1
|
Joukhadar R, Trethowan RM, Thistlethwaite R, Hayden MJ, Stangoulis J, Cu S, Tibbits J, Daetwyler HD. Stable pleotropic loci controlling the accumulation of multiple nutritional elements in wheat. TAG. THEORETICAL AND APPLIED GENETICS. THEORETISCHE UND ANGEWANDTE GENETIK 2025; 138:95. [PMID: 40205176 PMCID: PMC11982167 DOI: 10.1007/s00122-025-04877-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 03/08/2025] [Indexed: 04/11/2025]
Abstract
Understanding the genetic basis of nutrient accumulation in wheat is crucial for improving its nutritional content and addressing global food security challenges. Here, we identified stable pleiotropic loci controlling the accumulation of 13 nutritional elements in wheat across diverse environments using a large wheat population of 1470 individuals. Our analysis revealed significant variability in SNP-based heritability values across 13 essential elements. Genetic correlations among elements uncovered complex relations, with positive correlations observed within two distinct groups, where calcium (Ca), cobalt (Co), potassium (K), and sodium (Na) formed one group, and copper (Cu), iron (Fe), magnesium (Mg), manganese (Mn), molybdenum (Mo), nickel (Ni), phosphorus (P), and zinc (Zn) formed the other. Negative correlations were observed among elements across both groups. Through MetaGWAS analysis, we identified stable QTL associated with individual elements and elements with high positive correlations. We identified 67 stable QTL across environments that are independent from grain yield, of which 56 were detected using the MetaGWAS analysis indicating their pleiotropic effect on multiple elements. A major QTL on chromosome 7D that can shift the phenotype up to one standard deviation compared to the mean phenotype in the population exhibited differential effects on multiple elements belonging to both groups. Our findings offer novel insights into the genetic architecture of nutrient accumulation in wheat and have practical implications for breeding programmes aimed at enhancing multiple nutrients simultaneously. By targeting stable QTL, breeders can develop wheat varieties with improved nutritional profiles, contributing to global food security and human health.
Collapse
Affiliation(s)
- Reem Joukhadar
- Agriculture Victoria, Centre for AgriBioscience, AgriBio, Bundoora, VIC, Australia.
| | - Richard M Trethowan
- School of Life and Environmental Sciences, Plant Breeding Institute, Sydney Institute of Agriculture, The University of Sydney, Narrabri, NSW, Australia.
- School of Life and Environmental Sciences, Plant Breeding Institute, Sydney Institute of Agriculture, The University of Sydney, Cobbitty, NSW, Australia.
| | - Rebecca Thistlethwaite
- School of Life and Environmental Sciences, Plant Breeding Institute, Sydney Institute of Agriculture, The University of Sydney, Narrabri, NSW, Australia
| | - Matthew J Hayden
- Agriculture Victoria, Centre for AgriBioscience, AgriBio, Bundoora, VIC, Australia
- School of Applied Systems Biology, La Trobe University, Bundoora, VIC, Australia
| | - James Stangoulis
- College of Science and Engineering, Flinders University, Sturt Road, Bedford Park, South Australia, 5042, Australia
| | - Suong Cu
- College of Science and Engineering, Flinders University, Sturt Road, Bedford Park, South Australia, 5042, Australia
| | - Josquin Tibbits
- Agriculture Victoria, Centre for AgriBioscience, AgriBio, Bundoora, VIC, Australia
| | - Hans D Daetwyler
- Agriculture Victoria, Centre for AgriBioscience, AgriBio, Bundoora, VIC, Australia
| |
Collapse
|
2
|
Li Y, Dang X, Chen R, Teng Z, Wang J, Li S, Yue Y, Mitchell BL, Zeng Y, Yao YG, Li M, Liu Z, Yuan Y, Li T, Zhang Z, Luo XJ. Cross-ancestry genome-wide association study and systems-level integrative analyses implicate new risk genes and therapeutic targets for depression. Nat Hum Behav 2025; 9:806-823. [PMID: 39994458 DOI: 10.1038/s41562-024-02073-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 10/23/2024] [Indexed: 02/26/2025]
Abstract
Deciphering the genetic architecture of depression is pivotal for characterizing the associated pathophysiological processes and development of new therapeutics. Here we conducted a cross-ancestry genome-wide meta-analysis on depression (416,437 cases and 1,308,758 controls) and identified 287 risk loci, of which 49 are new. Variant-level fine mapping prioritized potential causal variants and functional genomic analysis identified variants that regulate the binding of transcription factors. We validated that 80% of the identified functional variants are regulatory variants, and expression quantitative trait loci analysis uncovered the potential target genes regulated by the prioritized risk variants. Gene-level analysis, including transcriptome and proteome-wide association studies, colocalization and Mendelian randomization-based analyses, prioritized potential causal genes and drug targets. Gene prioritization analyses highlighted likely causal genes, including TMEM106B, CTNND1, AREL1 and so on. Pathway analysis indicated significant enrichment of depression risk genes in synapse-related pathways. Finally, knockdown of Tmem106b in mice resulted in depression-like behaviours, supporting the involvement of Tmem106b in depression. Our study identified new risk loci, likely causal variants and genes for depression, providing important insights into the genetic architecture of depression and potential therapeutic targets.
Collapse
Affiliation(s)
- Yifan Li
- Department of Psychiatry and Psychosomatics, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Southeast University, Nanjing, China
| | - Xinglun Dang
- Department of Psychiatry and Psychosomatics, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Southeast University, Nanjing, China
| | - Rui Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Zhaowei Teng
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Yunnan Provincial Department of Education Gut Microbiota Transplantation Engineering Research Center, Kunming, China
| | - Junyang Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Shiwu Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Yingying Yue
- Department of Psychiatry and Psychosomatics, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Southeast University, Nanjing, China
| | - Brittany L Mitchell
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Yong Zeng
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Yunnan Provincial Department of Education Gut Microbiota Transplantation Engineering Research Center, Kunming, China
| | - Yong-Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Zhongchun Liu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Yonggui Yuan
- Department of Psychiatry and Psychosomatics, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Southeast University, Nanjing, China.
| | - Tao Li
- Affiliated Mental Health Center, Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Zhijun Zhang
- Department of Psychiatry and Psychosomatics, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Southeast University, Nanjing, China.
- Department of Mental Health and Public Health, Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| | - Xiong-Jian Luo
- Department of Psychiatry and Psychosomatics, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Southeast University, Nanjing, China.
| |
Collapse
|
3
|
Cai X, Zhang W, Gao N, Wei C, Wu X, Si J, Gao Y, Li J, Yin T, Zhang Z. Integrating large-scale meta-analysis of genome-wide association studies improve the genomic prediction accuracy for combined pig populations. J Anim Breed Genet 2025; 142:223-236. [PMID: 39215551 DOI: 10.1111/jbg.12896] [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: 12/26/2023] [Revised: 07/18/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024]
Abstract
The strategy of combining reference populations has been widely recognized as an effective way to enhance the accuracy of genomic prediction (GP). This study investigated the efficiency of genomic prediction using prior information and combined reference population. In total, prior information considering trait-associated single nucleotide polymorphisms (SNPs) obtained from meta-analysis of genome-wide association studies (GWAS meta-analysis) was incorporated into three models to assess the performance of GP using combined reference populations. Two different Yorkshire populations with imputed whole genome sequence (WGS) data (9,741,620 SNPs), named as P1 (1259 individuals) and P2 (1018 individuals), were used to predict genomic estimated breeding values for three live carcass traits, including backfat thickness, loin muscle area, and loin muscle depth. A 10 × 5 fold cross-validation was used to evaluate the prediction accuracy of 203 randomly selected candidate pigs from the P2 population and the reference population consisted of the remaining pigs from P2 and the stepwise added pigs from P1. By integrating SNPs with different p-value thresholds from GWAS meta-analysis downloaded from PigGTEx Project, the prediction accuracy of GBLUP, genomic feature BLUP (GFBLUP) and GBLUP given genetic architecture (BLUP|GA) were compared. Moreover, we explored effects of reference population size and heritability enrichment of genomic features on the prediction accuracy improvement of GFBLUP and BLUP|GA relative to GBLUP. The prediction accuracy of GBLUP using all WGS markers showed average improvement of 4.380% using the P1 + P2 reference population compared with the P2 reference population. Using the combined reference population, GFBLUP and BLUP|GA yielded 6.179% and 5.525% higher accuracies than GBLUP using all SNPs based on the single reference population, respectively. Positive regression coefficients were estimated in relation to the improvement in prediction accuracy (between GFBLUP/BLUP|GA and GBLUP) and the size of the reference as well as the heritability enrichment of genomic features. Compared to the classic GBLUP model, GFBLUP and BLUP|GA models integrating GWAS meta-analysis information increase the prediction accuracy and using combined populations with enlarged reference population size further enhances prediction accuracy of the two approaches. The heritability enrichment of genomic features can be used as an indicator to reflect weather prior information is accurately presented.
Collapse
Affiliation(s)
- Xiaodian Cai
- National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Wenjing Zhang
- National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Ning Gao
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Chen Wei
- National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Xibo Wu
- Guangxi State Farmd Yongxin Animal Husbandry Group Co., Ltd, Nanning, China
| | - Jinglei Si
- Guangxi State Farmd Yongxin Animal Husbandry Group Co., Ltd, Nanning, China
| | - Yahui Gao
- National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Jiaqi Li
- National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Tong Yin
- Institute of Animal Breeding and Genetics, Justus Liebig University, Giessen, Germany
| | - Zhe Zhang
- National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
4
|
Bravo JI, Zhang L, Benayoun BA. Multi-ancestry GWAS reveals loci linked to human variation in LINE-1- and Alu-insertion numbers. TRANSLATIONAL MEDICINE OF AGING 2025; 9:25-40. [PMID: 40051556 PMCID: PMC11883834 DOI: 10.1016/j.tma.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2025] Open
Abstract
LINE-1 (L1) and Alu are two families of transposable elements (TEs) occupying ~17% and ~11% of the human genome, respectively. Though only a small fraction of L1 copies is able to produce the machinery to mobilize autonomously, Alu and degenerate L1s can hijack their functional machinery and mobilize in trans. The expression and subsequent mobilization of L1 and Alu can exert pathological effects on their hosts. These features have made them promising focus subjects in studies of aging where they can become active. However, mechanisms regulating TE activity are incompletely characterized, especially in diverse human populations. To address these gaps, we leveraged genomic data from the 1000 Genomes Project to carry out a trans-ethnic GWAS of L1/Alu insertion singletons. These are rare, recently acquired insertions observed in only one person and which we used as proxies for variation in L1/Alu insertion numbers. Our approach identified SNVs in genomic regions containing genes with potential and known TE regulatory properties, and it enriched for SNVs in regions containing known regulators of L1 expression. Moreover, we identified reference TE copies and structural variants that associated with L1/Alu singletons, suggesting their potential contribution to TE insertion number variation. Finally, a transcriptional analysis of lymphoblastoid cells highlighted potential cell cycle alterations in a subset of samples harboring L1/Alu singletons. Collectively, our results suggest that known TE regulatory mechanisms may be active in diverse human populations, expand the list of loci implicated in TE insertion number variability, and reinforce links between TEs and disease.
Collapse
Affiliation(s)
- Juan I. Bravo
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Lucia Zhang
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
- Quantitative and Computational Biology Department, USC Dornsife College of Letters, Arts and Sciences, Los Angeles, California, USA
| | - Bérénice A. Benayoun
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
- Molecular and Computational Biology Department, USC Dornsife College of Letters, Arts and Sciences, Los Angeles, CA 90089, USA
- Biochemistry and Molecular Medicine Department, USC Keck School of Medicine, Los Angeles, CA 90089, USA
- USC Norris Comprehensive Cancer Center, Epigenetics and Gene Regulation, Los Angeles, CA 90089, USA
- USC Stem Cell Initiative, Los Angeles, CA 90089, USA
| |
Collapse
|
5
|
Nakatsuka N, Adler D, Jiang L, Hartman A, Cheng E, Klann E, Satija R. A Reproducibility Focused Meta-Analysis Method for Single-Cell Transcriptomic Case-Control Studies Uncovers Robust Differentially Expressed Genes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.15.618577. [PMID: 39463993 PMCID: PMC11507907 DOI: 10.1101/2024.10.15.618577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
We assessed the reproducibility of differentially expressed genes (DEGs) in previously published Alzheimer's (AD), Parkinson's (PD), Schizophrenia (SCZ), and COVID-19 scRNA-seq studies. While transcriptional scores from DEGs of individual PD and COVID-19 datasets had moderate predictive power for case-control status of other datasets (AUC=0.77 and 0.75), genes from individual AD and SCZ datasets had poor predictive power (AUC=0.68 and 0.55). We developed a non-parametric meta-analysis method, SumRank, based on reproducibility of relative differential expression ranks across datasets, and found DEGs with improved predictive power (AUC=0.88, 0.91, 0.78, and 0.62). By multiple other metrics, specificity and sensitivity of these genes were substantially higher than those discovered by dataset merging and inverse variance weighted p-value aggregation methods. The DEGs revealed known and novel biological pathways, and we validate BCAT1 as down-regulated in AD mouse oligodendrocytes. Lastly, we evaluate factors influencing reproducibility of individual studies as a prospective guide for experimental design.
Collapse
|
6
|
Bravo JI, Zhang L, Benayoun BA. Multi-ancestry GWAS reveals loci linked to human variation in LINE-1- and Alu-insertion numbers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.09.10.612283. [PMID: 39314493 PMCID: PMC11419044 DOI: 10.1101/2024.09.10.612283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
LINE-1 (L1) and Alu are two families of transposable elements (TEs) occupying ~17% and ~11% of the human genome, respectively. Though only a small fraction of L1 copies is able to produce the machinery to mobilize autonomously, Alu and degenerate L1s can hijack their functional machinery and mobilize in trans. The expression and subsequent mobilization of L1 and Alu can exert pathological effects on their hosts. These features have made them promising focus subjects in studies of aging where they can become active. However, mechanisms regulating TE activity are incompletely characterized, especially in diverse human populations. To address these gaps, we leveraged genomic data from the 1000 Genomes Project to carry out a trans-ethnic GWAS of L1/Alu insertion singletons. These are rare, recently acquired insertions observed in only one person and which we used as proxies for variation in L1/Alu insertion numbers. Our approach identified SNVs in genomic regions containing genes with potential and known TE regulatory properties, and it enriched for SNVs in regions containing known regulators of L1 expression. Moreover, we identified reference TE copies and structural variants that associated with L1/Alu singletons, suggesting their potential contribution to TE insertion number variation. Finally, a transcriptional analysis of lymphoblastoid cells highlighted potential cell cycle alterations in a subset of samples harboring L1/Alu singletons. Collectively, our results suggest that known TE regulatory mechanisms may be active in diverse human populations, expand the list of loci implicated in TE insertion number variability, and reinforce links between TEs and disease.
