI: 10.1093/bib/bbac600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/30/2022] [Accepted: 12/06/2022] [Indexed: 01/09/2023] Open
Abstract
Genome-wide association studies (GWAS) have benefited greatly from enhanced high-throughput technology in recent decades. GWAS meta-analysis has become increasingly popular to highlight the genetic architecture of complex traits, informing about the replicability and variability of effect estimations across human ancestries. A wealth of GWAS meta-analysis methodologies have been developed depending on the input data and the outcome information of interest. We present a survey of current approaches from SNP to pathway-based meta-analysis by acknowledging the range of resources and methodologies in the field, and we provide a comprehensive review of different categories of Genome-Wide Meta-analysis methods employed. These methods highlight different levels at which GWAS meta-analysis may be done, including Single Nucleotide Polymorphisms, Genes and Pathways, for which we describe their framework outline. We also discuss the strengths and pitfalls of each approach and make suggestions regarding each of them.
Collapse
Affiliation(s)
- Joel Defo
- Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, 7925, Observatory, South Africa.,South African Medical Research Council Genomic and Personalized Medicine Research Unit
| | - Denis Awany
- South African Tuberculosis Vaccine Initiative (SATVI), University of Cape Town, 7925, South Africa
| | - Raj Ramesar
- Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, 7925, Observatory, South Africa.,South African Medical Research Council Genomic and Personalized Medicine Research Unit
| |
Collapse
|
6
|
Lai J, Wang X, Zhao K, Zheng S. Block-diagonal test for high-dimensional covariance matrices. TEST-SPAIN 2022. [DOI: 10.1007/s11749-022-00842-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
7
|
Chen X, Zhang H, Liu M, Deng HW, Wu Z. Simultaneous detection of novel genes and SNPs by adaptive p-value combination. Front Genet 2022; 13:1009428. [DOI: 10.3389/fgene.2022.1009428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 11/03/2022] [Indexed: 11/18/2022] Open
Abstract
Combining SNP p-values from GWAS summary data is a promising strategy for detecting novel genetic factors. Existing statistical methods for the p-value-based SNP-set testing confront two challenges. First, the statistical power of different methods depends on unknown patterns of genetic effects that could drastically vary over different SNP sets. Second, they do not identify which SNPs primarily contribute to the global association of the whole set. We propose a new signal-adaptive analysis pipeline to address these challenges using the omnibus thresholding Fisher’s method (oTFisher). The oTFisher remains robustly powerful over various patterns of genetic effects. Its adaptive thresholding can be applied to estimate important SNPs contributing to the overall significance of the given SNP set. We develop efficient calculation algorithms to control the type I error rate, which accounts for the linkage disequilibrium among SNPs. Extensive simulations show that the oTFisher has robustly high power and provides a higher balanced accuracy in screening SNPs than the traditional Bonferroni and FDR procedures. We applied the oTFisher to study the genetic association of genes and haplotype blocks of the bone density-related traits using the summary data of the Genetic Factors for Osteoporosis Consortium. The oTFisher identified more novel and literature-reported genetic factors than existing p-value combination methods. Relevant computation has been implemented into the R package TFisher to support similar data analysis.
Collapse
|
8
|
Association between Immunologic Markers and Cirrhosis in Individuals from a Prospective Chronic Hepatitis C Cohort. Cancers (Basel) 2022; 14:cancers14215280. [PMID: 36358697 PMCID: PMC9657502 DOI: 10.3390/cancers14215280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/19/2022] [Accepted: 10/23/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Chronic hepatitis C virus (HCV) infection can affect immune response and inflammatory pathways, leading to severe liver diseases such as cirrhosis and hepatocellular carcinoma (HCC). Methods: In a prospective cohort of chronically HCV-infected individuals, we sampled 68 individuals who developed cirrhosis, 91 controls who did not develop cirrhosis, and 94 individuals who developed HCC. Unconditional odds ratios (ORs) from polytomous logistic regression models and canonical discriminant analyses (CDAs) were used to compare categorical (C) baseline plasma levels for 102 markers in individuals who developed cirrhosis vs. controls and those who developed HCC vs. cirrhosis. Leave-one-out cross validation was used to produce receiver operating characteristic curves to assess predictive ability of markers. Lastly, biological pathways were assessed in association with cirrhotic development compared to controls. Results: After multivariable adjustment, DEFA-1 (OR: C2v.C1 = 7.73; p < 0.0001), ITGAM (OR: C2v.C1 = 4.03; p = 0.0002), SCF (OR: C4v.C1 = 0.19; p-trend = 0.0001), and CCL11 (OR: C4v.C1 = 0.31; p-trend= 0.002) were all associated with development of cirrhosis compared to controls; these markers, together with clinical/demographics variables, improved prediction of cirrhosis from 55.7% (in clinical/demographic-only model) to 74.9% accuracy. A twelve-marker model based on CDA results further increased prediction of cirrhosis to 88.0%. While six biological pathways were found to be associated with cirrhosis, cell adhesion was the only pathway associated with cirrhosis after Bonferroni correction. In contrast to cirrhosis, DEFA-1 and ITGAM levels were inversely associated with HCC risk. Conclusions: Pending validation, these findings highlight the important role of immunological markers in predicting HCV-related cirrhosis even 11 years post-enrollment.
Collapse
|
9
|
Duroux D, Climente-González H, Azencott CA, Van Steen K. Interpretable network-guided epistasis detection. Gigascience 2022; 11:giab093. [PMID: 35134928 PMCID: PMC8848319 DOI: 10.1093/gigascience/giab093] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 10/12/2021] [Accepted: 12/13/2021] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Detecting epistatic interactions at the gene level is essential to understanding the biological mechanisms of complex diseases. Unfortunately, genome-wide interaction association studies involve many statistical challenges that make such detection hard. We propose a multi-step protocol for epistasis detection along the edges of a gene-gene co-function network. Such an approach reduces the number of tests performed and provides interpretable interactions while keeping type I error controlled. Yet, mapping gene interactions into testable single-nucleotide polymorphism (SNP)-interaction hypotheses, as well as computing gene pair association scores from SNP pair ones, is not trivial. RESULTS Here we compare 3 SNP-gene mappings (positional overlap, expression quantitative trait loci, and proximity in 3D structure) and use the adaptive truncated product method to compute gene pair scores. This method is non-parametric, does not require a known null distribution, and is fast to compute. We apply multiple variants of this protocol to a genome-wide association study dataset on inflammatory bowel disease. Different configurations produced different results, highlighting that various mechanisms are implicated in inflammatory bowel disease, while at the same time, results overlapped with known disease characteristics. Importantly, the proposed pipeline also differs from a conventional approach where no network is used, showing the potential for additional discoveries when prior biological knowledge is incorporated into epistasis detection.
Collapse
Affiliation(s)
- Diane Duroux
- BIO3 - Systems Genetics, GIGA-R Medical Genomics, University of Liège, 4000 Liège, Belgium, 11 Liège 4000, Belgium
| | - Héctor Climente-González
- Institut Curie, PSL Research University, F-75005 Paris, France
- INSERM, U900, F-75005 Paris, France
- CBIO-Centre for Computational Biology, Mines ParisTech, PSL Research University, 75006 Paris, France
- High-Dimensional Statistical Modeling Team, RIKEN Center for Advanced Intelligence Project, Chuo-ku, Tokyo 103-0027, Japan
| | - Chloé-Agathe Azencott
- Institut Curie, PSL Research University, F-75005 Paris, France
- INSERM, U900, F-75005 Paris, France
- CBIO-Centre for Computational Biology, Mines ParisTech, PSL Research University, 75006 Paris, France
| | - Kristel Van Steen
- BIO3 - Systems Genetics, GIGA-R Medical Genomics, University of Liège, 4000 Liège, Belgium, 11 Liège 4000, Belgium
- BIO3 - Systems Medicine, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium, 49 3000 Leuven, Belgium
| |
Collapse
|
10
|
Marczyk M, Macioszek A, Tobiasz J, Polanska J, Zyla J. Importance of SNP Dependency Correction and Association Integration for Gene Set Analysis in Genome-Wide Association Studies. Front Genet 2021; 12:767358. [PMID: 34956320 PMCID: PMC8696167 DOI: 10.3389/fgene.2021.767358] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/10/2021] [Indexed: 11/13/2022] Open
Abstract
A typical genome-wide association study (GWAS) analyzes millions of single-nucleotide polymorphisms (SNPs), several of which are in a region of the same gene. To conduct gene set analysis (GSA), information from SNPs needs to be unified at the gene level. A widely used practice is to use only the most relevant SNP per gene; however, there are other methods of integration that could be applied here. Also, the problem of nonrandom association of alleles at two or more loci is often neglected. Here, we tested the impact of incorporation of different integrations and linkage disequilibrium (LD) correction on the performance of several GSA methods. Matched normal and breast cancer samples from The Cancer Genome Atlas database were used to evaluate the performance of six GSA algorithms: Coincident Extreme Ranks in Numerical Observations (CERNO), Gene Set Enrichment Analysis (GSEA), GSEA-SNP, improved GSEA for GWAS (i-GSEA4GWAS), Meta-Analysis Gene-set Enrichment of variaNT Associations (MAGENTA), and Over-Representation Analysis (ORA). Association of SNPs to phenotype was calculated using modified McNemar's test. Results for SNPs mapped to the same gene were integrated using Fisher and Stouffer methods and compared with the minimum p-value method. Four common measures were used to quantify the performance of all combinations of methods. Results of GSA analysis on GWAS were compared to the one performed on gene expression data. Comparing all evaluation metrics across different GSA algorithms, integrations, and LD correction, we highlighted CERNO, and MAGENTA with Stouffer as the most efficient. Applying LD correction increased prioritization and specificity of enrichment outcomes for all tested algorithms. When Fisher or Stouffer were used with LD, sensitivity and reproducibility were also better. Using any integration method was beneficial in comparison with a minimum p-value method in specific combinations. The correlation between GSA results from genomic and transcriptomic level was the highest when Stouffer integration was combined with LD correction. We thoroughly evaluated different approaches to GSA in GWAS in terms of performance to guide others to select the most effective combinations. We showed that LD correction and Stouffer integration could increase the performance of enrichment analysis and encourage the usage of these techniques.
Collapse
Affiliation(s)
- Michal Marczyk
- Department of Data Science and Engineering, Silesian University of Technology, Gliwice, Poland.,Yale Cancer Center, Yale School of Medicine, New Haven, CT, United States
| | - Agnieszka Macioszek
- Department of Data Science and Engineering, Silesian University of Technology, Gliwice, Poland
| | - Joanna Tobiasz
- Department of Data Science and Engineering, Silesian University of Technology, Gliwice, Poland
| | - Joanna Polanska
- Department of Data Science and Engineering, Silesian University of Technology, Gliwice, Poland
| | - Joanna Zyla
- Department of Data Science and Engineering, Silesian University of Technology, Gliwice, Poland
| |
Collapse
|
11
|
Heard NA. Standardized Partial Sums and Products of p-Values. J Comput Graph Stat 2021. [DOI: 10.1080/10618600.2021.1999822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- N. A. Heard
- Department of Mathematics, Imperial College London, London, UK
| |
Collapse
|
12
|
|
13
|
Dutta D, VandeHaar P, Fritsche LG, Zöllner S, Boehnke M, Scott LJ, Lee S. A powerful subset-based method identifies gene set associations and improves interpretation in UK Biobank. Am J Hum Genet 2021; 108:669-681. [PMID: 33730541 DOI: 10.1016/j.ajhg.2021.02.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 02/19/2021] [Indexed: 02/06/2023] Open
Abstract
Tests of association between a phenotype and a set of genes in a biological pathway can provide insights into the genetic architecture of complex phenotypes beyond those obtained from single-variant or single-gene association analysis. However, most existing gene set tests have limited power to detect gene set-phenotype association when a small fraction of the genes are associated with the phenotype and cannot identify the potentially "active" genes that might drive a gene set-based association. To address these issues, we have developed Gene set analysis Association Using Sparse Signals (GAUSS), a method for gene set association analysis that requires only GWAS summary statistics. For each significantly associated gene set, GAUSS identifies the subset of genes that have the maximal evidence of association and can best account for the gene set association. Using pre-computed correlation structure among test statistics from a reference panel, our p value calculation is substantially faster than other permutation- or simulation-based approaches. In simulations with varying proportions of causal genes, we find that GAUSS effectively controls type 1 error rate and has greater power than several existing methods, particularly when a small proportion of genes account for the gene set signal. Using GAUSS, we analyzed UK Biobank GWAS summary statistics for 10,679 gene sets and 1,403 binary phenotypes. We found that GAUSS is scalable and identified 13,466 phenotype and gene set association pairs. Within these gene sets, we identify an average of 17.2 (max = 405) genes that underlie these gene set associations.
Collapse
Affiliation(s)
- Diptavo Dutta
- Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biostatistics, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Peter VandeHaar
- Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lars G Fritsche
- Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sebastian Zöllner
- Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Michael Boehnke
- Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Laura J Scott
- Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Seunggeun Lee
- Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA; Graduate School of Data Science, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
14
|
Yoon S, Baik B, Park T, Nam D. Powerful p-value combination methods to detect incomplete association. Sci Rep 2021; 11:6980. [PMID: 33772054 PMCID: PMC7997958 DOI: 10.1038/s41598-021-86465-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 03/08/2021] [Indexed: 12/13/2022] Open
Abstract
Meta-analyses increase statistical power by combining statistics from multiple studies. Meta-analysis methods have mostly been evaluated under the condition that all the data in each study have an association with the given phenotype. However, specific experimental conditions in each study or genetic heterogeneity can result in "unassociated statistics" that are derived from the null distribution. Here, we show that power of conventional meta-analysis methods rapidly decreases as an increasing number of unassociated statistics are included, whereas the classical Fisher's method and its weighted variant (wFisher) exhibit relatively high power that is robust to addition of unassociated statistics. We also propose another robust method based on joint distribution of ordered p-values (ordmeta). Simulation analyses for t-test, RNA-seq, and microarray data demonstrated that wFisher and ordmeta, when only a small number of studies have an association, outperformed existing meta-analysis methods. We performed meta-analyses of nine microarray datasets (prostate cancer) and four association summary datasets (body mass index), where our methods exhibited high biological relevance and were able to detect genes that the-state-of-the-art methods missed. The metapro R package that implements the proposed methods is available from both CRAN and GitHub ( http://github.com/unistbig/metapro ).
Collapse
Affiliation(s)
- Sora Yoon
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, 44919, Republic of Korea
| | - Bukyung Baik
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, 44919, Republic of Korea
| | - Taesung Park
- Department of Statistics, Seoul National University, Seoul, 08826, Republic of Korea
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826, Republic of Korea
| | - Dougu Nam
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, 44919, Republic of Korea.
- Department of Mathematical Sciences, Ulsan National Institute of Science and Technology, Ulsan, 44919, Republic of Korea.
| |
Collapse
|
15
|
Li Z, Qin S, Li Q. A novel test by combining the maximum and minimum values among a large number of dependent Z-scores with application to genome wide association study. Stat Med 2021; 40:2422-2434. [PMID: 33665825 DOI: 10.1002/sim.8912] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 01/19/2021] [Accepted: 01/30/2021] [Indexed: 12/22/2022]
Abstract
In this article, we propose a novel test via combining the maximum and minimum values among a large number of dependent Z-scores for testing the hypothesis with sparse signals. The proposed test employs the information about different signs of maximum and minimum Z-scores and thus power is gained. Its asymptotic null distribution is derived under the null hypothesis and some regular conditions. Extensive simulation studies are conducted to show the advantages of the proposed test by comparing with two existing ones. A real application to the lipids genome wide association study further shows its performances.
Collapse
Affiliation(s)
- Zhengbang Li
- School of Mathematics and Statistics, Central China Normal University, Wuhan, China
| | - Sanan Qin
- School of Mathematics and Statistics, Central China Normal University, Wuhan, China
| | - Qizhai Li
- LSC, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing, China.,School of Mathematical Sciences, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
16
|
Abstract
Many high-dimensional hypothesis tests aim to globally examine marginal or low-dimensional features of a high-dimensional joint distribution, such as testing of mean vectors, covariance matrices and regression coefficients. This paper constructs a family of U-statistics as unbiased estimators of the ℓ p -norms of those features. We show that under the null hypothesis, the U-statistics of different finite orders are asymptotically independent and normally distributed. Moreover, they are also asymptotically independent with the maximum-type test statistic, whose limiting distribution is an extreme value distribution. Based on the asymptotic independence property, we propose an adaptive testing procedure which combines p-values computed from the U-statistics of different orders. We further establish power analysis results and show that the proposed adaptive procedure maintains high power against various alternatives.
Collapse
Affiliation(s)
- Yinqiu He
- Department of Statistics, University of Michigan
| | - Gongjun Xu
- Department of Statistics, University of Michigan
| | - Chong Wu
- Department of Statistics, Florida State University
| | - Wei Pan
- Division of Biostatistics, School of Public Health, University of Minnesota
| |
Collapse
|
17
|
Wendeu-Foyet MG, Cénée S, Koudou Y, Trétarre B, Rébillard X, Cancel-Tassin G, Cussenot O, Boland A, Olaso R, Deleuze JF, Blanché H, Lamy PJ, Mulot C, Laurent-Puig P, Truong T, Menegaux F. Circadian genes polymorphisms, night work and prostate cancer risk: Findings from the EPICAP study. Int J Cancer 2020; 147:3119-3129. [PMID: 32506468 DOI: 10.1002/ijc.33139] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 05/04/2020] [Accepted: 05/18/2020] [Indexed: 11/10/2022]
Abstract
Over the past two decades, several studies have attempted to understand the hypothesis that disrupting the circadian rhythm may promote the development of cancer. Some have suggested that night work and some circadian genes polymorphisms are associated with cancer, including prostate cancer. Our study aims to test the hypothesis that prostate cancer risk among night workers may be modulated by genetic polymorphisms in the circadian pathway genes based on data from the EPICAP study, a population-based case-control study including 1511 men (732 cases/779 controls) with genotyped data. We estimated odds ratio (ORs) and P values of the association between prostate cancer and circadian gene variants using logistic regression models. We tested the interaction between circadian genes variants and night work indicators that were significantly associated with prostate cancer at pathway, gene and SNP levels. Analyses were also stratified by each of these night work indicators and by cancer aggressiveness. The circadian pathway was significantly associated with aggressive prostate cancer among night workers (P = .004), particularly for men who worked at night for <20 years (P = .0002) and those who performed long night shift (>10 hours, P = .001). At the gene level, we observed among night workers significant associations between aggressive prostate cancer and ARNTL, NPAS2 and RORA. At the SNP-level, no significant association was observed. Our findings provide some clues of a potential modulating effect of circadian genes in the relationship between night work and prostate cancer. Further investigation is warranted to confirm these findings and to better elucidate the biological pathways involved.
Collapse
Affiliation(s)
| | - Sylvie Cénée
- Université Paris-Saclay, UVSQ, Inserm, CESP, Villejuif, France
| | - Yves Koudou
- Université Paris-Saclay, UVSQ, Inserm, CESP, Villejuif, France
| | - Brigitte Trétarre
- Registre des Tumeurs de l'Hérault, EA 2415, ICM, Montpellier, France
| | | | - Géraldine Cancel-Tassin
- CeRePP, Hopital Tenon, Paris, France
- Sorbonne Université, GRC no. 5, ONCOTYPE-URO, AP-HP, Hôpital Tenon, Paris, France
| | - Olivier Cussenot
- CeRePP, Hopital Tenon, Paris, France
- Sorbonne Université, GRC no. 5, ONCOTYPE-URO, AP-HP, Hôpital Tenon, Paris, France
| | - Anne Boland
- Centre National de Recherche en Génomique Humaine (CNRGH), Institut de Biologie François Jacob, CEA, Université Paris-Saclay, Saint-Aubin, France
| | - Robert Olaso
- Centre National de Recherche en Génomique Humaine (CNRGH), Institut de Biologie François Jacob, CEA, Université Paris-Saclay, Saint-Aubin, France
| | - Jean-François Deleuze
- Centre National de Recherche en Génomique Humaine (CNRGH), Institut de Biologie François Jacob, CEA, Université Paris-Saclay, Saint-Aubin, France
| | - Hélène Blanché
- Centre d'Etude du Polymorphisme Humain (CEPH), Fondation Jean Dausset, Paris, France
| | - Pierre-Jean Lamy
- Clinique Beau Soleil, Service Urologie, Montpellier, France
- Institut médical d'Analyse Génomique-Imagenome, Labosud, Montpellier, France
| | - Claire Mulot
- Université Paris Descartes, INSERM UMR-S1147 EPIGENETEC, Paris, France
| | | | - Thérèse Truong
- Université Paris-Saclay, UVSQ, Inserm, CESP, Villejuif, France
| | | |
Collapse
|
18
|
Deng Y, Wu S, Fan H. Genome-wide pathway-based quantitative multiple phenotypes analysis. PLoS One 2020; 15:e0240910. [PMID: 33175855 PMCID: PMC7657528 DOI: 10.1371/journal.pone.0240910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 10/06/2020] [Indexed: 11/18/2022] Open
Abstract
For complex diseases, genome-wide pathway association studies have become increasingly promising. Currently, however, pathway-based association analysis mainly focus on a single phenotype, which may insufficient to describe the complex diseases and physiological processes. This work proposes a combination model to evaluate the association between a pathway and multiple phenotypes and to reduce the run time based on asymptotic results. For a single phenotype, we propose a semi-supervised maximum kernel-based U-statistics (mSKU) method to assess the pathway-based association analysis. For multiple phenotypes, we propose the fisher combination function with dependent phenotypes (FC) to transform the p-values between the pathway and each marginal phenotype individually to achieve pathway-based multiple phenotypes analysis. With real data from the Alzheimer Disease Neuroimaging Initiative (ADNI) study and Human Liver Cohort (HLC) study, the FC-mSKU method allows us to specify which pathways are specific to a single phenotype or contribute to common genetic constructions of multiple phenotypes. If we only focus on single-phenotype tests, we may miss some findings for etiology studies. Through extensive simulation studies, the FC-mSKU method demonstrates its advantages compared with its counterparts.
Collapse
Affiliation(s)
- Yamin Deng
- Statistics Center, First Hospital of Shanxi Medical University, Taiyuan, China.,Division of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Shiman Wu
- Statistics Center, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Huifang Fan
- Statistics Center, First Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
19
|
Zhang L, Papachristou C, Choudhary PK, Biswas S. A Bayesian Hierarchical Framework for Pathway Analysis in Genome-Wide Association Studies. Hum Hered 2020; 84:240-255. [PMID: 32966977 DOI: 10.1159/000508664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 05/14/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Pathway analysis allows joint consideration of multiple SNPs belonging to multiple genes, which in turn belong to a biologically defined pathway. This type of analysis is usually more powerful than single-SNP analyses for detecting joint effects of variants in a pathway. METHODS We develop a Bayesian hierarchical model by fully modeling the 3-level hierarchy, namely, SNP-gene-pathway that is naturally inherent in the structure of the pathways, unlike the currently used ad hoc ways of combining such information. We model the effects at each level conditional on the effects of the levels preceding them within the generalized linear model framework. To deal with the high dimensionality, we regularize the regression coefficients through an appropriate choice of priors. The model is fit using a combination of iteratively weighted least squares and expectation-maximization algorithms to estimate the posterior modes and their standard errors. A normal approximation is used for inference. RESULTS We conduct simulations to study the proposed method and find that our method has higher power than some standard approaches in several settings for identifying pathways with multiple modest-sized variants. We illustrate the method by analyzing data from two genome-wide association studies on breast and renal cancers. CONCLUSION Our method can be helpful in detecting pathway association.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Mathematical Sciences, University of Texas at Dallas, Richardson, Texas, USA
| | | | - Pankaj K Choudhary
- Department of Mathematical Sciences, University of Texas at Dallas, Richardson, Texas, USA
| | - Swati Biswas
- Department of Mathematical Sciences, University of Texas at Dallas, Richardson, Texas, USA,
| |
Collapse
|
20
|
Ni J, Deng B, Zhu M, Wang Y, Yan C, Wang T, Liu Y, Li G, Ding Y, Jin G. Integration of GWAS and eQTL Analysis to Identify Risk Loci and Susceptibility Genes for Gastric Cancer. Front Genet 2020; 11:679. [PMID: 32754194 PMCID: PMC7366424 DOI: 10.3389/fgene.2020.00679] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 06/03/2020] [Indexed: 02/05/2023] Open
Abstract
Genome-wide association studies (GWAS) have identified several susceptibility loci for gastric cancer (GC), but the majority of identified single-nucleotide polymorphisms (SNPs) fall within the non-coding region and are likely to exert their biological function by modulating gene expression. To systematically estimate expression-associated SNPs (eSNPs) that confer genetic predisposition to GC, we evaluated the associations of 314,203 stomach tissue-specific eSNPs with GC risk in three GWAS datasets (2,631 cases and 4,373 controls). Subsequently, we conducted a gene-based analysis to calculate the cumulative effect of eSNPs through sequence kernel association combined test and Sherlock integrative analysis. At the SNP-level, we identified two novel variants (rs836545 at 7p22.1 and rs1892252 at 6p22.2) associated with GC risk. The risk allele carriers of rs836545-T and rs1892252-G exhibited higher expression levels of DAGLB (P = 3.70 × 10–18) and BTN3A2 (P = 3.20 × 10–5), respectively. Gene-based analyses identified DAGLB and FBXO43 as novel susceptibility genes for GC. DAGLB and FBXO43 were significantly overexpressed in GC tissues than in their adjacent tissues (P = 5.59 × 10–7 and P = 3.90 × 10–6, respectively), and high expression level of these two genes was associated with an unfavorable prognosis of GC patients (P = 1.30 × 10–7 and P = 7.60 × 10–3, respectively). Co-expression genes with these two novel genes in normal stomach tissues were significantly enriched in several cancer-related pathways, including P53, MAPK and TGF-beta pathways. In summary, our findings confirm the importance of eSNPs in dissecting the genetic basis of GC, and the identified eSNPs and relevant genes will provide new insight into the genetic and biological basis for the mechanism of GC development.