Collapse
Affiliation(s)
- Juan I. Bravo
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Lucia Zhang
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
- Quantitative and Computational Biology Department, USC Dornsife College of Letters, Arts and Sciences, Los Angeles, California, USA
| | - Bérénice A. Benayoun
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
- Molecular and Computational Biology Department, USC Dornsife College of Letters, Arts and Sciences, Los Angeles, CA 90089, USA
- Biochemistry and Molecular Medicine Department, USC Keck School of Medicine, Los Angeles, CA 90089, USA
- USC Norris Comprehensive Cancer Center, Epigenetics and Gene Regulation, Los Angeles, CA 90089, USA
- USC Stem Cell Initiative, Los Angeles, CA 90089, USA
| |
Collapse
|
7
|
Kuang A, Hivert MF, Hayes MG, Lowe WL, Scholtens DM. Multi-ancestry genome-wide association analyses: a comparison of meta- and mega-analyses in the Hyperglycemia and Adverse Pregnancy Outcome (HAPO) study. BMC Genomics 2025; 26:65. [PMID: 39849370 PMCID: PMC11755808 DOI: 10.1186/s12864-025-11229-1] [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: 05/22/2024] [Accepted: 01/08/2025] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND There is increasing need for effective incorporation of high-dimensional genetics data from individuals with varied ancestry in genome-wide association (GWAS) analyses. Classically, multi-ancestry GWAS analyses are performed using statistical meta-analysis to combine results conducted within homogeneous ancestry groups. The emergence of cosmopolitan reference panels makes collective preprocessing of GWAS data possible, but impact on downstream GWAS results in a mega-analysis framework merits investigation. We utilized GWAS data from the multi-national Hyperglycemia and Adverse Pregnancy Outcome Study to investigate differences in GWAS findings using a homogeneous ancestry meta-analysis versus a heterogeneous ancestry mega-analysis pipeline. Maternal fasting and 1-hr glucose and metabolomics measured during a 2-hr 75-gram oral glucose tolerance test during early third trimester pregnancy were evaluated as phenotypes. RESULTS For the homogeneous ancestry meta-analysis pipeline, variant data were prepared by identifying sets of individuals with similar ancestry and imputing to ancestry-specific reference panels. GWAS was conducted within each ancestry group and results were combined using random-effects meta-analysis. For the heterogeneous ancestry mega-analysis pipeline, data for all individuals were collectively imputed to the Trans-Omics for Precision Medicine (TOPMed) cosmopolitan reference panel, and GWAS was conducted using a unified mega-analysis. The meta-analysis pipeline identified genome-wide significant associations for 15 variants in a region close to GCK on chromosome 7 with maternal fasting glucose and no significant findings for 1-hr glucose. Associations in this same region were identified using the mega-analysis pipeline, along with a well-documented association at MTNR1B on chromosome 11 with both fasting and 1-hr maternal glucose. For metabolomics analyses, the number of significant findings in the heterogeneous ancestry mega-analysis far exceeded those from the homogeneous ancestry meta-analysis and confirmed many previously documented associations, but genomic inflation factors were much more variable. CONCLUSIONS For multi-ancestry GWAS, heterogeneous ancestry mega-analysis generates a rich set of variants for analysis using a cosmopolitan reference panel and results in vastly more significant, biologically credible and previously documented associations than a homogeneous ancestry meta-analysis approach. Genomic inflation factors do indicate that findings from the mega-analysis pipeline may merit cautious interpretation and further follow-up.
Collapse
Affiliation(s)
- Alan Kuang
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Marie-France Hivert
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Population Medicine, Harvard Pilgrim Health Care Institute, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - M Geoffrey Hayes
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - William L Lowe
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Denise M Scholtens
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
8
|
Zhao F, Wang L, Xu S. Identification of QTL-by-environment interaction by controlling polygenic background effect. J Genet Genomics 2025:S1673-8527(25)00018-9. [PMID: 39805530 DOI: 10.1016/j.jgg.2025.01.003] [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: 07/23/2024] [Revised: 12/30/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025]
Abstract
The quantitative trait loci (QTL)-by-environment (Q × E) interaction effect is hard to detect because there are no effective ways to control the genomic background. In this study, we propose a linear mixed model that simultaneously analyzes data from multiple environments to detect Q × E interactions. This model incorporates two different kinship matrices derived from the genome-wide markers to control both main and interaction polygenic background effects. Simulation studies demonstrate that our approach is more powerful than the meta-analysis and inclusive composite interval mapping methods. We further analyze four agronomic traits of rice across four environments. A main effect QTL is identified for 1000-grain weight (KGW), while no QTL are found for tiller number. Additionally, a large QTL with a significant Q × E interaction is detected on chromosome 7 affecting grain number, yield, and KGW. This region harbors two important genes, PROG1 and Ghd7. Furthermore, we apply our mixed model to analyze lodging in barley across six environments. The six regions exhibiting Q × E interaction effects identified by our approach overlap with the SNPs previously identified using EM and MCMC-based Bayesian methods, further validating the robustness of our approach. Both simulation studies and empirical data analyses show that our method outperformed all other methods compared.
Collapse
Affiliation(s)
- Fuping Zhao
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Lixian Wang
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Shizhong Xu
- Department of Botany and Plant Sciences, University of California, Riverside, CA 92521, USA.
| |
Collapse
|
9
|
da Silva MRG, Veroneze R, Marques DBD, da Silva DA, Machado II, Brito LF, Lopes PS. A meta-analysis of genome-wide association studies to identify candidate genes associated with feed efficiency traits in pigs. J Anim Sci 2025; 103:skaf010. [PMID: 39847436 PMCID: PMC11833465 DOI: 10.1093/jas/skaf010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 01/21/2025] [Indexed: 01/24/2025] Open
Abstract
Pig production is an agricultural sector of great economic and social relevance to Brazil and global markets. Feed efficiency traits directly influence the sustainability of pig production due to the economic impact of feed costs on the production system and the environmental footprint of the industry. Therefore, breeding for improved feed efficiency has been a target of worldwide pig breeding programs. Genome-wide association studies (GWAS) enable the assessment of the genetic background of complex traits, which contributes to a better understanding of the biological mechanisms regulating their phenotypic expression. In this context, the primary objective of this study was to identify and validate genomic regions and candidate genes associated with feed conversion ratio (FCR) and residual feed intake (RFI) in pigs based on a comprehensive systematic review and meta-analysis of GWAS. The METAL software was used to implement the meta-analysis and the Bonferroni multiple testing correction considering a significance threshold 0.05. The significant single nucleotide polymorphisms (SNPs) in the meta-analysis were used to identify candidate genes, followed by a functional genomic enrichment analysis. The systematic review identified 13 studies, of which 7 evaluated FCR, 3 evaluated RFI, and 3 studies investigated both traits, with 160 and 96 SNPs identified for FCR and RFI, respectively. After the meta-analysis, 145 markers were significantly associated with FCR and 90 with RFI. The gene annotation process resulted in 105 and 114 genes for FCR and RFI, respectively. The enrichment analysis for FCR resulted in 16 significant gene ontology (GO) terms, while 6 terms were identified for RFI. The main GO terms were actin cytoskeleton (GO_BP:0030036), membrane (GO_CC:0016020), integral components of the peroxisomal membrane (GO_CC:0005779), and carbohydrate-binding (GO_MF:0030246). The main candidate genes identified were MED18, PHACTR4, ABCC2, TRHDE, FRS2, FAR2 and FIS1 for FCR, and ADGRL2, ASGR1, ASGR2, and MAN2B1 for RFI. These findings contribute to a better understanding of the genetic mechanisms associated with feed efficiency traits in pigs, providing a foundation for future improvements in pig breeding programs.
Collapse
Affiliation(s)
- Maria Rita Gonçalves da Silva
- Department of Animal Science, Federal University of Viçosa, Viçosa, MG, Brazil
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Renata Veroneze
- Department of Animal Science, Federal University of Viçosa, Viçosa, MG, Brazil
| | - Daniele B D Marques
- Department of Animal Science, Federal University of Viçosa, Viçosa, MG, Brazil
| | - Delvan A da Silva
- Department of Animal Science, Federal University of Viçosa, Viçosa, MG, Brazil
| | - Inaê I Machado
- Department of Animal Science, Federal University of Viçosa, Viçosa, MG, Brazil
| | - Luiz F Brito
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Paulo S Lopes
- Department of Animal Science, Federal University of Viçosa, Viçosa, MG, Brazil
| |
Collapse
|
10
|
Hou L, Wu S, Yuan Z, Xue F, Li H. TEMR: Trans-ethnic mendelian randomization method using large-scale GWAS summary datasets. Am J Hum Genet 2025; 112:28-43. [PMID: 39689714 PMCID: PMC11739928 DOI: 10.1016/j.ajhg.2024.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 12/19/2024] Open
Abstract
Available large-scale genome-wide association study (GWAS) summary datasets predominantly stem from European populations, while sample sizes for other ethnicities, notably Central/South Asian, East Asian, African, Hispanic, etc., remain comparatively limited, resulting in low precision of causal effect estimations within these ethnicities when using Mendelian randomization (MR). In this paper, we propose a trans-ethnic MR method, TEMR, to improve the statistical power and estimation precision of MR in a target population that is underrepresented, using trans-ethnic large-scale GWAS summary datasets. TEMR incorporates trans-ethnic genetic correlation coefficients through a conditional likelihood-based inference framework, producing calibrated p values with substantially improved MR power. In the simulation study, compared with other existing MR methods, TEMR exhibited superior precision and statistical power in causal effect estimation within the target populations. Finally, we applied TEMR to infer causal relationships between concentrations of 16 blood biomarkers and the risk of developing five diseases (hypertension, ischemic stroke, type 2 diabetes, schizophrenia, and major depression disorder) in East Asian, African, and Hispanic/Latino populations, leveraging biobank-scale GWAS summary data obtained from individuals of European descent. We found that the causal biomarkers were mostly validated by previous MR methods, and we also discovered 17 causal relationships that were not identified using previously published MR methods.
Collapse
Affiliation(s)
- Lei Hou
- Department of Medical Data, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250000, P.R. China; Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan 250000, P.R. China
| | - Sijia Wu
- Department of Medical Data, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250000, P.R. China; Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan 250000, P.R. China
| | - Zhongshang Yuan
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan 250000, P.R. China; Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250000, P.R. China
| | - Fuzhong Xue
- Department of Medical Data, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250000, P.R. China; Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan 250000, P.R. China; Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250000, P.R. China.
| | - Hongkai Li
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan 250000, P.R. China; Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250000, P.R. China.
| |
Collapse
|
11
|
Gao S, Zhu P, Wang T, Han Z, Xue Y, Zhang Y, Wang L, Zhang H, Chen Y, Liu G. Alzheimer's disease genome-wide association studies in the context of statistical heterogeneity. Mol Psychiatry 2025; 30:342-348. [PMID: 38965422 DOI: 10.1038/s41380-024-02654-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/06/2024]
Affiliation(s)
- Shan Gao
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, 100069, Beijing, China
| | - Ping Zhu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, 100069, Beijing, China
| | - Tao Wang
- Chinese Institute for Brain Research, 102206, Beijing, China
| | - Zhifa Han
- Center of Respiratory Medicine, China-Japan Friendship Hospital, National Center for Respiratory Medicine, Institute of Respiratory Medicine, Chinese Acadamy of Medical Sciences, National Clinical Research Center for Respiratory Diseases, 100029, Beijing, China
| | - Yanli Xue
- School of Biomedical Engineering, Capital Medical University, 10069, Beijing, China
| | - Yan Zhang
- Department of Pathology, The Affiliated Hospital of Weifang Medical University, Weifang, 261053, China
| | - Longcai Wang
- Department of Anesthesiology, The Affiliated Hospital of Weifang Medical University, Weifang, 261053, China
| | - Haihua Zhang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, 100069, Beijing, China
| | - Yan Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Wannan Medical College, No. 22, Wenchang Road, Wuhu, 241002, Anhui, China
- Institute of Chronic Disease Prevention and Control, Wannan Medical College, No.22, Wenchang Road, Wuhu, 241002, Anhui, China
| | - Guiyou Liu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, 100069, Beijing, China.
- Chinese Institute for Brain Research, 102206, Beijing, China.
- Department of Epidemiology and Biostatistics, School of Public Health, Wannan Medical College, No. 22, Wenchang Road, Wuhu, 241002, Anhui, China.
- Institute of Chronic Disease Prevention and Control, Wannan Medical College, No.22, Wenchang Road, Wuhu, 241002, Anhui, China.
- Beijing Key Laboratory of Hypoxia Translational Medicine, Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, National Engineering Laboratory of Internet Medical Diagnosis and Treatment Technology, Xuanwu Hospital, Capital Medical University, 100053, Beijing, China.
- Brain Hospital, Shengli Oilfield Central Hospital, Dongying, 257000, Shandong, China.
| |
Collapse
|
12
|
De Walsche A, Vergne A, Rincent R, Roux F, Nicolas S, Welcker C, Mezmouk S, Charcosset A, Mary-Huard T. metaGE: Investigating genotype x environment interactions through GWAS meta-analysis. PLoS Genet 2025; 21:e1011553. [PMID: 39792927 PMCID: PMC11756807 DOI: 10.1371/journal.pgen.1011553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 01/23/2025] [Accepted: 12/23/2024] [Indexed: 01/12/2025] Open
Abstract
Elucidating the genetic components of plant genotype-by-environment interactions is of key importance in the context of increasing climatic instability, diversification of agricultural practices and pest pressure due to phytosanitary treatment limitations. The genotypic response to environmental stresses can be investigated through multi-environment trials (METs). However, genome-wide association studies (GWAS) of MET data are significantly more complex than that of single environments. In this context, we introduce metaGE, a flexible and computationally efficient meta-analysis approach for jointly analyzing single-environment GWAS of any MET experiment. The metaGE procedure accounts for the heterogeneity of quantitative trait loci (QTL) effects across the environmental conditions and allows the detection of QTL whose allelic effect variations are strongly correlated to environmental cofactors. We evaluated the performance of the proposed methodology and compared it to two competing procedures through simulations. We also applied metaGE to two emblematic examples: the detection of flowering QTLs whose effects are modulated by competition in Arabidopsis and the detection of yield QTLs impacted by drought stresses in maize. The procedure identified known and new QTLs, providing valuable insights into the genetic architecture of complex traits and QTL effects dependent on environmental stress conditions. The whole statistical approach is available as an R package.
Collapse
Affiliation(s)
- Annaïg De Walsche
- Génétique Quantitative et Evolution - Le Moulon, INRAE, CNRS, AgroParisTech, Université Paris-Saclay, Gif-sur-Yvette, France
- MIA Paris-Saclay, INRAE, AgroParisTech, Université Paris-Saclay, Palaiseau, France
| | | | - Renaud Rincent
- Génétique Quantitative et Evolution - Le Moulon, INRAE, CNRS, AgroParisTech, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Fabrice Roux
- LIPME, INRAE, CNRS, Université de Toulouse, Castanet-Tolosan, France
| | - Stéphane Nicolas
- Génétique Quantitative et Evolution - Le Moulon, INRAE, CNRS, AgroParisTech, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Claude Welcker
- LEPSE, Université de Montpellier, INRAE, Institut Agro, Montpellier, France
| | | | - Alain Charcosset
- Génétique Quantitative et Evolution - Le Moulon, INRAE, CNRS, AgroParisTech, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Tristan Mary-Huard
- Génétique Quantitative et Evolution - Le Moulon, INRAE, CNRS, AgroParisTech, Université Paris-Saclay, Gif-sur-Yvette, France
- MIA Paris-Saclay, INRAE, AgroParisTech, Université Paris-Saclay, Palaiseau, France
| |
Collapse
|
13
|
Escriba-Montagut X, Marcon Y, Anguita-Ruiz A, Avraam D, Urquiza J, Morgan AS, Wilson RC, Burton P, Gonzalez JR. Federated privacy-protected meta- and mega-omics data analysis in multi-center studies with a fully open-source analytic platform. PLoS Comput Biol 2024; 20:e1012626. [PMID: 39652598 DOI: 10.1371/journal.pcbi.1012626] [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: 10/27/2023] [Revised: 12/19/2024] [Accepted: 11/10/2024] [Indexed: 12/21/2024] Open
Abstract
The importance of maintaining data privacy and complying with regulatory requirements is highlighted especially when sharing omic data between different research centers. This challenge is even more pronounced in the scenario where a multi-center effort for collaborative omics studies is necessary. OmicSHIELD is introduced as an open-source tool aimed at overcoming these challenges by enabling privacy-protected federated analysis of sensitive omic data. In order to ensure this, multiple security mechanisms have been included in the software. This innovative tool is capable of managing a wide range of omic data analyses specifically tailored to biomedical research. These include genome and epigenome wide association studies and differential gene expression analyses. OmicSHIELD is designed to support both meta- and mega-analysis, so that it offers a wide range of capabilities for different analysis designs. We present a series of use cases illustrating some examples of how the software addresses real-world analyses of omic data.