Collapse
Affiliation(s)
- Jing Ni
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, China
| | - Bin Deng
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou, China
| | - Meng Zhu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, China
| | - Yuzhuo Wang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, China
| | - Caiwang Yan
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, China
| | - Tianpei Wang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, China
| | - Yaqian Liu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, China
| | - Gang Li
- Department of General Surgery, Jiangsu Institute of Cancer Research, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Yanbing Ding
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou, China
| | - Guangfu Jin
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
21
|
Statistical Method Based on Bayes-Type Empirical Score Test for Assessing Genetic Association with Multilocus Genotype Data. Int J Genomics 2020; 2020:4708152. [PMID: 32455126 PMCID: PMC7229558 DOI: 10.1155/2020/4708152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 04/21/2020] [Indexed: 12/20/2022] Open
Abstract
Simultaneous testing of multiple genetic variants for association is widely recognized as a valuable complementary approach to single-marker tests. As such, principal component regression (PCR) has been found to have competitive power. We focus on exploring a robust test for an unknown genetic mode of all SNPs, an unknown Hardy-Weinberg equilibrium (HWE) in a population, and a large number of all SNPs. First, we propose a new global test by means of the use of codominant codes for all markers and PCR. The new global test is built on an empirical Bayes-type score statistic for testing marginal associations with each single marker. The new global test gains power by robustly exploiting the Hardy-Weinberg equilibrium in the control population and effectively using linkage disequilibrium among test markers. The new global test reduces to PCR when the genotype for each marker is coded as the number of minor alleles. This connection lends insight into the power of the new global test relative to PCR and some other popular multimarker test methods. Second, we propose a robust test method based on the new global test and the ordinary PCR test built on a prospective score statistic for testing marginal associations with each single marker when the genotype for each marker is coded as the number of minor alleles by taking the minimum p value of these two tests. Finally, through extensive simulation studies and analysis of the association between pancreatic cancer and some genes of interest, we show that the proposed robust test method has desirable power and can often identify association signals that may be missed by existing methods.
Collapse
|
22
|
Fifteen Years of Gene Set Analysis for High-Throughput Genomic Data: A Review of Statistical Approaches and Future Challenges. ENTROPY 2020; 22:e22040427. [PMID: 33286201 PMCID: PMC7516904 DOI: 10.3390/e22040427] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/18/2020] [Accepted: 04/03/2020] [Indexed: 12/22/2022]
Abstract
Over the last decade, gene set analysis has become the first choice for gaining insights into underlying complex biology of diseases through gene expression and gene association studies. It also reduces the complexity of statistical analysis and enhances the explanatory power of the obtained results. Although gene set analysis approaches are extensively used in gene expression and genome wide association data analysis, the statistical structure and steps common to these approaches have not yet been comprehensively discussed, which limits their utility. In this article, we provide a comprehensive overview, statistical structure and steps of gene set analysis approaches used for microarrays, RNA-sequencing and genome wide association data analysis. Further, we also classify the gene set analysis approaches and tools by the type of genomic study, null hypothesis, sampling model and nature of the test statistic, etc. Rather than reviewing the gene set analysis approaches individually, we provide the generation-wise evolution of such approaches for microarrays, RNA-sequencing and genome wide association studies and discuss their relative merits and limitations. Here, we identify the key biological and statistical challenges in current gene set analysis, which will be addressed by statisticians and biologists collectively in order to develop the next generation of gene set analysis approaches. Further, this study will serve as a catalog and provide guidelines to genome researchers and experimental biologists for choosing the proper gene set analysis approach based on several factors.
Collapse
|
23
|
Boag AM, Short A, Kennedy LJ, Syme H, Graham PA, Catchpole B. Polymorphisms in the CTLA4 promoter sequence are associated with canine hypoadrenocorticism. Canine Med Genet 2020; 7:2. [PMID: 32835228 PMCID: PMC7371821 DOI: 10.1186/s40575-020-0081-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 02/18/2020] [Indexed: 12/24/2022] Open
Abstract
Background Canine hypoadrenocorticism is an immune-mediated endocrinopathy that shares both clinical and pathophysiological similarities with Addison’s disease in humans. Several dog breeds are overrepresented in the disease population, suggesting that a genetic component is involved, although this is likely to be polygenic. Previous research has implicated CTLA4 as a potential susceptibility gene. CTLA4 is an important regulator of T cell function and polymorphisms/mutations in CTLA4 have been associated with a number of autoimmune phenotypes in both humans and rodent models of autoimmunity. The aim of the current study was to undertake a case:control association study of CTLA4 promotor polymorphisms in three dog breeds, cocker spaniels, springer spaniels and West Highland white terriers (WHWT). Results Polymorphisms in the CTLA4 promoter were determined by PCR and sequence-based typing. There were significant associations with three promoter haplotypes in cocker spaniels (p = 0.003). A series of SNPs were also associated with hypoadrenocorticism in cocker spaniels and springer spaniels, including polymorphisms in predicted NFAT and SP1 transcription factor binding sites. Conclusions This study provides further evidence that CTLA4 promotor polymorphisms are associated with this complex genetic disease and supports an immune mediated aetiopathogenesis of canine hypoadrenocorticism.
Collapse
Affiliation(s)
- Alisdair M Boag
- Pathobiology and Population Sciences, The Royal Veterinary College, University of London, London, UK.,The Queen's Medical Research Institute, Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Andrea Short
- Centre for Integrated Genomic Medical Research, University of Manchester, Manchester, UK
| | - Lorna J Kennedy
- Centre for Integrated Genomic Medical Research, University of Manchester, Manchester, UK
| | - Hattie Syme
- Clinical Science and Services, The Royal Veterinary College, University of London, London, UK
| | - Peter A Graham
- Faculty of Medicine & Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Nottingham, UK
| | - Brian Catchpole
- Pathobiology and Population Sciences, The Royal Veterinary College, University of London, London, UK
| |
Collapse
|
24
|
Zhang H, Tong T, Landers J, Wu Z. TFisher: A powerful truncation and weighting procedure for combining $p$-values. Ann Appl Stat 2020. [DOI: 10.1214/19-aoas1302] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
25
|
Chimusa ER, Dalvie S, Dandara C, Wonkam A, Mazandu GK. Post genome-wide association analysis: dissecting computational pathway/network-based approaches. Brief Bioinform 2020; 20:690-700. [PMID: 29701762 DOI: 10.1093/bib/bby035] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 04/04/2018] [Indexed: 02/02/2023] Open
Abstract
Over thousands of genetic associations to diseases have been identified by genome-wide association studies (GWASs), which conceptually is a single-marker-based approach. There are potentially many uses of these identified variants, including a better understanding of the pathogenesis of diseases, new leads for studying underlying risk prediction and clinical prediction of treatment. However, because of inadequate power, GWAS might miss disease genes and/or pathways with weak genetic or strong epistatic effects. Driven by the need to extract useful information from GWAS summary statistics, post-GWAS approaches (PGAs) were introduced. Here, we dissect and discuss advances made in pathway/network-based PGAs, with a particular focus on protein-protein interaction networks that leverage GWAS summary statistics by combining effects of multiple loci, subnetworks or pathways to detect genetic signals associated with complex diseases. We conclude with a discussion of research areas where further work on summary statistic-based methods is needed.
Collapse
Affiliation(s)
- Emile R Chimusa
- Division of Human Genetics, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Level 3, Wernher and Beit North, Private Bag, Rondebosch, 7700, Anzio road, Observatory Cape Town, South Africa
| | - Shareefa Dalvie
- Department of Psychiatry and Mental Health, University of Cape Town, Observatory, 7925, Cape Town, South Africa
| | - Collet Dandara
- Division of Human Genetics, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Private Bag, Rondebosch, 7700, Cape Town, South Africa
| | - Ambroise Wonkam
- Division of Human Genetics, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Private Bag, Rondebosch, 7700, Cape Town, South Africa
| | - Gaston K Mazandu
- Division of Human Genetics, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Private Bag, Rondebosch, 7700, Cape Town, South Africa; African Institute for Mathematical Sciences, 7945 Muizenberg, Cape Town, South Africa and Computational Biology Division, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Medical School, Anzio Road, Observatory, 7925, Cape Town, South Africa
| |
Collapse
|
26
|
Vsevolozhskaya OA, Hu F, Zaykin DV. Detecting Weak Signals by Combining Small P-Values in Genetic Association Studies. Front Genet 2019; 10:1051. [PMID: 31824555 PMCID: PMC6879667 DOI: 10.3389/fgene.2019.01051] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/30/2019] [Indexed: 01/31/2023] Open
Abstract
We approach the problem of combining top-ranking association statistics or P-values from a new perspective which leads to a remarkably simple and powerful method. Statistical methods, such as the rank truncated product (RTP), have been developed for combining top-ranking associations, and this general strategy proved to be useful in applications for detecting combined effects of multiple disease components. To increase power, these methods aggregate signals across top ranking single nucleotide polymorphisms (SNPs), while adjusting for their total number assessed in a study. Analytic expressions for combined top statistics or P-values tend to be unwieldy, which complicates interpretation and practical implementation and hinders further developments. Here, we propose the augmented rank truncation (ART) method that retains main characteristics of the RTP but is substantially simpler to implement. ART leads to an efficient form of the adaptive algorithm, an approach where the number of top ranking SNPs is varied to optimize power. We illustrate our methods by strengthening previously reported associations of μ-opioid receptor variants with sensitivity to pain.
Collapse
Affiliation(s)
- Olga A. Vsevolozhskaya
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, KY, United States
| | - Fengjiao Hu
- Biostatistics and Computational Biology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - Dmitri V. Zaykin
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
27
|
Fedirko V, Mandle HB, Zhu W, Hughes DJ, Siddiq A, Ferrari P, Romieu I, Riboli E, Bueno-de-Mesquita B, van Duijnhoven FJB, Siersema PD, Tjønneland A, Olsen A, Perduca V, Carbonnel F, Boutron-Ruault MC, Kühn T, Johnson T, Krasimira A, Trichopoulou A, Makrythanasis P, Thanos D, Panico S, Krogh V, Sacerdote C, Skeie G, Weiderpass E, Colorado-Yohar S, Sala N, Barricarte A, Sanchez MJ, Quirós R, Amiano P, Gylling B, Harlid S, Perez-Cornago A, Heath AK, Tsilidis KK, Aune D, Freisling H, Murphy N, Gunter MJ, Jenab M. Vitamin D-Related Genes, Blood Vitamin D Levels and Colorectal Cancer Risk in Western European Populations. Nutrients 2019; 11:E1954. [PMID: 31434255 PMCID: PMC6722852 DOI: 10.3390/nu11081954] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 08/09/2019] [Accepted: 08/12/2019] [Indexed: 12/11/2022] Open
Abstract
Higher circulating 25-hydroxyvitamin D levels (25(OH)D) have been found to be associated with lower risk for colorectal cancer (CRC) in prospective studies. Whether this association is modified by genetic variation in genes related to vitamin D metabolism and action has not been well studied in humans. We investigated 1307 functional and tagging single-nucleotide polymorphisms (SNPs; individually, and by gene/pathway) in 86 vitamin D-related genes in 1420 incident CRC cases matched to controls from the European Prospective Investigation into Cancer and Nutrition (EPIC) cohort. We also evaluated the association between these SNPs and circulating 25(OH)D in a subset of controls. We confirmed previously reported CRC risk associations between SNPs in the VDR, GC, and CYP27B1 genes. We also identified additional associations with 25(OH)D, as well as CRC risk, and several potentially novel SNPs in genes related to vitamin D transport and action (LRP2, CUBN, NCOA7, and HDAC9). However, none of these SNPs were statistically significant after Benjamini-Hochberg (BH) multiple testing correction. When assessed by a priori defined functional pathways, tumor growth factor β (TGFβ) signaling was associated with CRC risk (P ≤ 0.001), with most statistically significant genes being SMAD7 (PBH = 0.008) and SMAD3 (PBH = 0.008), and 18 SNPs in the vitamin D receptor (VDR) binding sites (P = 0.036). The 25(OH)D-gene pathway analysis suggested that genetic variants in the genes related to VDR complex formation and transcriptional activity are associated with CRC depending on 25(OH)D levels (interaction P = 0.041). Additional studies in large populations and consortia, especially with measured circulating 25(OH)D, are needed to confirm our findings.
Collapse
Affiliation(s)
- Veronika Fedirko
- Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA.
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA.
| | - Hannah B Mandle
- Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - Wanzhe Zhu
- Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - David J Hughes
- Cancer Biology and Therapeutics Group (CBT), Conway Institute, School of Biomolecular and Biomedical Science (SBBS), University College Dublin, Dublin, Ireland
| | - Afshan Siddiq
- Genomics England, London EC1M 6BQ, UK
- Imperial College London, London SW7 2AZ, UK
| | - Pietro Ferrari
- Section of Nutrition and Metabolism, International Agency for Research on Cancer (IARC-WHO), Lyon 69372, France
| | - Isabelle Romieu
- Section of Nutrition and Metabolism, International Agency for Research on Cancer (IARC-WHO), Lyon 69372, France
| | - Elio Riboli
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London SW7 2AZ, UK
| | - Bas Bueno-de-Mesquita
- Division of Human Nutrition & Health, Wageningen University & Research, 6700 AA Wageningen, The Netherlands
| | - Fränzel J B van Duijnhoven
- Division of Human Nutrition & Health, Wageningen University & Research, 6700 AA Wageningen, The Netherlands
| | - Peter D Siersema
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Anne Tjønneland
- Danish Cancer Society Research Center, 2100 Copenhagen, Denmark
| | - Anja Olsen
- Danish Cancer Society Research Center, 2100 Copenhagen, Denmark
| | - Vittorio Perduca
- Laboratoire de Mathématiques Appliquées MAP5, Université Paris Descartes, 75006 Paris, France
- CESP, Fac. de médecine-Univ. Paris-Sud, Fac. de médecine-UVSQ, INSERM, Université Paris-Saclay, F-94805 Villejuif, France
- Gustave Roussy, F-94805 Villejuif, France
| | - Franck Carbonnel
- CESP, Fac. de médecine-Univ. Paris-Sud, Fac. de médecine-UVSQ, INSERM, Université Paris-Saclay, F-94805 Villejuif, France
- Gustave Roussy, F-94805 Villejuif, France
- Department of Gastroenterology, Bicêtre University Hospital, Assistance Publique des Hôpitaux de Paris, 94270 Le Kremlin Bicêtre, France
| | - Marie-Christine Boutron-Ruault
- CESP, Fac. de médecine-Univ. Paris-Sud, Fac. de médecine-UVSQ, INSERM, Université Paris-Saclay, F-94805 Villejuif, France
- Gustave Roussy, F-94805 Villejuif, France
| | - Tilman Kühn
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Theron Johnson
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Aleksandrova Krasimira
- Nutrition, Immunity and Metabolism, Department of Epidemiology, German Institute for Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert Allee, 14558 Nuthetal, Germany
| | | | - Periklis Makrythanasis
- Hellenic Health Foundation, 115 27 Athens, Greece
- Biomedical Research Foundation of the Academy of Athens, 115 27 Athens, Greece
| | - Dimitris Thanos
- Hellenic Health Foundation, 115 27 Athens, Greece
- Biomedical Research Foundation of the Academy of Athens, 115 27 Athens, Greece
| | - Salvatore Panico
- Dipartimento Di Medicina Clinica E Chirurgia, Federico Ii University, 80138 Naples, Italy
| | - Vittorio Krogh
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian, 20133 Milano, Italy
| | - Carlotta Sacerdote
- Unit of Cancer Epidemiology, Città della Salute e della Scienza University-Hospital and Center for Cancer Prevention (CPO), 10126 Turin, Italy
| | - Guri Skeie
- Department of Community Medicine, Faculty of Health Sciences, University of Tromsø, The Arctic University of Norway, 9019 Tromsø, Norway
| | - Elisabete Weiderpass
- Department of Community Medicine, Faculty of Health Sciences, University of Tromsø, The Arctic University of Norway, 9019 Tromsø, Norway
- Department of Research, Cancer Registry of Norway, Institute of Population-Based Cancer Research, N-0304 Oslo, Norway
- Department of Medical Epidemiology and Biostatistics, Karolinska Institut, SE-171 77 Stockholm, Sweden
- Genetic Epidemiology Group, Folkhälsan Research Center and Faculty of Medicine, Helsinki University, Helsinki 00014, Finland
- International Agency for Research on Cancer (IARC-WHO), Lyon 69372, France
| | - Sandra Colorado-Yohar
- Department of Epidemiology, Murcia Regional Health Council, IMIB-Arrixaca, Murcia 30008, Spain
- CIBER Epidemiology and Public Healh (CIBERESP), Madrid 28029, Spain
- Research Group on Demography and Health, National Faculty of Public Health, University of Antioquia, Cl. 67 ##53-108 Medellín, Colombia
| | - Núria Sala
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Program, and Translational Research Laboratory, Catalan Institute of Oncology (ICO)-IDIBELL, 08908 Barcelona, Spain
| | - Aurelio Barricarte
- CIBER Epidemiology and Public Healh (CIBERESP), Madrid 28029, Spain
- Navarra Public Health Institute, Pamplona 31008, Spain
| | - Maria-Jose Sanchez
- CIBER Epidemiology and Public Healh (CIBERESP), Madrid 28029, Spain
- Escuela Andaluza de Salud Pública, Instituto de Investigación Biosanitaria (ibs.GRANADA), Granada 18012, Spain
| | - Ramón Quirós
- Public Health Directorate, Asturias 33006, Spain
| | - Pilar Amiano
- CIBER Epidemiology and Public Healh (CIBERESP), Madrid 28029, Spain
- Public Health Division of Gipuzkoa, BioDonostia Research Institute, San Sebastian 20014, Spain
| | - Björn Gylling
- Department of Medical Biosciences, Pathology, Umeå University, 901 87 Umeå, Sweden
| | - Sophia Harlid
- Department of Radiation Sciences, Oncology, Umeå University, 901 87 Umeå, Sweden
| | - Aurora Perez-Cornago
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Alicia K Heath
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London SW7 2AZ, UK
| | - Konstantinos K Tsilidis
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London SW7 2AZ, UK
- Department of Hygiene and Epidemiology, University of Ioannina School of Medicine, Ioannina 45110, Greece
| | - Dagfinn Aune
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London SW7 2AZ, UK
- Department of Nutrition, Bjørknes University College, 0456 Oslo, Norway
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, 0372 Oslo, Norway
| | - Heinz Freisling
- Section of Nutrition and Metabolism, International Agency for Research on Cancer (IARC-WHO), Lyon 69372, France
| | - Neil Murphy
- Section of Nutrition and Metabolism, International Agency for Research on Cancer (IARC-WHO), Lyon 69372, France
| | - Marc J Gunter
- Section of Nutrition and Metabolism, International Agency for Research on Cancer (IARC-WHO), Lyon 69372, France
| | - Mazda Jenab
- Section of Nutrition and Metabolism, International Agency for Research on Cancer (IARC-WHO), Lyon 69372, France.
| |
Collapse
|
28
|
Khlebus E, Kutsenko V, Meshkov A, Ershova A, Kiseleva A, Shevtsov A, Shcherbakova N, Zharikova A, Lankin V, Tikhaze A, Chazova I, Yarovaya E, Drapkina O, Boytsov S. Multiple rare and common variants in APOB gene locus associated with oxidatively modified low-density lipoprotein levels. PLoS One 2019; 14:e0217620. [PMID: 31150472 PMCID: PMC6544350 DOI: 10.1371/journal.pone.0217620] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 05/15/2019] [Indexed: 01/17/2023] Open
Abstract
Oxidatively modified low-density lipoproteins (oxLDL) play an important role in the occurrence and progression of atherosclerosis. To identify the genetic factors influencing the oxLDL levels, we have genotyped 776 DNA samples of Russian individuals for 196,725 single-nucleotide polymorphisms (SNPs) using the Cardio-MetaboChip (Illumina, USA) and conducted genome-wide association study (GWAS). Fourteen common variants in the locus including APOB gene were significantly associated with the oxLDL levels (P < 2.18 × 10−7). These variants explained only 6% of the variation in the oxLDL levels. Then, we assessed the contribution of rare coding variants of APOB gene to the oxLDL levels. Individuals with the extreme oxLDL levels (48 with the lowest and 48 with the highest values) were selected for targeted sequencing of the region including APOB gene. To evaluate the contribution of the SNPs to the oxLDL levels we used various statistical methods for the association analysis of rare variants: WST, SKAT, and SKAT-O. We revealed that both synonymous and nonsynonymous SNPs affected the oxLDL levels. For the joint analysis of the rare and common variants, we conducted the SKAT-C testing and found a group of 15 SNPs significantly associated with the oxLDL levels (P = 2.14 × 10−9). Our results indicate that the oxLDL levels depend on both common and rare variants of the APOB gene.
Collapse
Affiliation(s)
- Eleonora Khlebus
- Federal State Institution National Medical Research Center for Preventive Medicine of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
- Moscow Institute of Physics and Technology (State University), Moscow, Russia
- * E-mail:
| | - Vladimir Kutsenko
- Federal State Institution National Medical Research Center for Preventive Medicine of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
- Lomonosov Moscow State University, Moscow, Russia
| | - Alexey Meshkov
- Federal State Institution National Medical Research Center for Preventive Medicine of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Alexandra Ershova
- Federal State Institution National Medical Research Center for Preventive Medicine of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Anna Kiseleva
- Federal State Institution National Medical Research Center for Preventive Medicine of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | | | - Natalia Shcherbakova
- Federal State Institution National Medical Research Center for Preventive Medicine of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Anastasiia Zharikova
- Federal State Institution National Medical Research Center for Preventive Medicine of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Vadim Lankin
- Federal State Budget Organization National Medical Research Center of Cardiology of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Alla Tikhaze
- Federal State Budget Organization National Medical Research Center of Cardiology of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Irina Chazova
- Federal State Budget Organization National Medical Research Center of Cardiology of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | | | - Oksana Drapkina
- Federal State Institution National Medical Research Center for Preventive Medicine of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Sergey Boytsov
- Federal State Budget Organization National Medical Research Center of Cardiology of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| |
Collapse
|
29
|
Fedirko V, Jenab M, Méplan C, Jones JS, Zhu W, Schomburg L, Siddiq A, Hybsier S, Overvad K, Tjønneland A, Omichessan H, Perduca V, Boutron-Ruault MC, Kühn T, Katzke V, Aleksandrova K, Trichopoulou A, Karakatsani A, Kotanidou A, Tumino R, Panico S, Masala G, Agnoli C, Naccarati A, Bueno-de-Mesquita B, Vermeulen RCH, Weiderpass E, Skeie G, Nøst TH, Lujan-Barroso L, Quirós JR, Huerta JM, Rodríguez-Barranco M, Barricarte A, Gylling B, Harlid S, Bradbury KE, Wareham N, Khaw KT, Gunter M, Murphy N, Freisling H, Tsilidis K, Aune D, Riboli E, Hesketh JE, Hughes DJ. Association of Selenoprotein and Selenium Pathway Genotypes with Risk of Colorectal Cancer and Interaction with Selenium Status. Nutrients 2019; 11:E935. [PMID: 31027226 PMCID: PMC6520820 DOI: 10.3390/nu11040935] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 04/19/2019] [Accepted: 04/22/2019] [Indexed: 12/16/2022] Open
Abstract
Selenoprotein genetic variations and suboptimal selenium (Se) levels may contribute to the risk of colorectal cancer (CRC) development. We examined the association between CRC risk and genotype for single nucleotide polymorphisms (SNPs) in selenoprotein and Se metabolic pathway genes. Illumina Goldengate assays were designed and resulted in the genotyping of 1040 variants in 154 genes from 1420 cases and 1421 controls within the European Prospective Investigation into Cancer and Nutrition (EPIC) study. Multivariable logistic regression revealed an association of 144 individual SNPs from 63 Se pathway genes with CRC risk. However, regarding the selenoprotein genes, only TXNRD1 rs11111979 retained borderline statistical significance after adjustment for correlated tests (PACT = 0.10; PACT significance threshold was P < 0.1). SNPs in Wingless/Integrated (Wnt) and Transforming growth factor (TGF) beta-signaling genes (FRZB, SMAD3, SMAD7) from pathways affected by Se intake were also associated with CRC risk after multiple testing adjustments. Interactions with Se status (using existing serum Se and Selenoprotein P data) were tested at the SNP, gene, and pathway levels. Pathway analyses using the modified Adaptive Rank Truncated Product method suggested that genes and gene x Se status interactions in antioxidant, apoptosis, and TGF-beta signaling pathways may be associated with CRC risk. This study suggests that SNPs in the Se pathway alone or in combination with suboptimal Se status may contribute to CRC development.
Collapse
Affiliation(s)
- Veronika Fedirko
- Department of Epidemiology, Rollins School of Public Health & Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA.
| | - Mazda Jenab
- Section of Nutrition and Metabolism, International Agency for Research on Cancer, 69372 Lyon, France.
| | - Catherine Méplan
- School of Biomedical Sciences, Newcastle University, Newcastle upon Tyne NE1 7RU, UK.
| | - Jeb S Jones
- Department of Epidemiology, Rollins School of Public Health & Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA.
| | - Wanzhe Zhu
- Department of Epidemiology, Rollins School of Public Health & Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA.
| | - Lutz Schomburg
- Institute for Experimental Endocrinology, University Medical School, D-13353 Berlin, Germany.
| | - Afshan Siddiq
- Department of Epidemiology and Biostatistics, The School of Public Health, Imperial College London, London W2 1PG, UK.
| | - Sandra Hybsier
- Institute for Experimental Endocrinology, University Medical School, D-13353 Berlin, Germany.
| | - Kim Overvad
- Department of Public Health, Section for Epidemiology, Aarhus University, 8000 Aarhus, Denmark.
| | - Anne Tjønneland
- Diet, Genes and Environment Unit, Danish Cancer Society Research Center, DK 2100 Copenhagen, Denmark.
| | - Hanane Omichessan
- Faculty of Medicine, CESP, University of Paris-Sud, Faculty of Medicine UVSQ, INSERM, University of Paris-Saclay, 94805 Villejuif, France.