Collapse
Affiliation(s)
- Xavier Escriba-Montagut
- Barcelona Institute for Global Health (ISGlobal), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | | | - Augusto Anguita-Ruiz
- Barcelona Institute for Global Health (ISGlobal), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Demetris Avraam
- Department of Public Health, Policy and Systems, University of Liverpool, Liverpool, United Kingdom
| | - Jose Urquiza
- Barcelona Institute for Global Health (ISGlobal), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Centro de Investigación Biomédica en Red en Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Andrei S Morgan
- Université Paris Cité, Centre of Research in Epidemiology and StatisticS (CRESS), Obstetrical Perinatal and Pediatric Epidemiology Research Team (EPOPé), INSERM, INRAE, F-75006, Paris, France
- Elizabeth Garrett Anderson Institute for Women's Health London, University College London, London, United Kingdom
| | - Rebecca C Wilson
- Department of Public Health, Policy and Systems, University of Liverpool, Liverpool, United Kingdom
| | - Paul Burton
- Population Health Sciences Institute, Newcastle University, Newcastle, United Kingdom
| | - Juan R Gonzalez
- Barcelona Institute for Global Health (ISGlobal), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Centro de Investigación Biomédica en Red en Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| |
Collapse
|
14
|
Mancin E, Maltecca C, Jiang J, Huang YJ, Tiezzi F. Capturing resilience from phenotypic deviations: a case study using feed consumption and whole genome data in pigs. BMC Genomics 2024; 25:1128. [PMID: 39574040 PMCID: PMC11583387 DOI: 10.1186/s12864-024-11052-0] [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: 06/16/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND In recent years, interest has grown in quantifying resilience in livestock by examining deviations in target phenotypes. This method is based on the idea that variability in these phenotypes reflects an animal's ability to adapt to external factors. By utilizing routinely collected time-series feed intake data in pigs, researchers can obtain a broad measure of resilience. This measure extends beyond specific conditions, capturing the impact of various unknown external factors that influence phenotype variations. Importantly, this method does not require additional phenotyping investments. Despite growing interest, the relationship between resilience indicators-calculated as deviations from longitudinally recorded target traits-and the mean of those traits remains largely unexplored. This gap raises the risk of inadvertently selecting for the mean rather than accurately capturing true resilience. Additionally, distinguishing between random phenotype fluctuations (white noise) and structural variations linked to resilience poses a challenge. With the aim of developing general resilience indicators applicable to commercial swine populations, we devised four resilience indicators utilizing daily feed consumption as the target trait. These include a canonical resilience indicator (BALnVar) and three novel ones (BAMaxArea, SPLnVar, and SPMaxArea), designed to minimize noise and ensure independence from daily feed consumption. We subsequently integrated these indicators with Whole Genome Sequencing using SLEMM algorithm, data from 1,250 animals to assess their efficacy in capturing resilience and their independence from the mean of daily feed consumption. RESULTS Our findings revealed that conventional resilience indicators failed to differentiate from the mean of daily feed consumption, underscoring potential limitations in accurately capturing true resilience. Notably, significant associations involving conventional resilience indicators were identified on chromosome 1, which is commonly linked to body weight. CONCLUSION We observed that deviations in feed consumption can effectively serve as indicators for selecting resilience in commercial pig farming, as confirmed by the identification of genes such as PKN1 and GYPC. However, the identification of other genes, such as RNF152, related to growth, suggests that common resilience quantification methods may be more closely related to the mean of daily feed consumption rather than capturing true resilience.
Collapse
Affiliation(s)
- Enrico Mancin
- Department of Agronomy, Natural Resources, Animals and Environment, (DAFNAE), University of Padova, Viale del Università 14, Legnaro (Padova), Food, 35020, Italy
| | - Christian Maltecca
- Department of Animal Science, North Carolina State University, Raleigh, NC, 27695, USA
- Department of Agriculture, Food, Environment and Forestry (DAGRI), University of Florence, Piazzale delle Cascine 18, Firenze, 50144, Italy
| | - Jicaj Jiang
- Department of Animal Science, North Carolina State University, Raleigh, NC, 27695, USA
| | - Yi Jian Huang
- Smithfield Premium Genetics, Rose Hill, NC, 28458, USA
| | - Francesco Tiezzi
- Department of Agriculture, Food, Environment and Forestry (DAGRI), University of Florence, Piazzale delle Cascine 18, Firenze, 50144, Italy.
| |
Collapse
|
15
|
Rout M, Tung GK, Singh JR, Mehra NK, Wander GS, Ralhan S, Sanghera DK. Polygenic Risk Score Assessment for Coronary Artery Disease in Asian Indians. J Cardiovasc Transl Res 2024; 17:1086-1096. [PMID: 38658478 DOI: 10.1007/s12265-024-10511-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 04/11/2024] [Indexed: 04/26/2024]
Abstract
We evaluated the performance of various polygenic risk score (PRS) models derived from European (EU), South Asian (SA), and Punjabi Asian Indians (AI) studies on 13,974 subjects from AI ancestry. While all models successfully predicted Coronary artery disease (CAD) risk, the AI, SA, and EU + AI were superior predictors and more transportable than the EU model; the predictive performance in training and test sets was 18% and 22% higher in AI and EU + AI models, respectively than in EU. Comparing individuals with extreme PRS quartiles, the AI and EU + AI captured individuals with high CAD risk showed 2.6 to 4.6 times higher efficiency than the EU. Interestingly, including the clinical risk score did not significantly change the performance of any genetic model. The enrichment of diversity variants in EU PRS improves risk prediction and transportability. Establishing population-specific normative and risk factors and inclusion into genetic models would refine the risk stratification and improve the clinical utility of CAD PRS.
Collapse
Affiliation(s)
- Madhusmita Rout
- Department of Pediatrics, Section of Genetics, College of Medicine, University of Oklahoma Health Sciences Center, 940 Stanton L. Young Blvd., Rm 317 BMSB, Oklahoma City, OK, 73104, USA
| | - Gurleen Kaur Tung
- Department of Pediatrics, Section of Genetics, College of Medicine, University of Oklahoma Health Sciences Center, 940 Stanton L. Young Blvd., Rm 317 BMSB, Oklahoma City, OK, 73104, USA
| | | | | | | | - Sarju Ralhan
- Hero DMC Heart Institute, Ludhiana, Punjab, India
| | - Dharambir K Sanghera
- Department of Pediatrics, Section of Genetics, College of Medicine, University of Oklahoma Health Sciences Center, 940 Stanton L. Young Blvd., Rm 317 BMSB, Oklahoma City, OK, 73104, USA.
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
16
|
Whankaew S, Suksri P, Sinprasertporn A, Thawonsuwan J, Sathapondecha P. Development of DNA Markers for Acute Hepatopancreatic Necrosis Disease Tolerance in Litopenaeus vannamei through a Genome-Wide Association Study. BIOLOGY 2024; 13:731. [PMID: 39336158 PMCID: PMC11429464 DOI: 10.3390/biology13090731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/04/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024]
Abstract
Shrimp aquaculture is facing a serious disease, acute hepatopancreatic necrosis disease (AHPND), caused by Vibrio paraheamolyticus (VpAPHND). For sustainable shrimp aquaculture, massive losses of shrimp infected with VpAPHND must be prevented. Research and selection of shrimp tolerant to VpAPHND infection is a sustainable approach to reducing the risk of AHPND. This study focused on the identification and development of potential DNA markers associated with AHPND using DArT sequencing (DArTSeq) and a genome-wide association study. Three populations of post-larval Litopenaeus vannamei were immersed in VpAPHND to collect susceptible (D) and tolerant (S) samples. The 45 D and 48 S shrimp had their genotypes analyzed using DArTSeq. A total of 108,983 SNPs and 17,212 InDels were obtained from the DArTseq data, while the biallelic 516 SNPs and 2293 InDels were finally filtered with PIC < 0.1, MAF < 0.05, and a call rate ≥ 80%. The filtered variants were analyzed for their association with AHPND tolerance. Although there were no significantly associated SNPs and InDels above the Bonferroni correction threshold, candidate variants, four SNPs and 17 InDels corresponding to p < 0.01, were provided for further validation of the AHPND tolerance trait. The candidate SNPs are located on an exon of the zinc finger protein 239-like gene, an intron of an uncharacterized gene, and in intergenic regions. Most of the candidate InDels are in the intergenic regions, with fewer in the intronic and exonic regions. This study provides information on SNPs and InDels for white shrimp. These markers will support the variant database of shrimp and be useful in shrimp aquaculture for breeding selection.
Collapse
Affiliation(s)
- Sukhuman Whankaew
- Faculty of Technology and Community Development, Thaksin University, Phatthalung Campus, Phatthalung 93210, Thailand
| | - Phassorn Suksri
- Center for Genomics and Bioinformatics Research, Division of Biological Sciences, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
| | - Ammara Sinprasertporn
- Songkhla Aquatic Animal Health Research and Development Center, Department of Fisheries, Songkhla 90110, Thailand
| | - Jumroensri Thawonsuwan
- Songkhla Aquatic Animal Health Research and Development Center, Department of Fisheries, Songkhla 90110, Thailand
| | - Ponsit Sathapondecha
- Center for Genomics and Bioinformatics Research, Division of Biological Sciences, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
| |
Collapse
|
17
|
Kontou PI, Bagos PG. The goldmine of GWAS summary statistics: a systematic review of methods and tools. BioData Min 2024; 17:31. [PMID: 39238044 PMCID: PMC11375927 DOI: 10.1186/s13040-024-00385-x] [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: 02/09/2024] [Accepted: 08/27/2024] [Indexed: 09/07/2024] Open
Abstract
Genome-wide association studies (GWAS) have revolutionized our understanding of the genetic architecture of complex traits and diseases. GWAS summary statistics have become essential tools for various genetic analyses, including meta-analysis, fine-mapping, and risk prediction. However, the increasing number of GWAS summary statistics and the diversity of software tools available for their analysis can make it challenging for researchers to select the most appropriate tools for their specific needs. This systematic review aims to provide a comprehensive overview of the currently available software tools and databases for GWAS summary statistics analysis. We conducted a comprehensive literature search to identify relevant software tools and databases. We categorized the tools and databases by their functionality, including data management, quality control, single-trait analysis, and multiple-trait analysis. We also compared the tools and databases based on their features, limitations, and user-friendliness. Our review identified a total of 305 functioning software tools and databases dedicated to GWAS summary statistics, each with unique strengths and limitations. We provide descriptions of the key features of each tool and database, including their input/output formats, data types, and computational requirements. We also discuss the overall usability and applicability of each tool for different research scenarios. This comprehensive review will serve as a valuable resource for researchers who are interested in using GWAS summary statistics to investigate the genetic basis of complex traits and diseases. By providing a detailed overview of the available tools and databases, we aim to facilitate informed tool selection and maximize the effectiveness of GWAS summary statistics analysis.
Collapse
Affiliation(s)
| | - Pantelis G Bagos
- Department of Computer Science and Biomedical Informatics, University of Thessaly, 35131, Lamia, Greece.
| |
Collapse
|
18
|
Guo H, Li T, Shi Y, Wang X. MTML: An Efficient Multitrait Multilocus GWAS Method Based on the Cauchy Combination Test. Biom J 2024; 66:e202300130. [PMID: 39076046 DOI: 10.1002/bimj.202300130] [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: 05/15/2023] [Revised: 10/19/2023] [Accepted: 11/27/2023] [Indexed: 07/31/2024]
Abstract
Genome-wide association study (GWAS) by measuring the joint effect of multiple loci on multiple traits, has recently attracted interest, due to the decreased costs of high-throughput genotyping and phenotyping technologies. Previous studies mainly focused on either multilocus models that identify associations with a single trait or multitrait models that scan a single marker at a time. Since these types of models cannot fully utilize the association information, the powers of the tests are usually low. To potentially address this problem, we present here a multitrait multilocus (MTML) modeling framework that implements in three steps: (1) simplify the complex calculation; (2) reduce the model dimension; (3) integrate the joint contribution of single markers to multiple traits by Cauchy combination. The performances of MTML are evaluated and compared with other three published methods by Monte Carlo simulations. Simulation results show that MTML is more powerful for quantitative trait nucleotide detection and robust for various numbers of traits. In the meanwhile, MTML can effectively control type I error rate at a reasonable level. Real data analysis of Arabidopsis thaliana shows that MTML identifies more pleiotropic genetic associations. Therefore, we conclude that MTML is an efficient GWAS method for joint analysis of multiple quantitative traits. The R package MTML, which facilitates the implementation of the proposed method, is publicly available on GitHub https://github.com/Guohongping/MTML.
Collapse
Affiliation(s)
- Hongping Guo
- School of Mathematics and Statistics, Hubei Normal University, Huangshi, China
| | - Tong Li
- School of Mathematics and Statistics, Hubei Normal University, Huangshi, China
| | - Yao Shi
- School of Mathematics and Statistics, Qingdao University, Qingdao, China
| | - Xiao Wang
- School of Mathematics and Statistics, Qingdao University, Qingdao, China
| |
Collapse
|
19
|
Arivarasan VK, Diwakar D, Kamarudheen N, Loganathan K. Current approaches in CRISPR-Cas systems for diabetes. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 210:95-125. [PMID: 39824586 DOI: 10.1016/bs.pmbts.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2025]
Abstract
In the face of advancements in health care and a shift towards healthy lifestyle, diabetes mellitus (DM) still presents as a global health challenge. This chapter explores recent advancements in the areas of genetic and molecular underpinnings of DM, addressing the revolutionary potential of CRISPR-based genome editing technologies. We delve into the multifaceted relationship between genes and molecular pathways contributing to both type1 and type 2 diabetes. We highlight the importance of how improved genetic screening and the identification of susceptibility genes are aiding in early diagnosis and risk stratification. The spotlight then shifts to CRISPR-Cas9, a robust genome editing tool capable of various applications including correcting mutations in type 1 diabetes, enhancing insulin production in T2D, modulating genes associated with metabolism of glucose and insulin sensitivity. Delivery methods for CRISPR to targeted tissues and cells are explored, including viral and non-viral vectors, alongside the exciting possibilities offered by nanocarriers. We conclude by discussing the challenges and ethical considerations surrounding CRISPR-based therapies for DM. These include potential off-target effects, ensuring long-term efficacy and safety, and navigating the ethical implications of human genome modification. This chapter offers a comprehensive perspective on how genetic and molecular insights, coupled with the transformative power of CRISPR, are paving the way for potential cures and novel therapeutic approaches for DM.
Collapse
Affiliation(s)
- Vishnu Kirthi Arivarasan
- Department of Microbiology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India.
| | - Diksha Diwakar
- Department of Microbiology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| | - Neethu Kamarudheen
- The University of Texas, MD Anderson Cancer Center, Houston, TX, United States
| | | |
Collapse
|
20
|
Talwar JV, Klie A, Pagadala MS, Carter H. GRIEVOUS: your command-line general for resolving cross-dataset genotype inconsistencies. Bioinformatics 2024; 40:btae489. [PMID: 39078222 PMCID: PMC11322043 DOI: 10.1093/bioinformatics/btae489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 07/19/2024] [Accepted: 07/29/2024] [Indexed: 07/31/2024] Open
Abstract
SUMMARY Harmonizing variant indexing and allele assignments across datasets is crucial for data integrity in cross-dataset studies such as multi-cohort genome-wide association studies, meta-analyses, and the development, validation, and application of polygenic risk scores. Ensuring this indexing and allele consistency is a laborious, time-consuming, and error-prone process requiring a certain degree of computational proficiency. Here, we introduce GRIEVOUS, a command-line tool for cross-dataset variant homogenization. By means of an internal database and a custom indexing methodology, GRIEVOUS identifies, formats, and aligns all biallelic single nucleotide polymorphisms (SNPs) across all summary statistic and genotype files of interest. Upon completion of dataset harmonization, GRIEVOUS can also be used to extract the maximal set of biallelic SNPs common to all datasets. AVAILABILITY AND IMPLEMENTATION GRIEVOUS and all supporting documentation and tutorials can be found at https://github.com/jvtalwar/GRIEVOUS. It is freely and publicly available under the MIT license and can be installed via pip.
Collapse
Affiliation(s)
- James V Talwar
- Division of Medical Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, United States
- Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, CA 92093, United States
| | - Adam Klie
- Division of Medical Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, United States
- Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, CA 92093, United States
| | - Meghana S Pagadala
- Biomedical Science Program, University of California San Diego, La Jolla, CA 92093, United States
| | - Hannah Carter
- Division of Medical Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, United States
- Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, CA 92093, United States
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, United States
| |
Collapse
|
21
|
Ishikawa T, Masuda T, Hachiya T, Dina C, Simonet F, Nagata Y, Tanck MWT, Sonehara K, Glinge C, Tadros R, Khongphatthanayothin A, Lu TP, Higuchi C, Nakajima T, Hayashi K, Aizawa Y, Nakano Y, Nogami A, Morita H, Ohno S, Aiba T, Krijger Juárez C, Mauleekoonphairoj J, Poovorawan Y, Gourraud JB, Shimizu W, Probst V, Horie M, Wilde AAM, Redon R, Juang JMJ, Nademanee K, Bezzina CR, Barc J, Tanaka T, Okada Y, Schott JJ, Makita N. Brugada syndrome in Japan and Europe: a genome-wide association study reveals shared genetic architecture and new risk loci. Eur Heart J 2024; 45:2320-2332. [PMID: 38747976 DOI: 10.1093/eurheartj/ehae251] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 03/02/2024] [Accepted: 04/08/2024] [Indexed: 07/10/2024] Open
Abstract
BACKGROUND AND AIMS Brugada syndrome (BrS) is an inherited arrhythmia with a higher disease prevalence and more lethal arrhythmic events in Asians than in Europeans. Genome-wide association studies (GWAS) have revealed its polygenic architecture mainly in European populations. The aim of this study was to identify novel BrS-associated loci and to compare allelic effects across ancestries. METHODS A GWAS was conducted in Japanese participants, involving 940 cases and 1634 controls, followed by a cross-ancestry meta-analysis of Japanese and European GWAS (total of 3760 cases and 11 635 controls). The novel loci were characterized by fine-mapping, gene expression, and splicing quantitative trait associations in the human heart. RESULTS The Japanese-specific GWAS identified one novel locus near ZSCAN20 (P = 1.0 × 10-8), and the cross-ancestry meta-analysis identified 17 association signals, including six novel loci. The effect directions of the 17 lead variants were consistent (94.1%; P for sign test = 2.7 × 10-4), and their allelic effects were highly correlated across ancestries (Pearson's R = .91; P = 2.9 × 10-7). The genetic risk score derived from the BrS GWAS of European ancestry was significantly associated with the risk of BrS in the Japanese population [odds ratio 2.12 (95% confidence interval 1.94-2.31); P = 1.2 × 10-61], suggesting a shared genetic architecture across ancestries. Functional characterization revealed that a lead variant in CAMK2D promotes alternative splicing, resulting in an isoform switch of calmodulin kinase II-δ, favouring a pro-inflammatory/pro-death pathway. CONCLUSIONS This study demonstrates novel susceptibility loci implicating potentially novel pathogenesis underlying BrS. Despite differences in clinical expressivity and epidemiology, the polygenic architecture of BrS was substantially shared across ancestries.
Collapse
Affiliation(s)
- Taisuke Ishikawa
- Omics Research Center, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Tatsuo Masuda
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- StemRIM Institute of Regeneration-Inducing Medicine, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Tsuyoshi Hachiya
- Institute for Biomedical Sciences, Iwate Medical University, Iwate, Japan
| | - Christian Dina
- L'institut du thorax, Nantes Université, CHU Nantes, CNRS, INSERM, Nantes, France
| | - Floriane Simonet
- L'institut du thorax, Nantes Université, CHU Nantes, CNRS, INSERM, Nantes, France
| | - Yuki Nagata
- Bioresource Research Center, Tokyo Medical and Dental University, Tokyo, Japan
- Department of Human Genetics and Disease Diversity, Tokyo Medical and Dental University, Tokyo, Japan
| | - Michael W T Tanck
- Epidemiology and Data Science, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Kyuto Sonehara
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Genome Informatics, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | - Charlotte Glinge
- Department of Experimental Cardiology, Amsterdam UMC location University of Amsterdam, Heart Centre, Amsterdam, The Netherlands
- Heart Failure & Arrhythmias, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN GUARD-HEART https://guardheart.ern-net.eu)
| | - Rafik Tadros
- Department of Experimental Cardiology, Amsterdam UMC location University of Amsterdam, Heart Centre, Amsterdam, The Netherlands
- Heart Failure & Arrhythmias, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
- Montreal Heart Institute, Universite de Montreal, Cardiovascular Genetics Centre, Montreal, Quebec, Canada
| | - Apichai Khongphatthanayothin
- Department of Medicine, Center of Excellence in Arrhythmia Research Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Pediatrics, Division of Cardiology Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Cardiology, Bangkok Hospital, Bangkok, Thailand
| | - Tzu-Pin Lu
- Department of Public Health, Institute of Health Data Analytics and Statistics, National Taiwan University, Taipei, Taiwan
| | - Chihiro Higuchi
- Bioresource Research Center, Tokyo Medical and Dental University, Tokyo, Japan
- Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, Settsu, Japan
| | - Tadashi Nakajima
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Kenshi Hayashi
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Yoshiyasu Aizawa
- Department of Cardiovascular Medicine, International University of Health and Welfare, Narita, Japan
| | - Yukiko Nakano
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Akihiko Nogami
- Department of Cardiology, University of Tsukuba, Tsukuba, Japan
| | - Hiroshi Morita
- Department of Cardiovascular Therapeutics, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Seiko Ohno
- Medical Genome Center, National Cerebral and Cardiovascular Center, Suita, Japan
- Department of Cardiovascular Medicine, Shiga University of Medical Sciences, Otsu, Japan
| | - Takeshi Aiba
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Christian Krijger Juárez
- Department of Experimental Cardiology, Amsterdam UMC location University of Amsterdam, Heart Centre, Amsterdam, The Netherlands
- Heart Failure & Arrhythmias, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - John Mauleekoonphairoj
- Department of Medicine, Center of Excellence in Arrhythmia Research Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Yong Poovorawan
- Center of Excellence in Clinical Virology Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Jean-Baptiste Gourraud
- L'institut du thorax, Nantes Université, CHU Nantes, CNRS, INSERM, Nantes, France
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN GUARD-HEART https://guardheart.ern-net.eu)
| | - Wataru Shimizu
- Department of Cardiovascular Medicine, Nippon Medical School, Tokyo, Japan
| | - Vincent Probst
- L'institut du thorax, Nantes Université, CHU Nantes, CNRS, INSERM, Nantes, France
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN GUARD-HEART https://guardheart.ern-net.eu)
| | - Minoru Horie
- Department of Cardiovascular Medicine, Shiga University of Medical Sciences, Otsu, Japan
| | - Arthur A M Wilde
- Heart Failure & Arrhythmias, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN GUARD-HEART https://guardheart.ern-net.eu)
- Department of Cardiology, Amsterdam UMC location University of Amsterdam, Heart Centre, Amsterdam, The Netherlands
| | - Richard Redon
- L'institut du thorax, Nantes Université, CHU Nantes, CNRS, INSERM, Nantes, France
| | - Jyh-Ming Jimmy Juang
- Cardiovascular Center, Heart Failure Center and Department of Internal Medicine, Division of Cardiology, National Taiwan University Hospital, Taipei, Taiwan
- Department of Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Koonlawee Nademanee
- Department of Medicine, Center of Excellence in Arrhythmia Research Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Pacific Rim Electrophysiology Research Institute, Bumrungrad International Hospital, Bangkok, Thailand
| | - Connie R Bezzina
- Department of Experimental Cardiology, Amsterdam UMC location University of Amsterdam, Heart Centre, Amsterdam, The Netherlands
- Heart Failure & Arrhythmias, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN GUARD-HEART https://guardheart.ern-net.eu)
| | - Julien Barc
- L'institut du thorax, Nantes Université, CHU Nantes, CNRS, INSERM, Nantes, France
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN GUARD-HEART https://guardheart.ern-net.eu)
| | - Toshihiro Tanaka
- Bioresource Research Center, Tokyo Medical and Dental University, Tokyo, Japan
- Department of Human Genetics and Disease Diversity, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Genome Informatics, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Jean-Jacques Schott
- L'institut du thorax, Nantes Université, CHU Nantes, CNRS, INSERM, Nantes, France
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN GUARD-HEART https://guardheart.ern-net.eu)
| | - Naomasa Makita
- Department of Cardiology, Sapporo Teishinkai Hospital, N33, E1, Sapporo 065-0033, Japan
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, 6-1, Kishibe Shimmachi, 564-8565 Suita, Japan
| |
Collapse
|
22
|
Fan X, Monson KR, Peters BA, Whittington JM, Um CY, Oberstein PE, McCullough ML, Freedman ND, Huang WY, Ahn J, Hayes RB. Altered salivary microbiota associated with high-sugar beverage consumption. Sci Rep 2024; 14:13386. [PMID: 38862651 PMCID: PMC11167035 DOI: 10.1038/s41598-024-64324-w] [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: 09/11/2023] [Accepted: 06/07/2024] [Indexed: 06/13/2024] Open
Abstract
The human oral microbiome may alter oral and systemic disease risk. Consuming high sugar content beverages (HSB) can lead to caries development by altering the microbial composition in dental plaque, but little is known regarding HSB-specific oral microbial alterations. Therefore, we conducted a large, population-based study to examine associations of HSB intake with oral microbiome diversity and composition. Using mouthwash samples of 989 individuals in two nationwide U.S. cohorts, bacterial 16S rRNA genes were amplified, sequenced, and assigned to bacterial taxa. HSB intake was quantified from food frequency questionnaires as low (< 1 serving/week), medium (1-3 servings/week), or high (> 3 servings/week). We assessed overall bacterial diversity and presence of specific taxa with respect to HSB intake in each cohort separately and combined in a meta-analysis. Consistently in the two cohorts, we found lower species richness in high HSB consumers (> 3 cans/week) (p = 0.027), and that overall bacterial community profiles differed from those of non-consumers (PERMANOVA p = 0.040). Specifically, presence of a network of commensal bacteria (Lachnospiraceae, Peptostreptococcaceae, and Alloprevotella rava) was less common in high compared to non-consumers, as were other species including Campylobacter showae, Prevotella oulorum, and Mycoplasma faucium. Presence of acidogenic bacteria Bifodobacteriaceae and Lactobacillus rhamnosus was more common in high consumers. Abundance of Fusobacteriales and its genus Leptotrichia, Lachnoanaerobaculum sp., and Campylobacter were lower with higher HSB consumption, and their abundances were correlated. No significant interaction was found for these associations with diabetic status or with microbial markers for caries (S. mutans) and periodontitis (P. gingivalis). Our results suggest that soft drink intake may alter the salivary microbiota, with consistent results across two independent cohorts. The observed perturbations of overrepresented acidogenic bacteria and underrepresented commensal bacteria in high HSB consumers may have implications for oral and systemic disease risk.
Collapse
Affiliation(s)
- Xiaozhou Fan
- Division of Epidemiology, Department of Population Health, NYU Grossman School of Medicine, 180 Madison, New York, NY, 10016, USA
| | - Kelsey R Monson
- Division of Epidemiology, Department of Population Health, NYU Grossman School of Medicine, 180 Madison, New York, NY, 10016, USA
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Brandilyn A Peters
- Division of Epidemiology, Department of Population Health, NYU Grossman School of Medicine, 180 Madison, New York, NY, 10016, USA
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | - Caroline Y Um
- Department of Population Science, American Cancer Society, Atlanta, GA, USA
| | - Paul E Oberstein
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | | | - Neal D Freedman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Wen-Yi Huang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Jiyoung Ahn
- Division of Epidemiology, Department of Population Health, NYU Grossman School of Medicine, 180 Madison, New York, NY, 10016, USA
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Richard B Hayes
- Division of Epidemiology, Department of Population Health, NYU Grossman School of Medicine, 180 Madison, New York, NY, 10016, USA.
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA.
| |
Collapse
|
23
|
Smith JL, Tcheandjieu C, Dikilitas O, Iyer K, Miyazawa K, Hilliard A, Lynch J, Rotter JI, Chen YDI, Sheu WHH, Chang KM, Kanoni S, Tsao PS, Ito K, Kosel M, Clarke SL, Schaid DJ, Assimes TL, Kullo IJ. Multi-Ancestry Polygenic Risk Score for Coronary Heart Disease Based on an Ancestrally Diverse Genome-Wide Association Study and Population-Specific Optimization. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2024; 17:e004272. [PMID: 38380516 PMCID: PMC11372723 DOI: 10.1161/circgen.123.004272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 01/23/2024] [Indexed: 02/22/2024]
Abstract
BACKGROUND Predictive performance of polygenic risk scores (PRS) varies across populations. To facilitate equitable clinical use, we developed PRS for coronary heart disease (CHD; PRSCHD) for 5 genetic ancestry groups. METHODS We derived ancestry-specific and multi-ancestry PRSCHD based on pruning and thresholding (PRSPT) and ancestry-based continuous shrinkage priors (PRSCSx) applied to summary statistics from the largest multi-ancestry genome-wide association study meta-analysis for CHD to date, including 1.1 million participants from 5 major genetic ancestry groups. Following training and optimization in the Million Veteran Program, we evaluated the best-performing PRSCHD in 176,988 individuals across 9 diverse cohorts. RESULTS Multi-ancestry PRSPT and PRSCSx outperformed ancestry-specific PRSPT and PRSCSx across a range of tuning values. Two best-performing multi-ancestry PRSCHD (ie, PRSPTmult and PRSCSxmult) and 1 ancestry-specific (PRSCSxEUR) were taken forward for validation. PRSPTmult demonstrated the strongest association with CHD in individuals of South Asian ancestry and European ancestry (odds ratio per 1 SD [95% CI, 2.75 [2.41-3.14], 1.65 [1.59-1.72]), followed by East Asian ancestry (1.56 [1.50-1.61]), Hispanic/Latino ancestry (1.38 [1.24-1.54]), and African ancestry (1.16 [1.11-1.21]). PRSCSxmult showed the strongest associations in South Asian ancestry (2.67 [2.38-3.00]) and European ancestry (1.65 [1.59-1.71]), lower in East Asian ancestry (1.59 [1.54-1.64]), Hispanic/Latino ancestry (1.51 [1.35-1.69]), and the lowest in African ancestry (1.20 [1.15-1.26]). CONCLUSIONS The use of summary statistics from a large multi-ancestry genome-wide meta-analysis improved the performance of PRSCHD in most ancestry groups compared with single-ancestry methods. Despite the use of one of the largest and most diverse sets of training and validation cohorts to date, improvement of predictive performance was limited in African ancestry. This highlights the need for larger genome-wide association study datasets of underrepresented populations to enhance the performance of PRSCHD.