- Centre for Research in Epidemiology and Population Health (CESP), F-94805 Gustave Roussy, Villejuif, France.
| | - Vittorio Perduca
- Faculty of Medicine, CESP, University of Paris-Sud, Faculty of Medicine UVSQ, INSERM, University of Paris-Saclay, 94805 Villejuif, France.
- Centre for Research in Epidemiology and Population Health (CESP), F-94805 Gustave Roussy, Villejuif, France.
- Laboratory of Applied Mathematics, MAP5 (UMR CNRS 8145), University of Paris Descartes, 75270 Paris, France.
| | - Marie-Christine Boutron-Ruault
- Faculty of Medicine, CESP, University of Paris-Sud, Faculty of Medicine UVSQ, INSERM, University of Paris-Saclay, 94805 Villejuif, France.
- Centre for Research in Epidemiology and Population Health (CESP), F-94805 Gustave Roussy, Villejuif, France.
| | - Tilman Kühn
- Division of Cancer Epidemiology, German Cancer Research Centre (DKFZ), 69120 Heidelberg, Germany.
| | - Verena Katzke
- Division of Cancer Epidemiology, German Cancer Research Centre (DKFZ), 69120 Heidelberg, Germany.
| | - Krasimira Aleksandrova
- Department of Epidemiology, German Institute of Human Nutrition Potsdam-Rehbrücke, 14558 Nuthetal, Germany.
| | | | - Anna Karakatsani
- Hellenic Health Foundation, 115 27 Athens, Greece.
- 2nd Pulmonary Medicine Department, School of Medicine, National and Kapodistrian University of Athens, "ATTIKON" University Hospital, 106 79 Haidari, Greece.
| | - Anastasia Kotanidou
- Hellenic Health Foundation, 115 27 Athens, Greece.
- 1st Department of Critical Care Medicine and Pulmonary Services, University of Athens Medical School, Evangelismos Hospital, 106 76 Athens, Greece.
| | - Rosario Tumino
- Cancer Registry and Histopathology Department, Civic M.P. Arezzo Hospital, 97100 Ragusa, Italy.
| | - Salvatore Panico
- Department of Clinical Medicine and Surgery, Federico II University, 80138 Naples, Italy.
| | - Giovanna Masala
- Cancer Risk Factors and Life-Style Epidemiology Unit, Cancer Research and Prevention Institute-ISPO, 50141 Florence, Italy.
| | - Claudia Agnoli
- Epidemiology and Prevention Unit, IRCCS Foundation National Cancer Institute, 20133 Milan, Italy.
| | - Alessio Naccarati
- Molecular and Genetic Epidemiology Unit, Italian Institute for Genomic Medicine (IIGM) Torino, 10126 Torino, Italy.
| | - Bas Bueno-de-Mesquita
- Department of Epidemiology and Biostatistics, The School of Public Health, Imperial College London, London W2 1PG, UK.
- Department for Determinants of Chronic Diseases (DCD), National Institute for Public Health and the Environment (RIVM), 3720 Bilthoven, The Netherlands.
- Department of Gastroenterology and Hepatology, University Medical Centre, 3584 CX Utrecht, The Netherlands.
- Department of Social and Preventive Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia.
| | - Roel C H Vermeulen
- Institute of Risk Assessment Sciences, Utrecht University, 3512 JE Utrecht, The Netherlands.
| | - Elisabete Weiderpass
- Department of Research, Cancer Registry of Norway, Institute of Population-Based Cancer Research, N-0304 Oslo, Norway.
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, SE-171 77 Stockholm, Sweden.
- Genetic Epidemiology Group, Folkhälsan Research Center, and Faculty of Medicine, Helsinki University, 00014 Helsinki, Finland.
- Department of Community Medicine, University of Tromsø, The Arctic University of Norway, 9019 Tromsø, Norway.
| | - Guri Skeie
- Department of Community Medicine, University of Tromsø, The Arctic University of Norway, 9019 Tromsø, Norway.
| | - Therese Haugdahl Nøst
- Department of Community Medicine, University of Tromsø, The Arctic University of Norway, 9019 Tromsø, Norway.
| | - Leila Lujan-Barroso
- Unit of Nutrition and Cancer, Catalan Institute of Oncology (ICO-IDIBELL), L'Hospitalet de Llobregat, 08908 Barcelona, Spain.
| | - J Ramón Quirós
- EPIC Asturias, Public Health Directorate, 33006 Oviedo, Asturias, Spain.
| | - José María Huerta
- Department of Epidemiology, Murcia Regional Health Council, IMIB-Arrixaca, 30008 Murcia, Spain.
- CIBER Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain.
| | - Miguel Rodríguez-Barranco
- CIBER Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain.
- Andalucia School of Public Health, Institute for Biosanitary Research, University Hospital of Granada, University of Granada, 18011 Granada, Spain.
| | - Aurelio Barricarte
- CIBER Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain.
- Epidemiology, Prevention and Promotion Health Service, Navarra Public Health Institute, 31003 Pamplona, Spain.
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain.
| | - Björn Gylling
- Department of Medical Biosciences, Pathology, Umea University, 901 87 Umea, Sweden.
| | - Sophia Harlid
- Department of Radiation Sciences, Oncology, Umea University, 901 87 Umea, Sweden.
| | - Kathryn E Bradbury
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK.
| | - Nick Wareham
- MRC Epidemiology Unit, University of Cambridge, CB2 0QQ Cambridge, UK.
| | - Kay-Tee Khaw
- School of Clinical Medicine, University of Cambridge, Clinical Gerontology Unit, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK.
| | - Marc Gunter
- Section of Nutrition and Metabolism, International Agency for Research on Cancer, 69372 Lyon, France.
| | - Neil Murphy
- Section of Nutrition and Metabolism, International Agency for Research on Cancer, 69372 Lyon, France.
| | - Heinz Freisling
- Section of Nutrition and Metabolism, International Agency for Research on Cancer, 69372 Lyon, France.
| | - Kostas Tsilidis
- Department of Epidemiology and Biostatistics, The School of Public Health, Imperial College London, London W2 1PG, UK.
- Department of Hygiene and Epidemiology, University of Ioannina School of Medicine, 45110 Ioannina, Greece.
| | - Dagfinn Aune
- Department of Epidemiology and Biostatistics, The School of Public Health, Imperial College London, London W2 1PG, UK.
- Department of Nutrition, Bjørknes University College, 0456 Oslo, Norway.
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, 0372 Oslo, Norway.
| | - Elio Riboli
- Department of Epidemiology and Biostatistics, The School of Public Health, Imperial College London, London W2 1PG, UK.
| | - John E Hesketh
- School of Biomedical Sciences, Newcastle University, Newcastle upon Tyne NE1 7RU, UK.
| | - David J Hughes
- Cancer Biology and Therapeutics Group, UCD Conway Institute, School of Biomolecular and Biomedical Science, University College Dublin, D04 V1W8 Dublin, Ireland.
| |
Collapse
|
30
|
Berrandou T, Mulot C, Cordina-Duverger E, Arveux P, Laurent-Puig P, Truong T, Guénel P. Association of breast cancer risk with polymorphisms in genes involved in the metabolism of xenobiotics and interaction with tobacco smoking: A gene-set analysis. Int J Cancer 2019; 144:1896-1908. [PMID: 30303517 DOI: 10.1002/ijc.31917] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 09/20/2018] [Accepted: 09/25/2018] [Indexed: 12/14/2022]
Abstract
Single nucleotide polymorphisms (SNPs) in genes involved in xenobiotics metabolism (XM) are suspected to play a role in breast cancer risk. However, previous findings based on a SNP by SNP approach need to be replicated taking into account the combined effects of multiple SNPs. We used a gene-set analysis method to study the association between breast cancer risk and genetic variation in XM genes (seen as a set of SNPs) and in the XM pathway (seen as a set of genes). We also studied the interaction between variants in XM genes and tobacco smoking. The analysis was conducted in a case-control study of 1,125 cases and 1,172 controls. Using a dedicated chip, genotyping data of 585 SNPs in 68 XM genes were available. Genetic variation in the whole XM pathway was significantly associated with premenopausal breast cancer risk (p = 0.008). This association was mainly driven by genetic variation in NAT2, CYP2C18, CYP2C19, AKR1C2 and ALDH1A3. The association between the XM gene pathway and breast cancer was observed among current and previous smokers, but not among never smokers (p = 0.013 for interaction between XM genes and tobacco smoking status). The association with breast cancer risk indicates that XM genes variants may play a role in breast carcinogenesis through their detoxification function of environmental pollutants, such as those contained in tobacco smoke.
Collapse
Affiliation(s)
- Takiy Berrandou
- INSERM, Center for Research in Epidemiology and Population Health (CESP), Cancer and Environment team, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Claire Mulot
- INSERM, UMR-S 1147, CRB EPIGENETEC, Université Paris Descartes, Université Sorbonne Paris Cité, Paris, France
| | - Emilie Cordina-Duverger
- INSERM, Center for Research in Epidemiology and Population Health (CESP), Cancer and Environment team, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Patrick Arveux
- Breast and Gynaecologic Cancer Registry of Côte d'Or, Georges-François Leclerc Comprehensive Cancer Care Centre, Dijon, France
| | - Pierre Laurent-Puig
- INSERM, UMR-S 1147, CRB EPIGENETEC, Université Paris Descartes, Université Sorbonne Paris Cité, Paris, France
| | - Thérèse Truong
- INSERM, Center for Research in Epidemiology and Population Health (CESP), Cancer and Environment team, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Pascal Guénel
- INSERM, Center for Research in Epidemiology and Population Health (CESP), Cancer and Environment team, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
31
|
Sun R, Hui S, Bader GD, Lin X, Kraft P. Powerful gene set analysis in GWAS with the Generalized Berk-Jones statistic. PLoS Genet 2019; 15:e1007530. [PMID: 30875371 PMCID: PMC6436759 DOI: 10.1371/journal.pgen.1007530] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 03/27/2019] [Accepted: 02/28/2019] [Indexed: 11/19/2022] Open
Abstract
A common complementary strategy in Genome-Wide Association Studies (GWAS) is to perform Gene Set Analysis (GSA), which tests for the association between one phenotype of interest and an entire set of Single Nucleotide Polymorphisms (SNPs) residing in selected genes. While there exist many tools for performing GSA, popular methods often include a number of ad-hoc steps that are difficult to justify statistically, provide complicated interpretations based on permutation inference, and demonstrate poor operating characteristics. Additionally, the lack of gold standard gene set lists can produce misleading results and create difficulties in comparing analyses even across the same phenotype. We introduce the Generalized Berk-Jones (GBJ) statistic for GSA, a permutation-free parametric framework that offers asymptotic power guarantees in certain set-based testing settings. To adjust for confounding introduced by different gene set lists, we further develop a GBJ step-down inference technique that can discriminate between gene sets driven to significance by single genes and those demonstrating group-level effects. We compare GBJ to popular alternatives through simulation and re-analysis of summary statistics from a large breast cancer GWAS, and we show how GBJ can increase power by incorporating information from multiple signals in the same gene. In addition, we illustrate how breast cancer pathway analysis can be confounded by the frequency of FGFR2 in pathway lists. Our approach is further validated on two other datasets of summary statistics generated from GWAS of height and schizophrenia.
Collapse
Affiliation(s)
- Ryan Sun
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Shirley Hui
- The Donnelly Center, University of Toronto, Toronto, Ontario, Canada
| | - Gary D. Bader
- The Donnelly Center, University of Toronto, Toronto, Ontario, Canada
| | - Xihong Lin
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Peter Kraft
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| |
Collapse
|
32
|
Wendeu-Foyet MG, Koudou Y, Cénée S, Trétarre B, Rébillard X, Cancel-Tassin G, Cussenot O, Boland A, Bacq D, Deleuze JF, Lamy PJ, Mulot C, Laurent-Puig P, Truong T, Menegaux F. Circadian genes and risk of prostate cancer: Findings from the EPICAP study. Int J Cancer 2019; 145:1745-1753. [PMID: 30665264 DOI: 10.1002/ijc.32149] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 12/18/2018] [Indexed: 12/12/2022]
Abstract
Circadian rhythms regulate several physiological functions and genes controlling the circadian rhythm were found to regulate cell proliferation, cell cycle and apoptosis. Few studies have investigated the role of those circadian genes in prostate cancer occurrence. We aim to investigate the relationship between circadian genes polymorphisms and prostate cancer risk based on data from the EPICAP study, a population-based case-control study including 1,515 men (732 cases / 783 controls) with genotyped data. Odds Ratios (ORs) for association between prostate cancer and circadian gene variants were estimated for each of the 872 single nucleotide polymorphisms (SNPs) in 31 circadian clock genes. We also used a gene-based and pathway-based approach with a focus on the pathway including 9 core circadian genes. Separate analyses were conducted by prostate cancer aggressiveness. The core-circadian pathway (p = 0.0006) was significantly associated to prostate cancer, for either low (p = 0.002) or high (p = 0.01) grade tumor. At the gene level, we observed significant associations between all prostate cancer and NPAS2 and PER1 after correcting for multiple testing, while only RORA was significant for aggressive tumors. At the SNP-level, no significant association was observed. Our findings provide additional evidence of a potential link between genetic variants in circadian genes and prostate cancer risk. Further investigation is warranted to confirm these findings and to better understand the biological pathways involved.