Collapse
Affiliation(s)
- Johanna L Smith
- Department of Cardiovascular Medicine (J.L.S., O.D., I.J.K.), Mayo Clinic, Rochester, MN
| | - Catherine Tcheandjieu
- Department of Epidemiology and Biostatistics, University of California San Francisco (C.T.)
- Gladstone Institute of Data Science and Biotechnology, Gladstone Institute, San Francisco, CA (C.T.)
- VA Palo Alto Health Care System (C.T., A.H., P.S.T., S.L.C.)
| | - Ozan Dikilitas
- Department of Cardiovascular Medicine (J.L.S., O.D., I.J.K.), Mayo Clinic, Rochester, MN
| | - Kruthika Iyer
- Stanford University School of Medicine, Palo Alto, CA (K. Iyer, A.H.)
| | - Kazuo Miyazawa
- Riken Center for Integrative Medical Sciences, Yokohama City, Japan (K.M., K. Ito)
| | - Austin Hilliard
- VA Palo Alto Health Care System (C.T., A.H., P.S.T., S.L.C.)
- Stanford University School of Medicine, Palo Alto, CA (K. Iyer, A.H.)
| | | | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA (J.I.R., Y.-D.I.C.)
| | - Yii-Der Ida Chen
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA (J.I.R., Y.-D.I.C.)
| | - Wayne Huey-Herng Sheu
- Institute of Molecular and Genomic Medicine, National Health Research Institute (W.H.-H.S.)
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Taipei Veterans General Hospital (W.H.-H.S.)
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Taichung Veterans General Hospital, Taiwan (W.H.-H.S.)
| | - Kyong-Mi Chang
- Corporal Michael J Crescenz VA Medical Center, Philadelphia, PA (K.-M.C.)
| | - Stavroula Kanoni
- Queen Mary University of London, Cambridge, United Kingdom (S.K.)
| | - Philip S Tsao
- VA Palo Alto Health Care System (C.T., A.H., P.S.T., S.L.C.)
- Stanford University, Stanford, CA (P.S.T., S.L.C., T.L.A.)
| | - Kaoru Ito
- Riken Center for Integrative Medical Sciences, Yokohama City, Japan (K.M., K. Ito)
| | - Matthew Kosel
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN (M.K., D.J.S.)
| | - Shoa L Clarke
- VA Palo Alto Health Care System (C.T., A.H., P.S.T., S.L.C.)
- Stanford University, Stanford, CA (P.S.T., S.L.C., T.L.A.)
| | - Daniel J Schaid
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN (M.K., D.J.S.)
| | | | - Iftikhar J Kullo
- Department of Cardiovascular Medicine (J.L.S., O.D., I.J.K.), Mayo Clinic, Rochester, MN
| |
Collapse
|
24
|
Lai EY, Huang YT. Identifying pleiotropic genes via the composite test amidst the complexity of polygenic traits. Brief Bioinform 2024; 25:bbae327. [PMID: 39007593 PMCID: PMC11247409 DOI: 10.1093/bib/bbae327] [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: 04/09/2024] [Revised: 05/29/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
Identifying the causal relationship between genotype and phenotype is essential to expanding our understanding of the gene regulatory network spanning the molecular level to perceptible traits. A pleiotropic gene can act as a central hub in the network, influencing multiple outcomes. Identifying such a gene involves testing under a composite null hypothesis where the gene is associated with, at most, one trait. Traditional methods such as meta-analyses of top-hit $P$-values and sequential testing of multiple traits have been proposed, but these methods fail to consider the background of genome-wide signals. Since Huang's composite test produces uniformly distributed $P$-values for genome-wide variants under the composite null, we propose a gene-level pleiotropy test that entails combining the aforementioned method with the aggregated Cauchy association test. A polygenic trait involves multiple genes with different functions to co-regulate mechanisms. We show that polygenicity should be considered when identifying pleiotropic genes; otherwise, the associations polygenic traits initiate will give rise to false positives. In this study, we constructed gene-trait functional modules using the results of the proposed pleiotropy tests. Our analysis suite was implemented as an R package PGCtest. We demonstrated the proposed method with an application study of the Taiwan Biobank database and identified functional modules comprising specific genes and their co-regulated traits.
Collapse
Affiliation(s)
- En-Yu Lai
- Institute of Statistical Science, Academia Sinica, No.128, Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
| | - Yen-Tsung Huang
- Institute of Statistical Science, Academia Sinica, No.128, Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
| |
Collapse
|
25
|
Balech R, Maalouf F, Kaur S, Jighly A, Joukhadar R, Alsamman AM, Hamwieh A, Khater LA, Rubiales D, Kumar S. Identification of novel genes associated with herbicide tolerance in Lentil (Lens culinaris ssp. culinaris Medik.). Sci Rep 2024; 14:10215. [PMID: 38702403 PMCID: PMC11068770 DOI: 10.1038/s41598-024-59695-z] [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: 07/05/2023] [Accepted: 04/15/2024] [Indexed: 05/06/2024] Open
Abstract
Weeds pose a major constraint in lentil cultivation, leading to decrease farmers' revenues by reducing the yield and increasing the management costs. The development of herbicide tolerant cultivars is essential to increase lentil yield. Even though herbicide tolerant lines have been identified in lentils, breeding efforts are still limited and lack proper validation. Marker assisted selection (MAS) can increase selection accuracy at early generations. Total 292 lentil accessions were evaluated under different dosages of two herbicides, metribuzin and imazethapyr, during two seasons at Marchouch, Morocco and Terbol, Lebanon. Highly significant differences among accessions were observed for days to flowering (DF) and maturity (DM), plant height (PH), biological yield (BY), seed yield (SY), number of pods per plant (NP), as well as the reduction indices (RI) for PH, BY, SY and NP. A total of 10,271 SNPs markers uniformly distributed along the lentil genome were assayed using Multispecies Pulse SNP chip developed at Agriculture Victoria, Melbourne. Meta-GWAS analysis was used to detect marker-trait associations, which detected 125 SNPs markers associated with different traits and clustered in 85 unique quantitative trait loci. These findings provide valuable insights for initiating MAS programs aiming to enhance herbicide tolerance in lentil crop.
Collapse
Affiliation(s)
- Rind Balech
- International Center for Agricultural Research in the Dry Areas (ICARDA), Terbol, Lebanon.
| | - Fouad Maalouf
- International Center for Agricultural Research in the Dry Areas (ICARDA), Terbol, Lebanon.
| | - Sukhjiwan Kaur
- Department of Energy, AgriBio, Environment and Climate Action, Centre for AgriBioscience, 5 Ring Road, Bundoora, VIC, 3083, Australia
| | - Abdulqader Jighly
- Department of Energy, AgriBio, Environment and Climate Action, Centre for AgriBioscience, 5 Ring Road, Bundoora, VIC, 3083, Australia
| | - Reem Joukhadar
- Department of Energy, AgriBio, Environment and Climate Action, Centre for AgriBioscience, 5 Ring Road, Bundoora, VIC, 3083, Australia
| | | | | | - Lynn Abou Khater
- International Center for Agricultural Research in the Dry Areas (ICARDA), Terbol, Lebanon
| | - Diego Rubiales
- Institute for Sustainable Agriculture, CSIC, Córdoba, Spain
| | | |
Collapse
|
26
|
Wang X, Jiang Y, Sun Y. Revealing genomic heterogeneity and commonality: A penalized integrative analysis approach accounting for the adjacency structure of measurements. Genet Epidemiol 2024; 48:114-140. [PMID: 38317326 DOI: 10.1002/gepi.22549] [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: 09/26/2023] [Revised: 12/18/2023] [Accepted: 01/08/2024] [Indexed: 02/07/2024]
Abstract
Advancements in high-throughput genomic technologies have revolutionized the field of disease biomarker identification by providing large-scale genomic data. There is an increasing focus on understanding the relationships among diverse patient groups with distinct disease subtypes and characteristics. Complex diseases exhibit both heterogeneity and shared genomic factors, making it essential to investigate these patterns to accurately detect markers and comprehensively understand the diseases. Integrative analysis has emerged as a promising approach to address this challenge. However, existing studies have been limited by ignoring the adjacency structure of genomic measurements, such as single nucleotide polymorphisms (SNPs) and DNA methylations. In this study, we propose a structured integrative analysis method that incorporates a spline type penalty to accommodate this adjacency structure. We utilize a fused lasso type penalty to identify both heterogeneity and commonality across the groups. Extensive simulations demonstrate its superiority compared to several direct competing methods. The analysis of The Cancer Genome Atlas melanoma data with DNA methylation measurements and GENEVA diabetes data with SNP measurements exhibit that the proposed analysis lead to meaningful findings with better prediction performance and higher selection stability.
Collapse
Affiliation(s)
- Xindi Wang
- Center for Applied Statistics, School of Statistics, Renmin University of China, Beijing, China
| | - Yu Jiang
- School of Public Health, The University of Memphis, Memphis, Tennessee, USA
| | - Yifan Sun
- Center for Applied Statistics, School of Statistics, Renmin University of China, Beijing, China
| |
Collapse
|
27
|
Shi YM, Ou D, Li JT, Bao L, Liu XD, Zhang W, Ding H. Genetically Predicted Apolipoprotein E Levels with the Risk of Panvascular Diseases: A Mendelian Randomization Study. Cardiovasc Toxicol 2024; 24:385-395. [PMID: 38536640 DOI: 10.1007/s12012-024-09846-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 02/28/2024] [Indexed: 04/07/2024]
Abstract
The aim of this study was to comprehensively assess the causal relationship between the overall genetic effect of circulating ApoE levels and panvascular lesions using newer genome-wide association data and two-sample bidirectional Mendelian randomization (MR) analysis. Two-way MR using single-nucleotide polymorphisms of circulating ApoE as instrumental variables was performed using the highest-priority Genome-wide association study (GWAS) data, with factor-adjusted and data-corrected statistics, to estimate causal associations between circulating ApoE levels and 10 pan-vascular diseases in > 500,000 UK Biobank participants, > 400,000 participants of Finnish ancestry, and numerous participants in a consortium of predominantly European ancestry. Meta-analysis was conducted to assess positive results. After correcting for statistical results, elevated circulating ApoE levels were shown to have a significant protective effect against Cerebral ischemia (CI) [IVW odds ratio (OR) 0.888, 95% Confidence Interval (CI): 0.823-0.958, p = 2.3 × 10-3], Coronary heart disease [IVW OR 0.950,95% CI: 0.924-0.976, p = 2.0 × 10-4] had a significant protective effect and potentially suggestive protective causality against Angina pectoris [IVW odds ratio (OR) 0.961, 95%CI: 0.931-0.991, p = 1.1 × 10-2]. There was a potential causal effect for increased risk of Heart failure (HF) [IVW ratio (OR) 1.040, 95%CI: 1.006-1.060, p = 1.8 × 10-2]. (Bonferroni threshold p < 0.0026, PFDR < 0.05) Reverse MR analysis did not reveal significant evidence of a causal effect of PVD on changes in circulating ApoE levels. Meta-analysis increases reliability of results. Elevated circulating ApoE levels were particularly associated with an increased risk of heart failure. Elevated ApoE levels reduce the risk of cerebral ischemia, coronary heart disease, and angina pectoris, reflecting a protective effect. The possible pathophysiological role of circulating ApoE levels in the development of panvascular disease is emphasized.
Collapse
Affiliation(s)
- Yi-Ming Shi
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine On Prevention andTreatmentof Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Dian Ou
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine On Prevention andTreatmentof Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Jia-Ting Li
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine On Prevention andTreatmentof Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Le Bao
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine On Prevention andTreatmentof Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Xiao-Dan Liu
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine On Prevention andTreatmentof Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Wei Zhang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine On Prevention andTreatmentof Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China.
| | - Huang Ding
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine On Prevention andTreatmentof Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China.
| |
Collapse
|
28
|
Zhong Z, Li G, Xu Z, Zeng H, Teng J, Feng X, Diao S, Gao Y, Li J, Zhang Z. Evaluating three strategies of genome-wide association analysis for integrating data from multiple populations. Anim Genet 2024; 55:265-276. [PMID: 38185881 DOI: 10.1111/age.13394] [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: 07/28/2023] [Revised: 11/24/2023] [Accepted: 12/21/2023] [Indexed: 01/09/2024]
Abstract
In livestock, genome-wide association studies (GWAS) are usually conducted in a single population (single-GWAS) with limited sample size and detection power. To enhance the detection power of GWAS, meta-analysis of GWAS (meta-GWAS) and mega-analysis of GWAS (mega-GWAS) have been proposed to integrate data from multiple populations at the level of summary statistics or individual data, respectively. However, there is a lack of comparison for these different strategies, which makes it difficult to guide the best practice of GWAS integrating data from multiple study populations. To maximize the comparison of different association analysis strategies across multiple populations, we conducted single-GWAS, meta-GWAS, and mega-GWAS for the backfat thickness of 100 kg (BFT_100) and days to 100 kg (DAYS_100) within each of the three commercial pig breeds (Duroc, Yorkshire, and Landrace). Based on controlling the genome inflation factor to one, we calculated corrected p-values (pC ). In Yorkshire, with the largest sample size, mega-GWAS, meta-GWAS and single-GWAS detected 149, 38 and 20 significant SNPs (pC < 1E-5) associated with BFT_100, as well as 26, four, and one QTL, respectively. Among them, pC of SNPs from mega-GWAS was the lowest, followed by meta-GWAS and single-GWAS. The correlation of pC among the three GWAS strategies ranged from 0.60 to 0.75 and the correlation of SNP effect values between meta-GWAS and mega-GWAS was 0.74, all showing good agreement. Collectively, even though there are differences in the integration of individual data or summary statistics, integrating data from multiple populations is an effective means of genetic argument for complex traits, especially mega-GWAS versus single-GWAS.
Collapse
Affiliation(s)
- Zhanming Zhong
- National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Guangzhen Li
- National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Zhiting Xu
- National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Haonan Zeng
- National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Jinyan Teng
- National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Xueyan Feng
- National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Shuqi Diao
- National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yahui Gao
- National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Jiaqi Li
- National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Zhe Zhang
- National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
29
|
Gao S, Wang T, Han Z, Hu Y, Zhu P, Xue Y, Huang C, Chen Y, Liu G. Interpretation of 10 years of Alzheimer's disease genetic findings in the perspective of statistical heterogeneity. Brief Bioinform 2024; 25:bbae140. [PMID: 38711368 PMCID: PMC11074593 DOI: 10.1093/bib/bbae140] [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: 08/21/2023] [Revised: 02/22/2024] [Accepted: 03/14/2024] [Indexed: 05/08/2024] Open
Abstract
Common genetic variants and susceptibility loci associated with Alzheimer's disease (AD) have been discovered through large-scale genome-wide association studies (GWAS), GWAS by proxy (GWAX) and meta-analysis of GWAS and GWAX (GWAS+GWAX). However, due to the very low repeatability of AD susceptibility loci and the low heritability of AD, these AD genetic findings have been questioned. We summarize AD genetic findings from the past 10 years and provide a new interpretation of these findings in the context of statistical heterogeneity. We discovered that only 17% of AD risk loci demonstrated reproducibility with a genome-wide significance of P < 5.00E-08 across all AD GWAS and GWAS+GWAX datasets. We highlighted that the AD GWAS+GWAX with the largest sample size failed to identify the most significant signals, the maximum number of genome-wide significant genetic variants or maximum heritability. Additionally, we identified widespread statistical heterogeneity in AD GWAS+GWAX datasets, but not in AD GWAS datasets. We consider that statistical heterogeneity may have attenuated the statistical power in AD GWAS+GWAX and may contribute to explaining the low repeatability (17%) of genome-wide significant AD susceptibility loci and the decreased AD heritability (40-2%) as the sample size increased. Importantly, evidence supports the idea that a decrease in statistical heterogeneity facilitates the identification of genome-wide significant genetic loci and contributes to an increase in AD heritability. Collectively, current AD GWAX and GWAS+GWAX findings should be meticulously assessed and warrant additional investigation, and AD GWAS+GWAX should employ multiple meta-analysis methods, such as random-effects inverse variance-weighted meta-analysis, which is designed specifically for statistical heterogeneity.
Collapse
Affiliation(s)
- Shan Gao
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, No. 10, Xitoutiao, You’an Men Wai, Fengtai District, Beijing 100069, China
| | - Tao Wang
- Chinese Institute for Brain Research, No. 26, Kexueyuan Road, Changping District, Beijing 102206, China
| | - Zhifa Han
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, No. 5, Dongdan Santichao, Dongcheng District, Beijing 100193, China
| | - Yang Hu
- School of Computer Science and Technology, Harbin Institute of Technology, No. 92, Xidazhi Street, Nangang District, Harbin 150006, China
| | - Ping Zhu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, No. 10, Xitoutiao, You’an Men Wai, Fengtai District, Beijing 100069, China
| | - Yanli Xue
- School of Biomedical Engineering, Capital Medical University, No. 10 Xitoutiao, You'an Men Wai, Fengtai District, Beijing 100069, China
| | - Chen Huang
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida WaiLong, Taipa 999078, Macao SAR, China
| | - Yan Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Wannan Medical College, No. 22, Wenchang Road, Wuhu 241002, Anhui, China
- Institute of Chronic Disease Prevention and Control, Wannan Medical College, No. 22, Wenchang Road, Wuhu 241002, Anhui, China
| | - Guiyou Liu
- Chinese Institute for Brain Research, No. 26, Kexueyuan Road, Changping District, Beijing 102206, China
- Department of Epidemiology and Biostatistics, School of Public Health, Wannan Medical College, No. 22, Wenchang Road, Wuhu 241002, Anhui, China
- Institute of Chronic Disease Prevention and Control, Wannan Medical College, No. 22, Wenchang Road, Wuhu 241002, Anhui, China
- Key Laboratory of Cerebral Microcirculation in Universities of Shandong, Department of Neurology, Second Affiliated Hospital, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian 271000, Shandong, China
- Beijing Key Laboratory of Hypoxia Translational Medicine, National Engineering Laboratory of Internet Medical Diagnosis and Treatment Technology, Xuanwu Hospital, Capital Medical University, No. 45, Changchun Road, Xicheng District, Beijing 100053, China
| |
Collapse
|
30
|
Pagoni P, Higgins JPT, Lawlor DA, Stergiakouli E, Warrington NM, Morris TT, Tilling K. Meta-regression of genome-wide association studies to estimate age-varying genetic effects. Eur J Epidemiol 2024; 39:257-270. [PMID: 38183607 PMCID: PMC10995067 DOI: 10.1007/s10654-023-01086-1] [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: 01/30/2023] [Accepted: 11/15/2023] [Indexed: 01/08/2024]
Abstract
Fixed-effect meta-analysis has been used to summarize genetic effects on a phenotype across multiple Genome-Wide Association Studies (GWAS) assuming a common underlying genetic effect. Genetic effects may vary with age (or other characteristics), and not allowing for this in a GWAS might lead to bias. Meta-regression models between study heterogeneity and allows effect modification of the genetic effects to be explored. The aim of this study was to explore the use of meta-analysis and meta-regression for estimating age-varying genetic effects on phenotypes. With simulations we compared the performance of meta-regression to fixed-effect and random -effects meta-analyses in estimating (i) main genetic effects and (ii) age-varying genetic effects (SNP by age interactions) from multiple GWAS studies under a range of scenarios. We applied meta-regression on publicly available summary data to estimate the main and age-varying genetic effects of the FTO SNP rs9939609 on Body Mass Index (BMI). Fixed-effect and random-effects meta-analyses accurately estimated genetic effects when these did not change with age. Meta-regression accurately estimated both main genetic effects and age-varying genetic effects. When the number of studies or the age-diversity between studies was low, meta-regression had limited power. In the applied example, each additional minor allele (A) of rs9939609 was inversely associated with BMI at ages 0 to 3, and positively associated at ages 5.5 to 13. Our findings challenge the assumption that genetic effects are consistent across all ages and provide a method for exploring this. GWAS consortia should be encouraged to use meta-regression to explore age-varying genetic effects.
Collapse
Affiliation(s)
- Panagiota Pagoni
- MRC Integrative Epidemiology Unit at the University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK.
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK.
| | - Julian P T Higgins
- MRC Integrative Epidemiology Unit at the University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Deborah A Lawlor
- MRC Integrative Epidemiology Unit at the University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Evie Stergiakouli
- MRC Integrative Epidemiology Unit at the University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Nicole M Warrington
- MRC Integrative Epidemiology Unit at the University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
- Frazer Institute, University of Queensland, Brisbane, QLD, Australia
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
| | - Tim T Morris
- Centre for Longitudinal Studies, Social Research Institute, University College London, London, UK
| | - Kate Tilling
- MRC Integrative Epidemiology Unit at the University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| |
Collapse
|
31
|
Kolobkov D, Mishra Sharma S, Medvedev A, Lebedev M, Kosaretskiy E, Vakhitov R. Efficacy of federated learning on genomic data: a study on the UK Biobank and the 1000 Genomes Project. Front Big Data 2024; 7:1266031. [PMID: 38487517 PMCID: PMC10937521 DOI: 10.3389/fdata.2024.1266031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 01/31/2024] [Indexed: 03/17/2024] Open
Abstract
Combining training data from multiple sources increases sample size and reduces confounding, leading to more accurate and less biased machine learning models. In healthcare, however, direct pooling of data is often not allowed by data custodians who are accountable for minimizing the exposure of sensitive information. Federated learning offers a promising solution to this problem by training a model in a decentralized manner thus reducing the risks of data leakage. Although there is increasing utilization of federated learning on clinical data, its efficacy on individual-level genomic data has not been studied. This study lays the groundwork for the adoption of federated learning for genomic data by investigating its applicability in two scenarios: phenotype prediction on the UK Biobank data and ancestry prediction on the 1000 Genomes Project data. We show that federated models trained on data split into independent nodes achieve performance close to centralized models, even in the presence of significant inter-node heterogeneity. Additionally, we investigate how federated model accuracy is affected by communication frequency and suggest approaches to reduce computational complexity or communication costs.
Collapse
Affiliation(s)
- Dmitry Kolobkov
- GENXT, Hinxton, United Kingdom
- Laboratory of Ecological Genetics, Vavilov Institute of General Genetics, Moscow, Russia
| | - Satyarth Mishra Sharma
- GENXT, Hinxton, United Kingdom
- Center for Artificial Intelligence Technology, Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Aleksandr Medvedev
- GENXT, Hinxton, United Kingdom
- Center for Artificial Intelligence Technology, Skolkovo Institute of Science and Technology, Moscow, Russia
| | | | | | | |
Collapse
|
32
|
Chen H, King FJ, Zhou B, Wang Y, Canedy CJ, Hayashi J, Zhong Y, Chang MW, Pache L, Wong JL, Jia Y, Joslin J, Jiang T, Benner C, Chanda SK, Zhou Y. Drug target prediction through deep learning functional representation of gene signatures. Nat Commun 2024; 15:1853. [PMID: 38424040 PMCID: PMC10904399 DOI: 10.1038/s41467-024-46089-y] [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: 09/20/2023] [Accepted: 02/14/2024] [Indexed: 03/02/2024] Open
Abstract
Many machine learning applications in bioinformatics currently rely on matching gene identities when analyzing input gene signatures and fail to take advantage of preexisting knowledge about gene functions. To further enable comparative analysis of OMICS datasets, including target deconvolution and mechanism of action studies, we develop an approach that represents gene signatures projected onto their biological functions, instead of their identities, similar to how the word2vec technique works in natural language processing. We develop the Functional Representation of Gene Signatures (FRoGS) approach by training a deep learning model and demonstrate that its application to the Broad Institute's L1000 datasets results in more effective compound-target predictions than models based on gene identities alone. By integrating additional pharmacological activity data sources, FRoGS significantly increases the number of high-quality compound-target predictions relative to existing approaches, many of which are supported by in silico and/or experimental evidence. These results underscore the general utility of FRoGS in machine learning-based bioinformatics applications. Prediction networks pre-equipped with the knowledge of gene functions may help uncover new relationships among gene signatures acquired by large-scale OMICs studies on compounds, cell types, disease models, and patient cohorts.
Collapse
Affiliation(s)
- Hao Chen
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA, 92121, USA.
- Department of Computer Science and Engineering, University of California, Riverside, 900 University Avenue, Riverside, CA, 92521, USA.
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA, 15213, USA.
| | - Frederick J King
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA, 92121, USA
| | - Bin Zhou
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA, 92121, USA
| | - Yu Wang
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA, 92121, USA
| | - Carter J Canedy
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA, 92121, USA
| | - Joel Hayashi
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA, 92121, USA
| | - Yang Zhong
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA, 92121, USA
| | - Max W Chang
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Lars Pache
- NCI Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Julian L Wong
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA, 92121, USA
| | - Yong Jia
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA, 92121, USA
| | - John Joslin
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA, 92121, USA
| | - Tao Jiang
- Department of Computer Science and Engineering, University of California, Riverside, 900 University Avenue, Riverside, CA, 92521, USA
| | - Christopher Benner
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Sumit K Chanda
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA, 92037, USA
| | - Yingyao Zhou
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA, 92121, USA.
| |
Collapse
|
33
|
Li W, Chen R, Feng L, Dang X, Liu J, Chen T, Yang J, Su X, Lv L, Li T, Zhang Z, Luo XJ. Genome-wide meta-analysis, functional genomics and integrative analyses implicate new risk genes and therapeutic targets for anxiety disorders. Nat Hum Behav 2024; 8:361-379. [PMID: 37945807 DOI: 10.1038/s41562-023-01746-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 10/04/2023] [Indexed: 11/12/2023]
Abstract
Anxiety disorders are the most prevalent mental disorders. However, the genetic etiology of anxiety disorders remains largely unknown. Here we conducted a genome-wide meta-analysis on anxiety disorders by including 74,973 (28,392 proxy) cases and 400,243 (146,771 proxy) controls. We identified 14 risk loci, including 10 new associations near CNTNAP5, MAP2, RAB9BP1, BTN1A1, PRR16, PCLO, PTPRD, FARP1, CDH2 and RAB27B. Functional genomics and fine-mapping pinpointed the potential causal variants, and expression quantitative trait loci analysis revealed the potential target genes regulated by the risk variants. Integrative analyses, including transcriptome-wide association study, proteome-wide association study and colocalization analyses, prioritized potential causal genes (including CTNND1 and RAB27B). Evidence from multiple analyses revealed possibly causal genes, including RAB27B, BTN3A2, PCLO and CTNND1. Finally, we showed that Ctnnd1 knockdown affected dendritic spine density and resulted in anxiety-like behaviours in mice, revealing the potential role of CTNND1 in anxiety disorders. Our study identified new risk loci, potential causal variants and genes for anxiety disorders, providing insights into the genetic architecture of anxiety disorders and potential therapeutic targets.
Collapse
Affiliation(s)
- Wenqiang Li
- Henan Mental Hospital, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Rui Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Laipeng Feng
- Henan Mental Hospital, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Xinglun Dang
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Southeast University, Nanjing, China
| | - Jiewei Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Tengfei Chen
- Henan Mental Hospital, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Jinfeng Yang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Xi Su
- Henan Mental Hospital, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Luxian Lv
- Henan Mental Hospital, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Tao Li
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhijun Zhang
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Southeast University, Nanjing, China
- Department of Neurology, Affiliated Zhongda Hospital, Southeast University, Nanjing, China
- Department of Mental Health and Public Health, Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiong-Jian Luo
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Southeast University, Nanjing, China.
- Department of Neurology, Affiliated Zhongda Hospital, Southeast University, Nanjing, China.
| |
Collapse
|
34
|
Kim EE, Jang CS, Kim H, Han B. PASTRY: achieving balanced power for detecting risk and protective minor alleles in meta-analysis of association studies with overlapping subjects. BMC Bioinformatics 2024; 25:24. [PMID: 38216869 PMCID: PMC10790263 DOI: 10.1186/s12859-023-05627-z] [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: 07/15/2023] [Accepted: 12/20/2023] [Indexed: 01/14/2024] Open
Abstract
BACKGROUND Meta-analysis is a statistical method that combines the results of multiple studies to increase statistical power. When multiple studies participating in a meta-analysis utilize the same public dataset as controls, the summary statistics from these studies become correlated. To solve this challenge, Lin and Sullivan proposed a method to provide an optimal test statistic adjusted for the correlation. This method quickly became the standard practice. However, we identified an unexpected power asymmetry phenomenon in this standard framework. This can lead to unbalanced power for detecting protective minor alleles and risk minor alleles. RESULTS We found that the power asymmetry of the current framework is mainly due to the errors in approximating the correlation term. We then developed a meta-analysis method based on an accurate correlation estimator, called PASTRY (A method to avoid Power ASymmeTRY). PASTRY outperformed the standard method on both simulated and real datasets in terms of the power symmetry. CONCLUSIONS Our findings suggest that PASTRY can help to alleviate the power asymmetry problem. PASTRY is available at https://github.com/hanlab-SNU/PASTRY .
Collapse
Affiliation(s)
- Emma E Kim
- Department of Chemistry, Seoul National University, Seoul, 03080, Korea
| | - Chloe Soohyun Jang
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Hakin Kim
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 03080, Korea
| | - Buhm Han
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea.