Collapse
Affiliation(s)
- Méyomo G Wendeu-Foyet
- Université Paris-Saclay, Université Paris-Sud, CESP (Center for Research in Epidemiology and Population Health), Inserm, Team Cancer and Environment, Villejuif, France
| | - Yves Koudou
- Université Paris-Saclay, Université Paris-Sud, CESP (Center for Research in Epidemiology and Population Health), Inserm, Team Cancer and Environment, Villejuif, France
| | - Sylvie Cénée
- Université Paris-Saclay, Université Paris-Sud, CESP (Center for Research in Epidemiology and Population Health), Inserm, Team Cancer and Environment, Villejuif, France
| | | | | | - Géraldine Cancel-Tassin
- CeRePP, Hopital Tenon, Paris, France.,Sorbonne Université, GRC n°5, ONCOTYPE-URO, AP-HP, Hôpital Tenon, Paris
| | - Olivier Cussenot
- CeRePP, Hopital Tenon, Paris, France.,Sorbonne Université, GRC n°5, ONCOTYPE-URO, AP-HP, Hôpital Tenon, Paris
| | - Anne Boland
- Centre National de Recherche en Génomique Humaine (CNRGH), Institut de Biologie François Jacob, CEA, Université Paris-Saclay, Evry, France
| | - Delphine Bacq
- Centre National de Recherche en Génomique Humaine (CNRGH), Institut de Biologie François Jacob, CEA, Université Paris-Saclay, Evry, France
| | - Jean-François Deleuze
- Centre National de Recherche en Génomique Humaine (CNRGH), Institut de Biologie François Jacob, CEA, Université Paris-Saclay, Evry, France
| | - Pierre-Jean Lamy
- Clinique Beau Soleil, Montpellier, France.,Imagenome, Labosud, Montpellier, France
| | - Claire Mulot
- Université Paris Descartes, INSERM UMR-S1147 EPIGENETEC, Paris, France
| | | | - Thérèse Truong
- Université Paris-Saclay, Université Paris-Sud, CESP (Center for Research in Epidemiology and Population Health), Inserm, Team Cancer and Environment, Villejuif, France
| | - Florence Menegaux
- Université Paris-Saclay, Université Paris-Sud, CESP (Center for Research in Epidemiology and Population Health), Inserm, Team Cancer and Environment, Villejuif, France
| |
Collapse
|
33
|
Chien LC. A method for combining p-values in meta-analysis by gamma distributions. J Appl Stat 2019. [DOI: 10.1080/02664763.2018.1474857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Affiliation(s)
- Li-Chu Chien
- Center for Fundamental Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
34
|
Cai M, Li L. rPCMP: robust p-value combination by multiple partitions with applications to ATAC-seq data. BMC SYSTEMS BIOLOGY 2018; 12:141. [PMID: 30598086 PMCID: PMC6311921 DOI: 10.1186/s12918-018-0661-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND Evaluating the significance for a group of genes or proteins in a pathway or biological process for a disease could help researchers understand the mechanism of the disease. For example, identifying related pathways or gene functions for chromatin states of tumor-specific T cells will help determine whether T cells could reprogram or not, and further help design the cancer treatment strategy. Some existing p-value combination methods can be used in this scenario. However, these methods suffer from different disadvantages, and thus it is still challenging to design more powerful and robust statistical method. RESULTS The existing method of Group combined p-value (GCP) first partitions p-values to several groups using a set of several truncation points, but the method is often sensitive to these truncation points. Another method of adaptive rank truncated product method(ARTP) makes use of multiple truncation integers to adaptively combine the smallest p-values, but the method loses statistical power since it ignores the larger p-values. To tackle these problems, we propose a robust p-value combination method (rPCMP) by considering multiple partitions of p-values with different sets of truncation points. The proposed rPCMP statistic have a three-layer hierarchical structure. The inner-layer considers a statistic which combines p-values in a specified interval defined by two thresholds points, the intermediate-layer uses a GCP statistic which optimizes the statistic from the inner layer for a partition set of threshold points, and the outer-layer integrates the GCP statistic from multiple partitions of p-values. The empirical distribution of statistic under null distribution could be estimated by permutation procedure. CONCLUSIONS Our proposed rPCMP method has been shown to be more robust and have higher statistical power. Simulation study shows that our method can effectively control the type I error rates and have higher statistical power than the existing methods. We finally apply our rPCMP method to an ATAC-seq dataset for discovering the related gene functions with chromatin states in mouse tumors T cell.
Collapse
Affiliation(s)
- Menglan Cai
- School of Mathematics and Statistics, Xi'an Jiaotong University, Xianning West 28, Xi'an, China
| | - Limin Li
- School of Mathematics and Statistics, Xi'an Jiaotong University, Xianning West 28, Xi'an, China.
| |
Collapse
|
35
|
Benna C, Rajendran S, Spiro G, Tropea S, Del Fiore P, Rossi CR, Mocellin S. Associations of clock genes polymorphisms with soft tissue sarcoma susceptibility and prognosis. J Transl Med 2018; 16:338. [PMID: 30518396 PMCID: PMC6280400 DOI: 10.1186/s12967-018-1715-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 11/30/2018] [Indexed: 12/28/2022] Open
Abstract
Background Dysfunction of the circadian clock and polymorphisms of some circadian genes have been linked to cancer development and progression. We investigated the relationship between circadian genes germline variation and susceptibility or prognosis of patients with soft tissue sarcoma. Patients and methods We considered the 14 single nucleotide polymorphisms (SNPs) of 6 core circadian genes that have a minor allele frequency > 5% and that are known to be associated with cancer risk or prognosis. Genotyping was performed by q-PCR. Peripheral blood and clinic-pathological data were available for 162 patients with liposarcoma or leiomyosarcoma and 610 healthy donors. Associations between the selected clock genes polymorphisms and sarcoma susceptibility or prognosis were tested assuming 3 models of inheritance: additive, recessive and dominant. Subgroup analysis based on sarcoma histotype was performed under the additive genetic model. Multivariate logistic regression and multivariate Cox proportional hazard regression analyses were utilized to assess the association between SNPs with patient susceptibility and survival, respectively. Pathway variation analysis was conducted employing the Adaptive Rank Truncated Product method. Results Six out of the 14 analyzed SNPs were statistically significantly associated with susceptibility or prognosis of soft tissue sarcoma (P < 0.05). The present analysis suggested that carriers of the minor allele of the CLOCK polymorphism rs1801260 (C) or of PER2 rs934945 (T) had a reduced predisposition to sarcoma (26% and 35% respectively with the additive model) and liposarcoma (33% and 41% respectively). The minor allele (A) of NPAS2 rs895520 was associated with an increased predisposition to sarcoma of 33% and leiomyosarcoma of 44%. RORA rs339972 C allele was associated with a decreased predisposition to develop sarcoma assuming an additive model (29%) and leiomyosarcoma (36%). PER1 rs3027178 was associated with a reduced predisposition only in liposarcoma subgroup (32%). rs7602358 located upstream PER2 was significantly associated with liposarcoma survival (HR: 1.98; 95% CI 1.02–3.85; P = 0.04). Germline genetic variation in the circadian pathway was associated with the risk of developing soft tissue sarcoma (P = 0.035). Conclusions Genetic variation of circadian genes appears to play a role in the determinism of patient susceptibility and prognosis. These findings prompt further studies to fully dissect the molecular mechanisms. Electronic supplementary material The online version of this article (10.1186/s12967-018-1715-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Clara Benna
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padua, Italy. .,Clinica Chirurgica I, Azienda Ospedaliera Padova, Padua, Italy.
| | | | - Giovanna Spiro
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Saveria Tropea
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padua, Italy.,Surgical Oncology Unit, Istituto Oncologico Veneto (IOV-IRCCS), Padua, Italy
| | - Paolo Del Fiore
- Surgical Oncology Unit, Istituto Oncologico Veneto (IOV-IRCCS), Padua, Italy
| | - Carlo Riccardo Rossi
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padua, Italy.,Surgical Oncology Unit, Istituto Oncologico Veneto (IOV-IRCCS), Padua, Italy
| | - Simone Mocellin
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padua, Italy.,Surgical Oncology Unit, Istituto Oncologico Veneto (IOV-IRCCS), Padua, Italy
| |
Collapse
|
36
|
Heller R, Chatterjee N, Krieger A, Shi J. Post-Selection Inference Following Aggregate Level Hypothesis Testing in Large-Scale Genomic Data. J Am Stat Assoc 2018. [DOI: 10.1080/01621459.2017.1375933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Ruth Heller
- Department of Statistics and Operations Research, Tel-Aviv University, Tel-Aviv, Israel
- Biostatistics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD
| | - Nilanjan Chatterjee
- Department of Biostatistics, Bloomberg School of Public Health, and Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, MD
| | - Abba Krieger
- Department of Statistics, University of Pennsylvania, Philadelphia, PA
| | - Jianxin Shi
- Biostatistics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD
| |
Collapse
|
37
|
Wang S, Huo D, Ogundiran TO, Ojengbede O, Zheng W, Nathanson KL, Nemesure B, Ambs S, Olopade OI, Zheng Y. Genetic variation in the Hippo pathway and breast cancer risk in women of African ancestry. Mol Carcinog 2018; 57:1311-1318. [PMID: 29873413 PMCID: PMC6662580 DOI: 10.1002/mc.22845] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 05/18/2018] [Accepted: 06/01/2018] [Indexed: 12/14/2022]
Abstract
Gene expression changes within the Hippo pathway were found to be associated with large tumor size and metastasis in breast cancer. The combined effect of genetic variants in genes of this pathway may have a causal role in breast cancer development. We examined 7086 SNPs that were not highly correlated (r2 < 0.8) in 35 Hippo pathway genes using data from the genome-wide association study of breast cancer from the Root Consortium, which includes 3686 participants of African ancestry from Nigeria, United States of America, and Barbados: 1657 cases (403 estrogen receptor-positive [ER+], 374 ER-) and 2029 controls. Gene-level analyses were conducted using improved AdaJoint test for large-scale genetic association studies adjusting for age, study site and the first four eigenvectors from the principal component analysis. SNP-level analyses were conducted with logistic regression. The Hippo pathway was significantly associated with risk of ER+ breast cancer (pathway-level P = 0.019), with WWC1 (Padj = 0.04) being the leading gene. The pathway-level significance was lost without WWC1 (P = 0.12). rs147106204 in the WWC1 gene was the most statistically significant SNP after gene-level adjustment for multiple comparisons (OR = 0.53, 95%CI = 0.41-0.70, Padj = 0.025). We found evidence of an association between genetic variations in the Hippo pathway and ER+ breast cancer. Moreover, WWC1 was identified as the most important genetic susceptibility locus highlighting the importance of genetic epidemiology studies of breast cancer in understudied populations.
Collapse
Affiliation(s)
- Shengfeng Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
- Center for Clinical Cancer Genetics and Global Health, Department of Medicine, University of Chicago, Chicago, Illinois; USA
| | - Dezheng Huo
- Department of Public Health Sciences, University of Chicago, Chicago, Illinois, USA
| | | | - Oladosu Ojengbede
- Center for Population and Reproductive Health, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee, USA
| | | | - Barbara Nemesure
- Department of Preventive Medicine, State University of New York at Stony Brook, Stony Brook, New York, USA
| | - Stefan Ambs
- Laboratory of Human Carcinogenesis, National Cancer Institute, Bethesda, Maryland, USA
| | - Olufunmilayo I. Olopade
- Center for Clinical Cancer Genetics and Global Health, Department of Medicine, University of Chicago, Chicago, Illinois; USA
| | - Yonglan Zheng
- Center for Clinical Cancer Genetics and Global Health, Department of Medicine, University of Chicago, Chicago, Illinois; USA
| |
Collapse
|
38
|
Wang X, Boekstegers F, Brinster R. Methods and results from the genome-wide association group at GAW20. BMC Genet 2018; 19:79. [PMID: 30255814 PMCID: PMC6157187 DOI: 10.1186/s12863-018-0649-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND This paper summarizes the contributions from the Genome-wide Association Study group (GWAS group) of the GAW20. The GWAS group contributions focused on topics such as association tests, phenotype imputation, and application of empirical kinships. The goals of the GWAS group contributions were varied. A real or a simulated data set based on the Genetics of Lipid Lowering Drugs and Diet Network (GOLDN) study was employed by different methods. Different outcomes and covariates were considered, and quality control procedures varied throughout the contributions. RESULTS The consideration of heritability and family structure played a major role in some contributions. The inclusion of family information and adaptive weights based on data were found to improve power in genome-wide association studies. It was proven that gene-level approaches are more powerful than single-marker analysis. Other contributions focused on the comparison between pedigree-based kinship and empirical kinship matrices, and investigated similar results in heritability estimation, association mapping, and genomic prediction. A new approach for linkage mapping of triglyceride levels was able to identify a novel linkage signal. CONCLUSIONS This summary paper reports on promising statistical approaches and findings of the members of the GWAS group applied on real and simulated data which encompass the current topics of epigenetic and pharmacogenomics.
Collapse
Affiliation(s)
- Xuexia Wang
- University of North Texas, GAB 459, 1155 Union Circle #311430, Denton, TX 76203 USA
| | - Felix Boekstegers
- Institute of Medical Biometry and Informatics, University of Heidelberg, Im Neuenheimer Feld 130.3, 69120 Heidelberg, Germany
| | - Regina Brinster
- Institute of Medical Biometry and Informatics, University of Heidelberg, Im Neuenheimer Feld 130.3, 69120 Heidelberg, Germany
| |
Collapse
|
39
|
Cologne J, Loo L, Shvetsov YB, Misumi M, Lin P, Haiman CA, Wilkens LR, Le Marchand L. Stepwise approach to SNP-set analysis illustrated with the Metabochip and colorectal cancer in Japanese Americans of the Multiethnic Cohort. BMC Genomics 2018; 19:524. [PMID: 29986644 PMCID: PMC6038257 DOI: 10.1186/s12864-018-4910-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 06/29/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Common variants have explained less than the amount of heritability expected for complex diseases, which has led to interest in less-common variants and more powerful approaches to the analysis of whole-genome scans. Because of low frequency (low statistical power), less-common variants are best analyzed using SNP-set methods such as gene-set or pathway-based analyses. However, there is as yet no clear consensus regarding how to focus in on potential risk variants following set-based analyses. We used a stepwise, telescoping approach to analyze common- and rare-variant data from the Illumina Metabochip array to assess genomic association with colorectal cancer (CRC) in the Japanese sub-population of the Multiethnic Cohort (676 cases, 7180 controls). We started with pathway analysis of SNPs that are in genes and pathways having known mechanistic roles in colorectal cancer, then focused on genes within the pathways that evidenced association with CRC, and finally assessed individual SNPs within the genes that evidenced association. Pathway SNPs downloaded from the dbSNP database were cross-matched with Metabochip SNPs and analyzed using the logistic kernel machine regression approach (logistic SNP-set kernel-machine association test, or sequence kernel association test; SKAT) and related methods. RESULTS The TGF-β and WNT pathways were associated with all CRC, and the WNT pathway was associated with colon cancer. Individual genes demonstrating the strongest associations were TGFBR2 in the TGF-β pathway and SMAD7 (which is involved in both the TGF-β and WNT pathways). As partial validation of our approach, a known CRC risk variant in SMAD7 (in both the TGF-β and WNT pathways: rs11874392) was associated with CRC risk in our data. We also detected two novel candidate CRC risk variants (rs13075948 and rs17025857) in TGFBR2, a gene known to be associated with CRC risk. CONCLUSIONS A stepwise, telescoping approach identified some potentially novel risk variants associated with colorectal cancer, so it may be a useful method for following up on results of set-based SNP analyses. Further work is required to assess the statistical characteristics of the approach, and additional applications should aid in better clarifying its utility.
Collapse
Affiliation(s)
- John Cologne
- Department of Statistics, Radiation Effects Research Foundation, Hiroshima, 732-0815, Japan.
| | - Lenora Loo
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, 96813, USA
| | - Yurii B Shvetsov
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, 96813, USA
| | - Munechika Misumi
- Department of Statistics, Radiation Effects Research Foundation, Hiroshima, 732-0815, Japan
| | - Philip Lin
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, 96813, USA
| | - Christopher A Haiman
- Department of Preventive Medicine and Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Lynne R Wilkens
- Biostatistics and Informatics Shared Resource, University of Hawaii Cancer Center, Honolulu, HI, 96813, USA
| | - Loïc Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, 96813, USA
| |
Collapse
|
40
|
Zhang S, Zhu J, Li Z. Adaptive Group-combined P-values Test for Two-sample Location Problem with Applications to Microarray Data. Sci Rep 2018; 8:8117. [PMID: 29802271 PMCID: PMC5970252 DOI: 10.1038/s41598-018-26409-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 05/11/2018] [Indexed: 11/14/2022] Open
Abstract
The purpose of this article is to propose a test for two-sample location problem in high-dimensional data. In general highdimensional case, the data dimension can be much larger than the sample size and the underlying distribution may be far from normal. Existing tests requiring explicit relationship between the data dimension and sample size or designed for multivariate normal distributions may lose power significantly and even yield type I error rates strayed from nominal levels. To overcome this issue, we propose an adaptive group p-values combination test which is robust against both high dimensionality and normality. Simulation studies show that the proposed test controls type I error rates correctly and outperforms some existing tests in most situations. An Ageing Human Brain Microarray data are used to further exemplify the method.
Collapse
Affiliation(s)
- Shenghu Zhang
- School of Mathematics and Information Science, Jiangxi Normal University, Nanchang, 330022, China
- School of Mathematical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jiayan Zhu
- School of information engineering, Hubei University of Chinese Medicine, Wuhan, 430065, China
- School of Mathematics and Statistics & Hubei Key Laboratory of Mathematical Sciences, Central China Normal University, Wuhan, 430079, China
| | - Zhengbang Li
- School of Mathematics and Statistics & Hubei Key Laboratory of Mathematical Sciences, Central China Normal University, Wuhan, 430079, China.
| |
Collapse
|
41
|
Kim JE, Choi J, Park J, Park C, Lee SM, Park SE, Song N, Chung S, Sung H, Han W, Lee JW, Park SK, Kim MK, Noh DY, Yoo KY, Kang D, Choi JY. Associations between genetic polymorphisms of membrane transporter genes and prognosis after chemotherapy: meta-analysis and finding from Seoul Breast Cancer Study (SEBCS). THE PHARMACOGENOMICS JOURNAL 2018; 18:633-645. [PMID: 29618765 DOI: 10.1038/s41397-018-0016-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 10/13/2017] [Accepted: 12/04/2017] [Indexed: 12/30/2022]
Abstract
Membrane transporters can be major determinants of the pharmacokinetic profiles of anticancer drugs. The associations between genetic variations of ATP-binding cassette (ABC) and solute carrier (SLC) genes and cancer survival were investigated through a meta-analysis and an association study in the Seoul Breast Cancer Study (SEBCS). Including the SEBCS, the meta-analysis was conducted among 38 studies of genetic variations of transporters on various cancer survivors. The population of SEBCS consisted of 1338 breast cancer patients who had been treated with adjuvant chemotherapy. A total of 7750 SNPs were selected from 453 ABC and/or SLC genes typed by an Affymetrix 6.0 chip. ABCB1 rs1045642 was associated with poor progression-free survival in a meta-analysis (HR = 1.33, 95% CI: 1.07-1.64). ABCB1, SLC8A1, and SLC12A8 were associated with breast cancer survival in SEBCS (Pgene < 0.05). ABCB1 rs1202172 was differentially associated with survival depending on the chemotherapy (Pinteraction = 0.035). Our finding provides suggestive associations of membrane transporters on cancer survival.
Collapse
Affiliation(s)
- Ji-Eun Kim
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea
| | - Jaesung Choi
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea
| | - JooYong Park
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea
| | - Chulbum Park
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea
| | - Se Mi Lee
- College of Pharmacy Chonnam National University, Gwangju, Korea
| | - Seong Eun Park
- College of Pharmacy, Duksung Women's university, Seoul, Korea
| | - Nan Song
- Cancer Research Institute, Seoul National University, Seoul, Korea
| | - Seokang Chung
- Division for New Health Technology Assessment, National Evidence-based Healthcare Collaborating Agency, Seoul, Korea
| | - Hyuna Sung
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Wonshik Han
- Cancer Research Institute, Seoul National University, Seoul, Korea.,Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Jong Won Lee
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sue K Park
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea.,Cancer Research Institute, Seoul National University, Seoul, Korea.,Department of Preventive Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Mi Kyung Kim
- Division of Cancer Epidemiology and Management, National Cancer Center, Goyang, Korea
| | - Dong-Young Noh
- Cancer Research Institute, Seoul National University, Seoul, Korea.,Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Keun-Young Yoo
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul, Korea.,The Armed Forces Capital Hospital, Seongnam, Korea
| | - Daehee Kang
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea.,Cancer Research Institute, Seoul National University, Seoul, Korea.,Department of Preventive Medicine, Seoul National University College of Medicine, Seoul, Korea.,Institute of Environmental Medicine, Seoul National University Medical Research Center, Seoul, Korea
| | - Ji-Yeob Choi
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea. .,Cancer Research Institute, Seoul National University, Seoul, Korea. .,Department of Preventive Medicine, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
42
|
Chen L, Wang Y, Zhou Y. Association analysis of multiple traits by an approach of combining P values. J Genet 2018; 97:79-85. [PMID: 29666327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Increasing evidence shows that one variant can affect multiple traits, which is a widespread phenomenon in complex diseases. Joint analysis of multiple traits can increase statistical power of association analysis and uncover the underlying genetic mechanism. Although there are many statistical methods to analyse multiple traits, most of these methods are usually suitable for detecting common variants associated with multiple traits. However, because of low minor allele frequency of rare variant, these methods are not optimal for rare variant association analysis. In this paper, we extend an adaptive combination of P values method (termed ADA) for single trait to test association between multiple traits and rare variants in the given region. For a given region, we use reverse regression model to test each rare variant associated with multiple traits and obtain the P value of single-variant test. Further, we take the weighted combination of these P values as the test statistic. Extensive simulation studies show that our approach is more powerful than several other comparison methods in most cases and is robust to the inclusion of a high proportion of neutral variants and the different directions of effects of causal variants.
Collapse
Affiliation(s)
- Lili Chen
- Department of Mathematics, School of Sciences, Harbin Institute of Technology, Harbin 150001, People's Republic of China.
| | | | | |
Collapse
|
43
|
Mocellin S, Tropea S, Benna C, Rossi CR. Circadian pathway genetic variation and cancer risk: evidence from genome-wide association studies. BMC Med 2018; 16:20. [PMID: 29455641 PMCID: PMC5817863 DOI: 10.1186/s12916-018-1010-1] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 01/18/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Dysfunction of the circadian clock and single polymorphisms of some circadian genes have been linked to cancer susceptibility, although data are scarce and findings inconsistent. We aimed to investigate the association between circadian pathway genetic variation and risk of developing common cancers based on the findings of genome-wide association studies (GWASs). METHODS Single nucleotide polymorphisms (SNPs) of 17 circadian genes reported by three GWAS meta-analyses dedicated to breast (Discovery, Biology, and Risk of Inherited Variants in Breast Cancer (DRIVE) Consortium; cases, n = 15,748; controls, n = 18,084), prostate (Elucidating Loci Involved in Prostate Cancer Susceptibility (ELLIPSE) Consortium; cases, n = 14,160; controls, n = 12,724) and lung carcinoma (Transdisciplinary Research In Cancer of the Lung (TRICL) Consortium; cases, n = 12,160; controls, n = 16,838) in patients of European ancestry were utilized to perform pathway analysis by means of the adaptive rank truncated product (ARTP) method. Data were also available for the following subgroups: estrogen receptor negative breast cancer, aggressive prostate cancer, squamous lung carcinoma and lung adenocarcinoma. RESULTS We found a highly significant statistical association between circadian pathway genetic variation and the risk of breast (pathway P value = 1.9 × 10-6; top gene RORA, gene P value = 0.0003), prostate (pathway P value = 4.1 × 10-6; top gene ARNTL, gene P value = 0.0002) and lung cancer (pathway P value = 6.9 × 10-7; top gene RORA, gene P value = 2.0 × 10-6), as well as all their subgroups. Out of 17 genes investigated, 15 were found to be significantly associated with the risk of cancer: four genes were shared by all three malignancies (ARNTL, CLOCK, RORA and RORB), two by breast and lung cancer (CRY1 and CRY2) and three by prostate and lung cancer (NPAS2, NR1D1 and PER3), whereas four genes were specific for lung cancer (ARNTL2, CSNK1E, NR1D2 and PER2) and two for breast cancer (PER1, RORC). CONCLUSIONS Our findings, based on the largest series ever utilized for ARTP-based gene and pathway analysis, support the hypothesis that circadian pathway genetic variation is involved in cancer predisposition.