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 03080, Korea.
| |
Collapse
|
35
|
Mentis AFA, Liu L. Global impact and application of Precision Healthcare. THE NEW ERA OF PRECISION MEDICINE 2024:209-228. [DOI: 10.1016/b978-0-443-13963-5.00001-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
36
|
Ma Z, Chang Y, Brito LF, Li Y, Yang T, Wang Y, Yang N. Multitrait meta-analyses identify potential candidate genes for growth-related traits in Holstein heifers. J Dairy Sci 2023; 106:9055-9070. [PMID: 37641329 DOI: 10.3168/jds.2023-23462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 06/20/2023] [Indexed: 08/31/2023]
Abstract
Understanding the underlying pleiotropic relationships among growth and body size traits is important for refining breeding strategies in dairy cattle for optimal body size and growth rate. Therefore, we performed single-trait GWAS for monthly-recorded body weight (BW), hip height, body length, and chest girth from birth to 12 mo of age in Holstein animals, followed by stepwise multiple regression of independent or lowly-linked markers from GWAS loci using conditional and joint association analyses (COJO). Subsequently, we conducted a multitrait meta-analysis to detect pleiotropic markers. Based on the single-trait GWAS, we identified 170 significant SNPs, in which 59 of them remained significant after the COJO analyses. The most significant SNP, located at BTA7:3,676,741, explained 2.93% of the total phenotypic variance for BW6 (BW at 6 mo of age). We identified 17 SNPs with potential pleiotropic effects based on the multitrait meta-analyses, which resulted in 3 additional SNPs in comparison to those detected based on the single-trait GWAS. The identified quantitative trait loci regions overlap with genes known to influence human growth-related traits. According to positional and functional analyses, we proposed HMGA2, HNF4G, MED13L, BHLHE40, FRZB, DMP1, TRIB3, and GATAD2A as important candidate genes influencing the studied traits. The combination of single-trait GWAS and meta-analyses of GWAS results improved the efficiency of detecting associated SNPs, and provided new insights into the genetic mechanisms of growth and development in Holstein cattle.
Collapse
Affiliation(s)
- Z Ma
- Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture of China, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, 100193, Beijing, China; Beijing Sunlon Livestock Development Co. Ltd., 100029, Beijing, China
| | - Y Chang
- Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture of China, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, 100193, Beijing, China
| | - Luiz F Brito
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907
| | - Y Li
- Beijing Sunlon Livestock Development Co. Ltd., 100029, Beijing, China
| | - T Yang
- Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture of China, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, 100193, Beijing, China
| | - Y Wang
- Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture of China, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, 100193, Beijing, China.
| | - N Yang
- Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture of China, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, 100193, Beijing, China.
| |
Collapse
|
37
|
Beaumont RN, Flatley C, Vaudel M, Wu X, Chen J, Moen GH, Skotte L, Helgeland Ø, Solé-Navais P, Banasik K, Albiñana C, Ronkainen J, Fadista J, Stinson SE, Trajanoska K, Wang CA, Westergaard D, Srinivasan S, Sánchez-Soriano C, Bilbao JR, Allard C, Groleau M, Kuulasmaa T, Leirer DJ, White F, Jacques PÉ, Cheng H, Hao K, Andreassen OA, Åsvold BO, Atalay M, Bhatta L, Bouchard L, Brumpton BM, Brunak S, Bybjerg-Grauholm J, Ebbing C, Elliott P, Engelbrechtsen L, Erikstrup C, Estarlich M, Franks S, Gaillard R, Geller F, Grove J, Hougaard DM, Kajantie E, Morgen CS, Nohr EA, Nyegaard M, Palmer CNA, Pedersen OB, Rivadeneira F, Sebert S, Shields BM, Stoltenberg C, Surakka I, Thørner LW, Ullum H, Vaarasmaki M, Vilhjalmsson BJ, Willer CJ, Lakka TA, Gybel-Brask D, Bustamante M, Hansen T, Pearson ER, Reynolds RM, Ostrowski SR, Pennell CE, Jaddoe VWV, Felix JF, Hattersley AT, Melbye M, Lawlor DA, Hveem K, Werge T, Nielsen HS, Magnus P, Evans DM, Jacobsson B, Järvelin MR, Zhang G, Hivert MF, Johansson S, Freathy RM, Feenstra B, Njølstad PR. Genome-wide association study of placental weight identifies distinct and shared genetic influences between placental and fetal growth. Nat Genet 2023; 55:1807-1819. [PMID: 37798380 PMCID: PMC10632150 DOI: 10.1038/s41588-023-01520-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 08/31/2023] [Indexed: 10/07/2023]
Abstract
A well-functioning placenta is essential for fetal and maternal health throughout pregnancy. Using placental weight as a proxy for placental growth, we report genome-wide association analyses in the fetal (n = 65,405), maternal (n = 61,228) and paternal (n = 52,392) genomes, yielding 40 independent association signals. Twenty-six signals are classified as fetal, four maternal and three fetal and maternal. A maternal parent-of-origin effect is seen near KCNQ1. Genetic correlation and colocalization analyses reveal overlap with birth weight genetics, but 12 loci are classified as predominantly or only affecting placental weight, with connections to placental development and morphology, and transport of antibodies and amino acids. Mendelian randomization analyses indicate that fetal genetically mediated higher placental weight is causally associated with preeclampsia risk and shorter gestational duration. Moreover, these analyses support the role of fetal insulin in regulating placental weight, providing a key link between fetal and placental growth.
Collapse
Affiliation(s)
- Robin N Beaumont
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Christopher Flatley
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Genetics and Bioinformatics, Health Data and Digitalization, Norwegian Institute of Public Health, Oslo, Norway
| | - Marc Vaudel
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Xiaoping Wu
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Jing Chen
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Gunn-Helen Moen
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Frazer Institute, The University of Queensland, Brisbane, Queensland, Australia
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Line Skotte
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Øyvind Helgeland
- Department of Genetics and Bioinformatics, Health Data and Digitalization, Norwegian Institute of Public Health, Oslo, Norway
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Pol Solé-Navais
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Karina Banasik
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Clara Albiñana
- National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
| | | | - João Fadista
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
- Department of Clinical Sciences, Lund University Diabetes Centre, Malmö, Sweden
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Sara Elizabeth Stinson
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Katerina Trajanoska
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Human Genetics, McGill University, Montréal, Québec, Canada
| | - Carol A Wang
- School of Medicine and Public Health, College of Medicine, Public Health and Wellbeing, The University of Newcastle, Newcastle, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, Newcastle, New South Wales, Australia
| | - David Westergaard
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
- Department of Obstetrics and Gynecology, Copenhagen University Hospital, Hvidovre, Denmark
- Methods and Analysis, Statistics Denmark, Copenhagen, Denmark
| | - Sundararajan Srinivasan
- Division of Population Health and Genomics, School of Medicine, University of Dundee, Dundee, UK
| | | | - Jose Ramon Bilbao
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa, Spain
- Biobizkaia Health Research Institute, Barakaldo, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM), Barcelona, Spain
| | - Catherine Allard
- Centre de recherche du Centre Hospitalier de l'Universite de Sherbrooke, Sherbrooke, Québec, Canada
| | - Marika Groleau
- Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Teemu Kuulasmaa
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio Campus, Kuopio, Finland
| | - Daniel J Leirer
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Frédérique White
- Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Pierre-Étienne Jacques
- Centre de recherche du Centre Hospitalier de l'Universite de Sherbrooke, Sherbrooke, Québec, Canada
- Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Haoxiang Cheng
- Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Ke Hao
- Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Ole A Andreassen
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Bjørn Olav Åsvold
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- HUNT Research Centre, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Levanger, Norway
- Department of Endocrinology, Clinic of Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Mustafa Atalay
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio Campus, Kuopio, Finland
| | - Laxmi Bhatta
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
| | - Luigi Bouchard
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Clinical Department of Laboratory Medicine, Centre intégré universitaire de santé et de services sociaux (CIUSSS) du Saguenay-Lac-St-Jean-Hôpital Universitaire de Chicoutimi, Saguenay, Québec, Canada
| | - Ben Michael Brumpton
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- HUNT Research Centre, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Levanger, Norway
- Clinic of Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Jonas Bybjerg-Grauholm
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
| | - Cathrine Ebbing
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
| | - Paul Elliott
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Line Engelbrechtsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Obstetrics and Gynecology, Herlev Hospital, Herlev, Denmark
| | - Christian Erikstrup
- Department Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
- Department Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Marisa Estarlich
- Faculty of Nursing and Chiropody, Universitat de València, C/Menendez Pelayo, Valencia, Spain
- Epidemiology and Environmental Health Joint Research Unit, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region, FISABIO-Public Health, FISABIO-Universitat Jaume I-Universitat de València, Valencia, Spain
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Stephen Franks
- Institute of Reproductive and Developmental Biology, Imperial College London, London, UK
| | - Romy Gaillard
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Frank Geller
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Jakob Grove
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- Department of Biomedicine-Human Genetics and the iSEQ Center, Aarhus University, Aarhus, Denmark
- Center for Genomics and Personalized Medicine, Aarhus, Denmark
- Bioinformatics Research Centre, Aarhus University, Aarhus, Denmark
| | - David M Hougaard
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
| | - Eero Kajantie
- Research Unit of Clinical Medicine, Medical Research Center, University of Oulu, Oulu, Finland
- Population Health Unit, Finnish Institute for Health and Welfare, Helsinki and Oulu, Oulu, Finland
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Camilla S Morgen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- National Institute of Public Health, University of Southern Denmark, Copenhagen, Denmark
| | - Ellen A Nohr
- Institute of Clinical research, University of Southern Denmark, Odense, Denmark
| | - Mette Nyegaard
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Colin N A Palmer
- Division of Population Health and Genomics, School of Medicine, University of Dundee, Dundee, UK
| | - Ole Birger Pedersen
- Department of Clinical Immunology, Zealand University Hospital, Køge, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Sylvain Sebert
- Research Unit of Population Health, University of Oulu, Oulu, Finland
| | - Beverley M Shields
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Camilla Stoltenberg
- Norwegian Institute of Public Health, Oslo, Norway
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
| | - Ida Surakka
- Department of Internal Medicine, Division of Cardiology, University of Michigan, Ann Arbor, MI, USA
| | - Lise Wegner Thørner
- Department of Clinical Immunology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | | | - Marja Vaarasmaki
- Research Unit of Clinical Medicine, Medical Research Center, University of Oulu, Oulu, Finland
- Department of Obstetrics and Gynaecology, Oulu University Hospital, Oulu, Finland
| | - Bjarni J Vilhjalmsson
- National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
- Bioinformatics Research Centre, Aarhus University, Aarhus, Denmark
| | - Cristen J Willer
- Department of Internal Medicine, Division of Cardiology, University of Michigan, Ann Arbor, MI, USA
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Timo A Lakka
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio Campus, Kuopio, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio, Finland
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Dorte Gybel-Brask
- Psychotherapeutic Outpatient Clinic, Mental Health Services, Capital Region, Copenhagen, Denmark
| | - Mariona Bustamante
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Madrid, Spain
- ISGlobal, Institute for Global Health, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ewan R Pearson
- Division of Population Health and Genomics, School of Medicine, University of Dundee, Dundee, UK
| | - Rebecca M Reynolds
- Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK
| | - Sisse R Ostrowski
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Immunology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Craig E Pennell
- School of Medicine and Public Health, College of Medicine, Public Health and Wellbeing, The University of Newcastle, Newcastle, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, Newcastle, New South Wales, Australia
| | - Vincent W V Jaddoe
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Janine F Felix
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Andrew T Hattersley
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Mads Melbye
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Danish Cancer Institute, Copenhagen, Denmark
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Deborah A Lawlor
- Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Kristian Hveem
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- HUNT Research Centre, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Levanger, Norway
| | - Thomas Werge
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Institute of Biological Psychiatry, Mental Health Services, Copenhagen University Hospital, Copenhagen, Denmark
- Lundbeck Center for Geogenetics, GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
| | - Henriette Svarre Nielsen
- Department of Obstetrics and Gynecology, Copenhagen University Hospital, Hvidovre, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Per Magnus
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - David M Evans
- Frazer Institute, The University of Queensland, Brisbane, Queensland, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Bo Jacobsson
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Genetics and Bioinformatics, Health Data and Digitalization, Norwegian Institute of Public Health, Oslo, Norway
| | - Marjo-Riitta Järvelin
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Center for Life Course Health Research, University of Oulu, Oulu, Finland
- MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- Unit of Primary Health Care, Oulu University Hospital, OYS, Oulu, Finland
| | - Ge Zhang
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Center for Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- March of Dimes Prematurity Research Center Ohio Collaborative, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Marie-France Hivert
- Department of Population Medicine, Harvard Medical School, Harvard Pilgrim Health Care Institute, Boston, MA, USA
- Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Stefan Johansson
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway.
- Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway.
| | - Rachel M Freathy
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK.
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK.
| | - Bjarke Feenstra
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark.
- Department of Clinical Immunology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark.
| | - Pål R Njølstad
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway.
- Children and Youth Clinic, Haukeland University Hospital, Bergen, Norway.
| |
Collapse
|
38
|
Chiwina K, Xiong H, Bhattarai G, Dickson RW, Phiri TM, Chen Y, Alatawi I, Dean D, Joshi NK, Chen Y, Riaz A, Gepts P, Brick M, Byrne PF, Schwartz H, Ogg JB, Otto K, Fall A, Gilbert J, Shi A. Genome-Wide Association Study and Genomic Prediction of Fusarium Wilt Resistance in Common Bean Core Collection. Int J Mol Sci 2023; 24:15300. [PMID: 37894980 PMCID: PMC10607830 DOI: 10.3390/ijms242015300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/29/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
The common bean (Phaseolus vulgaris L.) is a globally cultivated leguminous crop. Fusarium wilt (FW), caused by Fusarium oxysporum f. sp. phaseoli (Fop), is a significant disease leading to substantial yield loss in common beans. Disease-resistant cultivars are recommended to counteract this. The objective of this investigation was to identify single nucleotide polymorphism (SNP) markers associated with FW resistance and to pinpoint potential resistant common bean accessions within a core collection, utilizing a panel of 157 accessions through the Genome-wide association study (GWAS) approach with TASSEL 5 and GAPIT 3. Phenotypes for Fop race 1 and race 4 were matched with genotypic data from 4740 SNPs of BARCBean6K_3 Infinium Bea Chips. After ranking the 157-accession panel and revealing 21 Fusarium wilt-resistant accessions, the GWAS pinpointed 16 SNPs on chromosomes Pv04, Pv05, Pv07, Pv8, and Pv09 linked to Fop race 1 resistance, 23 SNPs on chromosomes Pv03, Pv04, Pv05, Pv07, Pv09, Pv10, and Pv11 associated with Fop race 4 resistance, and 7 SNPs on chromosomes Pv04 and Pv09 correlated with both Fop race 1 and race 4 resistances. Furthermore, within a 30 kb flanking region of these associated SNPs, a total of 17 candidate genes were identified. Some of these genes were annotated as classical disease resistance protein/enzymes, including NB-ARC domain proteins, Leucine-rich repeat protein kinase family proteins, zinc finger family proteins, P-loopcontaining nucleoside triphosphate hydrolase superfamily, etc. Genomic prediction (GP) accuracy for Fop race resistances ranged from 0.26 to 0.55. This study advanced common bean genetic enhancement through marker-assisted selection (MAS) and genomic selection (GS) strategies, paving the way for improved Fop resistance.