Collapse
Affiliation(s)
- Simone Mocellin
- Department of Surgery Oncology and Gastroenterology, University of Padova, Via Giustiniani 2, 35128, Padova, Italy. .,Istituto Oncologico Veneto, IOV-IRCCS, Padova, Italy.
| | | | - Clara Benna
- Department of Surgery Oncology and Gastroenterology, University of Padova, Via Giustiniani 2, 35128, Padova, Italy
| | - Carlo Riccardo Rossi
- Department of Surgery Oncology and Gastroenterology, University of Padova, Via Giustiniani 2, 35128, Padova, Italy.,Istituto Oncologico Veneto, IOV-IRCCS, Padova, Italy
| |
Collapse
|
44
|
Tan J, Fu L, Chen H, Guan J, Chen Y, Fang J. Association study of genetic variation in the autophagy lysosome pathway genes and risk of eight kinds of cancers. Int J Cancer 2018; 143:80-87. [PMID: 29388190 DOI: 10.1002/ijc.31288] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 12/20/2017] [Accepted: 01/11/2018] [Indexed: 12/20/2022]
Abstract
The autophagy lysosome pathway is essential to maintain cell viability and homeostasis in response to many stressful environments, which is reported to play a vital role in cancer development and therapy. However, the association of genetic alterations of this pathway with risk of cancer remains unclear. Based on genome-wide association study data of eight kinds of cancers, we used an adaptive rank truncated product approach to perform a pathway-level and gene-level analysis, and used a logistic model to calculate SNP-level associations to examine whether an altered autophagy lysosome pathway contributes to cancer susceptibility. Among eight kinds of cancers, four of them showed significant statistics in the pathway-level analysis, including breast cancer (p = 0.00705), gastric cancer (p = 0.00880), lung cancer (p = 0.000100) and renal cell carcinoma (p = 0.00190). We also found that some autophagy lysosome genes had signals of association with cancer risk. Our results demonstrated that inherited genetic variants in the overall autophagy lysosome pathway and certain associated genes might contribute to cancer susceptibility, which warrant further evaluation in other independent datasets.
Collapse
Affiliation(s)
- Juan Tan
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, 145 Middle Shandong Road, Shanghai 200001, China
| | - Linna Fu
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, 145 Middle Shandong Road, Shanghai 200001, China
| | - Haoyan Chen
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, 145 Middle Shandong Road, Shanghai 200001, China
| | - Jian Guan
- Department of Otolaryngology Head and Neck Surgery & Center of Sleep Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Otolaryngological Institute of Shanghai Jiao Tong University, Shanghai, China
| | - Yingxuan Chen
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, 145 Middle Shandong Road, Shanghai 200001, China
| | - Jingyuan Fang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, 145 Middle Shandong Road, Shanghai 200001, China
| |
Collapse
|
45
|
Chen L, Wang Y, Zhou Y. Association analysis of multiple traits by an approach of combining
$$P$$
P
values. J Genet 2018. [DOI: 10.1007/s12041-018-0885-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
46
|
Fu LN, Tan J, Chen YX, Fang JY. Genetic variants in the histone methylation and acetylation pathway and their risks in eight types of cancers. J Dig Dis 2018; 19:102-111. [PMID: 29292860 DOI: 10.1111/1751-2980.12574] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 05/16/2017] [Accepted: 12/29/2017] [Indexed: 12/11/2022]
Abstract
OBJECTIVES The histone methylation and acetylation pathway genes regulate cell growth and survival. Aberrations in this pathway are implicated in a variety of cancers. This study aimed to identify germline genetic variants in histone methylation and acetylation pathway genes that may contribute to risk in eight types of cancers and to explore the relation between the whole pathway and their risks in these types of cancers. METHODS Germline genetic variants in 89 genes in the histone methylation and acetylation pathway were explored. Gene-based and pathway-based associations with eight types of cancers were analyzed using logistic regression models and the permutation-based adaptive rank-truncated product method, respectively. RESULTS Gene-level associations revealed that genetic variants in 45 genes were significantly associated with the risk of cancer. The total histone methylation and acetylation pathway was significantly associated with the risk of esophageal squamous cell carcinoma (P = 0.0492) and prostate (P = 0.0038), lung (P = 0.00015), and bladder cancer (P = 0.00135), but not with breast (P = 0.182), pancreatic (P = 0.336) and gastric cancer (P = 0.347) and renal cell carcinoma (P =0.828). CONCLUSIONS Our study suggested there is an association between germline genetic variation at the overall histone methylation and acetylation pathway level and some individual genes with cancer risk. Further studies are needed to validate these relations and to explore relative mechanisms.
Collapse
Affiliation(s)
- Lin Na Fu
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Juan Tan
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Ying Xuan Chen
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Jing-Yuan Fang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| |
Collapse
|
47
|
Wang Y, Wu W, Zhu M, Wang C, Shen W, Cheng Y, Geng L, Li Z, Zhang J, Dai J, Ma H, Chen L, Hu Z, Jin G, Shen H. Integrating expression-related SNPs into genome-wide gene- and pathway-based analyses identified novel lung cancer susceptibility genes. Int J Cancer 2017; 142:1602-1610. [DOI: 10.1002/ijc.31182] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 11/22/2017] [Accepted: 11/23/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Yuzhuo Wang
- Department of Epidemiology and Biostatistics, School of Public Health; Nanjing Medical University; Nanjing 211166 China
| | - Weibing Wu
- Department of Thoracic Surgery; First Affiliated Hospital of Nanjing Medical University; Nanjing 210029 China
| | - Meng Zhu
- Department of Epidemiology and Biostatistics, School of Public Health; Nanjing Medical University; Nanjing 211166 China
| | - Cheng Wang
- Department of Epidemiology and Biostatistics, School of Public Health; Nanjing Medical University; Nanjing 211166 China
| | - Wei Shen
- Department of Epidemiology and Biostatistics, School of Public Health; Nanjing Medical University; Nanjing 211166 China
| | - Yang Cheng
- Department of Epidemiology and Biostatistics, School of Public Health; Nanjing Medical University; Nanjing 211166 China
| | - Liguo Geng
- Department of Epidemiology and Biostatistics, School of Public Health; Nanjing Medical University; Nanjing 211166 China
| | - Zhihua Li
- Department of Epidemiology and Biostatistics, School of Public Health; Nanjing Medical University; Nanjing 211166 China
| | - Jiahui Zhang
- Department of Epidemiology and Biostatistics, School of Public Health; Nanjing Medical University; Nanjing 211166 China
| | - Juncheng Dai
- Department of Epidemiology and Biostatistics, School of Public Health; Nanjing Medical University; Nanjing 211166 China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment; Collaborative Innovation Center of Cancer Medicine, Nanjing Medical University; Nanjing 211166 China
| | - Hongxia Ma
- Department of Epidemiology and Biostatistics, School of Public Health; Nanjing Medical University; Nanjing 211166 China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment; Collaborative Innovation Center of Cancer Medicine, Nanjing Medical University; Nanjing 211166 China
| | - Liang Chen
- Department of Thoracic Surgery; First Affiliated Hospital of Nanjing Medical University; Nanjing 210029 China
| | - Zhibin Hu
- Department of Epidemiology and Biostatistics, School of Public Health; Nanjing Medical University; Nanjing 211166 China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment; Collaborative Innovation Center of Cancer Medicine, Nanjing Medical University; Nanjing 211166 China
| | - Guangfu Jin
- Department of Epidemiology and Biostatistics, School of Public Health; Nanjing Medical University; Nanjing 211166 China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment; Collaborative Innovation Center of Cancer Medicine, Nanjing Medical University; Nanjing 211166 China
| | - Hongbing Shen
- Department of Epidemiology and Biostatistics, School of Public Health; Nanjing Medical University; Nanjing 211166 China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment; Collaborative Innovation Center of Cancer Medicine, Nanjing Medical University; Nanjing 211166 China
| |
Collapse
|
48
|
Cirillo E, Parnell LD, Evelo CT. A Review of Pathway-Based Analysis Tools That Visualize Genetic Variants. Front Genet 2017; 8:174. [PMID: 29163640 PMCID: PMC5681904 DOI: 10.3389/fgene.2017.00174] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 10/24/2017] [Indexed: 01/04/2023] Open
Abstract
Pathway analysis is a powerful method for data analysis in genomics, most often applied to gene expression analysis. It is also promising for single-nucleotide polymorphism (SNP) data analysis, such as genome-wide association study data, because it allows the interpretation of variants with respect to the biological processes in which the affected genes and proteins are involved. Such analyses support an interactive evaluation of the possible effects of variations on function, regulation or interaction of gene products. Current pathway analysis software often does not support data visualization of variants in pathways as an alternate method to interpret genetic association results, and specific statistical methods for pathway analysis of SNP data are not combined with these visualization features. In this review, we first describe the visualization options of the tools that were identified by a literature review, in order to provide insight for improvements in this developing field. Tool evaluation was performed using a computational epistatic dataset of gene–gene interactions for obesity risk. Next, we report the necessity to include in these tools statistical methods designed for the pathway-based analysis with SNP data, expressly aiming to define features for more comprehensive pathway-based analysis tools. We conclude by recognizing that pathway analysis of genetic variations data requires a sophisticated combination of the most useful and informative visual aspects of the various tools evaluated.
Collapse
Affiliation(s)
- Elisa Cirillo
- Department of Bioinformatics - BiGCaT, Maastricht University, Maastricht, Netherlands
| | - Laurence D Parnell
- Jean Mayer-USDA Human Nutrition Research Center on Aging at Tufts University, Agricultural Research Service, USDA, Boston, MA, United States
| | - Chris T Evelo
- Department of Bioinformatics - BiGCaT, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
49
|
Adaptive combination of Bayes factors as a powerful method for the joint analysis of rare and common variants. Sci Rep 2017; 7:13858. [PMID: 29066733 PMCID: PMC5654754 DOI: 10.1038/s41598-017-13177-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 09/21/2017] [Indexed: 11/30/2022] Open
Abstract
Multi-marker association tests can be more powerful than single-locus analyses because they aggregate the variant information within a gene/region. However, combining the association signals of multiple markers within a gene/region may cause noise due to the inclusion of neutral variants, which usually compromises the power of a test. To reduce noise, the “adaptive combination of P-values” (ADA) method removes variants with larger P-values. However, when both rare and common variants are considered, it is not optimal to truncate variants according to their P-values. An alternative summary measure, the Bayes factor (BF), is defined as the ratio of the probability of the data under the alternative hypothesis to that under the null hypothesis. The BF quantifies the “relative” evidence supporting the alternative hypothesis. Here, we propose an “adaptive combination of Bayes factors” (ADABF) method that can be directly applied to variants with a wide spectrum of minor allele frequencies. The simulations show that ADABF is more powerful than single-nucleotide polymorphism (SNP)-set kernel association tests and burden tests. We also analyzed 1,109 case-parent trios from the Schizophrenia Trio Genomic Research in Taiwan. Three genes on chromosome 19p13.2 were found to be associated with schizophrenia at the suggestive significance level of 5 × 10−5.
Collapse
|
50
|
Wang S, Huo D, Ogundiran TO, Ojengbede O, Zheng W, Nathanson KL, Nemesure B, Ambs S, Olopade OI, Zheng Y. Association of breast cancer risk and the mTOR pathway in women of African ancestry in 'The Root' Consortium. Carcinogenesis 2017; 38:789-796. [PMID: 28582508 DOI: 10.1093/carcin/bgx055] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 06/02/2017] [Indexed: 12/16/2022] Open
Abstract
Functional studies have elucidated the role of the mammalian target of rapamycin (mTOR) pathway in breast carcinogenesis, but to date, there is a paucity of data on its contribution to breast cancer risk in women of African ancestry. We examined 47628 SNPs in 61 mTOR pathway genes in the genome wide association study of breast cancer in the African Diaspora study (The Root consortium), which included 3686 participants (1657 cases). Pathway- and gene-level analyses were conducted using the adaptive rank truncated product (ARTP) test for 10994 SNPs that were not highly correlated (r2 < 0.8). Odds ratio (OR) and 95% confidence interval (CI) were estimated with logistic regression for each single-nucleotide polymorphism. The mTOR pathway was significantly associated with overall and estrogen receptor-negative (ER-) breast cancer risk (P = 0.003 and 0.03, respectively). PRKAG3 (Padj = 0.0018) and RPS6KA3 (Padj = 0.061) were the leading genes for the associations with overall breast cancer risk and ER- breast cancer risk, respectively. rs190843378 in PRKAG3 was statistically significant after gene-level adjustment for multiple comparisons (OR = 0.50 for each T allele, 95% CI = 0.38-0.66, Padj = 3.6E-05), with a statistical power of 0.914. These results provide new insights on the biological relevance of the mTOR pathway in breast cancer progression and underscore the need for more genetic epidemiology studies of breast cancer in the African Diaspora.
Collapse
Affiliation(s)
- Shengfeng Wang
- Department of Medicine, Center for Clinical Cancer Genetics and Global Health, University of Chicago, Chicago, IL, USA
| | - Dezheng Huo
- Department of Public Health Sciences, University of Chicago, Chicago, IL, USA
| | - Temidayo O Ogundiran
- Department of Surgery, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Oladosu Ojengbede
- Center for Population and Reproductive Health, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN, USA
| | | | - Barbara Nemesure
- Department of Preventive Medicine, State University of New York at Stony Brook, Stony Brook, NY, USA
| | - Stefan Ambs
- Laboratory of Human Carcinogenesis, National Cancer Institute, Bethesda, MD, USA
| | | | - Yonglan Zheng
- To whom correspondence should be addressed. Tel: +1 773 702 1632; Fax: +1 773 834 1659;
| |
Collapse
|