Collapse
Affiliation(s)
- Kenani Chiwina
- Department of Horticulture, University of Arkansas, Fayetteville, AR 72701, USA; (K.C.); (G.B.); (R.W.D.); (T.M.P.); (Y.C.); (I.A.); (D.D.)
| | - Haizheng Xiong
- Department of Horticulture, University of Arkansas, Fayetteville, AR 72701, USA; (K.C.); (G.B.); (R.W.D.); (T.M.P.); (Y.C.); (I.A.); (D.D.)
| | - Gehendra Bhattarai
- Department of Horticulture, University of Arkansas, Fayetteville, AR 72701, USA; (K.C.); (G.B.); (R.W.D.); (T.M.P.); (Y.C.); (I.A.); (D.D.)
| | - Ryan William Dickson
- Department of Horticulture, University of Arkansas, Fayetteville, AR 72701, USA; (K.C.); (G.B.); (R.W.D.); (T.M.P.); (Y.C.); (I.A.); (D.D.)
| | - Theresa Makawa Phiri
- Department of Horticulture, University of Arkansas, Fayetteville, AR 72701, USA; (K.C.); (G.B.); (R.W.D.); (T.M.P.); (Y.C.); (I.A.); (D.D.)
| | - Yilin Chen
- Department of Horticulture, University of Arkansas, Fayetteville, AR 72701, USA; (K.C.); (G.B.); (R.W.D.); (T.M.P.); (Y.C.); (I.A.); (D.D.)
| | - Ibtisam Alatawi
- Department of Horticulture, University of Arkansas, Fayetteville, AR 72701, USA; (K.C.); (G.B.); (R.W.D.); (T.M.P.); (Y.C.); (I.A.); (D.D.)
| | - Derek Dean
- Department of Horticulture, University of Arkansas, Fayetteville, AR 72701, USA; (K.C.); (G.B.); (R.W.D.); (T.M.P.); (Y.C.); (I.A.); (D.D.)
| | - Neelendra K. Joshi
- Department of Entomology and Plant Pathology, University of Arkansas, Fayetteville, AR 72701, USA;
| | - Yuyan Chen
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR 72701, USA;
| | - Awais Riaz
- Department of Crop, Soil and Environmental Sciences, University of Arkansas, Fayetteville, AR 72701, USA;
| | - Paul Gepts
- Department of Plant Sciences, University of California, 1 Shields Avenue, Davis, CA 95616, USA;
| | - Mark Brick
- Department of Soil and Crop Sciences, Colorado State University, Fort Collins, CO 80523, USA; (M.B.); (P.F.B.); (J.B.O.); (A.F.); (J.G.)
| | - Patrick F. Byrne
- Department of Soil and Crop Sciences, Colorado State University, Fort Collins, CO 80523, USA; (M.B.); (P.F.B.); (J.B.O.); (A.F.); (J.G.)
| | - Howard Schwartz
- Department of Agricultural Biology, Colorado State University, Fort Collins, CO 80523, USA; (H.S.); (K.O.)
| | - James B. Ogg
- Department of Soil and Crop Sciences, Colorado State University, Fort Collins, CO 80523, USA; (M.B.); (P.F.B.); (J.B.O.); (A.F.); (J.G.)
| | - Kristin Otto
- Department of Agricultural Biology, Colorado State University, Fort Collins, CO 80523, USA; (H.S.); (K.O.)
| | - Amy Fall
- Department of Soil and Crop Sciences, Colorado State University, Fort Collins, CO 80523, USA; (M.B.); (P.F.B.); (J.B.O.); (A.F.); (J.G.)
| | - Jeremy Gilbert
- Department of Soil and Crop Sciences, Colorado State University, Fort Collins, CO 80523, USA; (M.B.); (P.F.B.); (J.B.O.); (A.F.); (J.G.)
| | - Ainong Shi
- Department of Horticulture, University of Arkansas, Fayetteville, AR 72701, USA; (K.C.); (G.B.); (R.W.D.); (T.M.P.); (Y.C.); (I.A.); (D.D.)
| |
Collapse
|
39
|
Sanchez MP, Tribout T, Kadri NK, Chitneedi PK, Maak S, Hozé C, Boussaha M, Croiseau P, Philippe R, Spengeler M, Kühn C, Wang Y, Li C, Plastow G, Pausch H, Boichard D. Sequence-based GWAS meta-analyses for beef production traits. Genet Sel Evol 2023; 55:70. [PMID: 37828440 PMCID: PMC10568825 DOI: 10.1186/s12711-023-00848-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 10/04/2023] [Indexed: 10/14/2023] Open
Abstract
BACKGROUND Combining the results of within-population genome-wide association studies (GWAS) based on whole-genome sequences into a single meta-analysis (MA) is an accurate and powerful method for identifying variants associated with complex traits. As part of the H2020 BovReg project, we performed sequence-level MA for beef production traits. Five partners from France, Switzerland, Germany, and Canada contributed summary statistics from sequence-based GWAS conducted with 54,782 animals from 15 purebred or crossbred populations. We combined the summary statistics for four growth, nine morphology, and 15 carcass traits into 16 MA, using both fixed effects and z-score methods. RESULTS The fixed-effects method was generally more informative to provide indication on potentially causal variants, although we combined substantially different traits in each MA. In comparison with within-population GWAS, this approach highlighted (i) a larger number of quantitative trait loci (QTL), (ii) QTL more frequently located in genomic regions known for their effects on growth and meat/carcass traits, (iii) a smaller number of genomic variants within the QTL, and (iv) candidate variants that were more frequently located in genes. MA pinpointed variants in genes, including MSTN, LCORL, and PLAG1 that have been previously associated with morphology and carcass traits. We also identified dozens of other variants located in genes associated with growth and carcass traits, or with a function that may be related to meat production (e.g., HS6ST1, HERC2, WDR75, COL3A1, SLIT2, MED28, and ANKAR). Some of these variants overlapped with expression or splicing QTL reported in the cattle Genotype-Tissue Expression atlas (CattleGTEx) and could therefore regulate gene expression. CONCLUSIONS By identifying candidate genes and potential causal variants associated with beef production traits in cattle, MA demonstrates great potential for investigating the biological mechanisms underlying these traits. As a complement to within-population GWAS, this approach can provide deeper insights into the genetic architecture of complex traits in beef cattle.
Collapse
Affiliation(s)
- Marie-Pierre Sanchez
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France.
| | - Thierry Tribout
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | | | - Praveen K Chitneedi
- Research Institute for Farm Animal Biology (FBN), 18196, Dummerstorf, Germany
| | - Steffen Maak
- Research Institute for Farm Animal Biology (FBN), 18196, Dummerstorf, Germany
| | - Chris Hozé
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
- Eliance, 75595, Paris, France
| | - Mekki Boussaha
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | - Pascal Croiseau
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | - Romain Philippe
- INRAE, USC1061 GAMAA, Université de Limoges, 87060, Limoges, France
| | | | - Christa Kühn
- Research Institute for Farm Animal Biology (FBN), 18196, Dummerstorf, Germany
- Agricultural and Environmental faculty, University Rostock, 18059, Rostock, Germany
- Friedrich-Loeffler-Institut (FLI), 17493, Greifswald, Insel Riems, Germany
| | - Yining Wang
- Lacombe Research and Development Centre, Agriculture and Agri-Food Canada, Lacombe, AB, T4L 1W1, Canada
| | - Changxi Li
- Lacombe Research and Development Centre, Agriculture and Agri-Food Canada, Lacombe, AB, T4L 1W1, Canada
- Department of Agricultural, Food and Nutritional Science, Livestock Gentec, University of Alberta, Edmonton, AB, T6G 2HI, Canada
| | - Graham Plastow
- Department of Agricultural, Food and Nutritional Science, Livestock Gentec, University of Alberta, Edmonton, AB, T6G 2HI, Canada
| | - Hubert Pausch
- Animal Genomics, ETH Zurich, 8092, Zurich, Switzerland
| | - Didier Boichard
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| |
Collapse
|
40
|
Beugnot A, Mary-Huard T, Bauland C, Combes V, Madur D, Lagardère B, Palaffre C, Charcosset A, Moreau L, Fievet JB. Identifying QTLs involved in hybrid performance and heterotic group complementarity: new GWAS models applied to factorial and admixed diallel maize hybrid panels. TAG. THEORETICAL AND APPLIED GENETICS. THEORETISCHE UND ANGEWANDTE GENETIK 2023; 136:219. [PMID: 37816986 PMCID: PMC10564676 DOI: 10.1007/s00122-023-04431-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 07/25/2023] [Indexed: 10/12/2023]
Abstract
KEY MESSAGE An original GWAS model integrating the ancestry of alleles was proposed and allowed the detection of background specific additive and dominance QTLs involved in heterotic group complementarity and hybrid performance. Maize genetic diversity is structured into genetic groups selected and improved relative to each other. This process increases group complementarity and differentiation over time and ensures that the hybrids produced from inter-group crosses exhibit high performances and heterosis. To identify loci involved in hybrid performance and heterotic group complementarity, we introduced an original association study model that disentangles allelic effects from the heterotic group origin of the alleles and compared it with a conventional additive/dominance model. This new model was applied on a factorial between Dent and Flint lines and a diallel between Dent-Flint admixed lines with two different layers of analysis: within each environment and in a multiple-environment context. We identified several strong additive QTLs for all traits, including some well-known additive QTLs for flowering time (in the region of Vgt1/2 on chromosome 8). Yield trait displayed significant non-additive effects in the diallel panel. Most of the detected Yield QTLs exhibited overdominance or, more likely, pseudo-overdominance effects. Apparent overdominance at these QTLs contributed to a part of the genetic group complementarity. The comparison between environments revealed a higher stability of additive QTL effects than non-additive ones. Several QTLs showed variations of effects according to the local heterotic group origin. We also revealed large chromosomic regions that display genetic group origin effects. Altogether, our results illustrate how admixed panels combined with dedicated GWAS modeling allow the identification of new QTLs that could not be revealed by a classical hybrid panel analyzed with traditional modeling.
Collapse
Affiliation(s)
- Aurélien Beugnot
- Université Paris-Saclay, INRAE, CNRS, AgroParisTech, UMR GQE-Le Moulon, 91272, Gif-Sur-Yvette, France
| | - Tristan Mary-Huard
- Université Paris-Saclay, INRAE, CNRS, AgroParisTech, UMR GQE-Le Moulon, 91272, Gif-Sur-Yvette, France
- Université Paris-Saclay, AgroParisTech, INRAE, UMR MIA Paris-Saclay, 91120, Palaiseau, France
| | - Cyril Bauland
- Université Paris-Saclay, INRAE, CNRS, AgroParisTech, UMR GQE-Le Moulon, 91272, Gif-Sur-Yvette, France
| | - Valerie Combes
- Université Paris-Saclay, INRAE, CNRS, AgroParisTech, UMR GQE-Le Moulon, 91272, Gif-Sur-Yvette, France
| | - Delphine Madur
- Université Paris-Saclay, INRAE, CNRS, AgroParisTech, UMR GQE-Le Moulon, 91272, Gif-Sur-Yvette, France
| | | | | | - Alain Charcosset
- Université Paris-Saclay, INRAE, CNRS, AgroParisTech, UMR GQE-Le Moulon, 91272, Gif-Sur-Yvette, France
| | - Laurence Moreau
- Université Paris-Saclay, INRAE, CNRS, AgroParisTech, UMR GQE-Le Moulon, 91272, Gif-Sur-Yvette, France
| | - Julie B Fievet
- Université Paris-Saclay, INRAE, CNRS, AgroParisTech, UMR GQE-Le Moulon, 91272, Gif-Sur-Yvette, France.
| |
Collapse
|
41
|
Uba CU, Oselebe HO, Tesfaye AA, Abtew WG. Association mapping in bambara groundnut [Vigna subterranea (L.) Verdc.] reveals loci associated with agro-morphological traits. BMC Genomics 2023; 24:593. [PMID: 37803263 PMCID: PMC10557193 DOI: 10.1186/s12864-023-09684-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 09/19/2023] [Indexed: 10/08/2023] Open
Abstract
BACKGROUND Genome-wide association studies (GWAS) are important for the acceleration of crop improvement through knowledge of marker-trait association (MTA). This report used DArT SNP markers to successfully perform GWAS on agro-morphological traits using 270 bambara groundnut [Vigna subterranea (L.) Verdc.] landraces sourced from diverse origins. The study aimed to identify marker traits association for nine agronomic traits using GWAS and their candidate genes. The experiment was conducted at two different locations laid out in alpha lattice design. The cowpea [Vigna unguiculata (L.) Walp.] reference genome (i.e. legume genome most closely related to bambara groundnut) assisted in the identification of candidate genes. RESULTS The analyses showed that linkage disequilibrium was found to decay rapidly with an average genetic distance of 148 kb. The broadsense heritability was relatively high and ranged from 48.39% (terminal leaf length) to 79.39% (number of pods per plant). The GWAS identified a total of 27 significant marker-trait associations (MTAs) for the nine studied traits explaining 5.27% to 24.86% of phenotypic variations. Among studied traits, the highest number of MTAs was obtained from seed coat colour (6) followed by days to flowering (5), while the least is days to maturity (1), explaining 5.76% to 11.03%, 14.5% to 19.49%, and 11.66% phenotypic variations, respectively. Also, a total of 17 candidate genes were identified, varying in number for different traits; seed coat colour (6), days to flowering (3), terminal leaf length (2), terminal leaf width (2), number of seed per pod (2), pod width (1) and days to maturity (1). CONCLUSION These results revealed the prospect of GWAS in identification of SNP variations associated with agronomic traits in bambara groundnut. Also, its present new opportunity to explore GWAS and marker assisted strategies in breeding of bambara groundnut for acceleration of the crop improvement.
Collapse
Affiliation(s)
- Charles U Uba
- Department of Horticulture and Plant Science, Jimma University, Jimma, Ethiopia.
| | | | - Abush A Tesfaye
- International Institute of Tropical Agriculture, Ibadan, Nigeria
| | - Wosene G Abtew
- Department of Horticulture and Plant Science, Jimma University, Jimma, Ethiopia
| |
Collapse
|
42
|
Beuchel C, Dittrich J, Becker S, Kirsten H, Tönjes A, Kovacs P, Stumvoll M, Loeffler M, Teren A, Thiery J, Isermann B, Ceglarek U, Scholz M. An atlas of genome-wide gene expression and metabolite associations and possible mediation effects towards body mass index. J Mol Med (Berl) 2023; 101:1305-1321. [PMID: 37672078 PMCID: PMC10560167 DOI: 10.1007/s00109-023-02362-z] [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: 11/29/2022] [Revised: 08/07/2023] [Accepted: 08/15/2023] [Indexed: 09/07/2023]
Abstract
Investigating the cross talk of different omics layers is crucial to understand molecular pathomechanisms of metabolic diseases like obesity. Here, we present a large-scale association meta-analysis of genome-wide whole blood and peripheral blood mononuclear cell (PBMC) gene expressions profiled with Illumina HT12v4 microarrays and metabolite measurements from dried blood spots (DBS) characterized by targeted liquid chromatography tandem mass spectrometry (LC-MS/MS) in three large German cohort studies with up to 7706 samples. We found 37,295 associations comprising 72 amino acids (AA) and acylcarnitine (AC) metabolites (including ratios) and 8579 transcripts. We applied this catalogue of associations to investigate the impact of associating transcript-metabolite pairs on body mass index (BMI) as an example metabolic trait. This is achieved by conducting a comprehensive mediation analysis considering metabolites as mediators of gene expression effects and vice versa. We discovered large mediation networks comprising 27,023 potential mediation effects within 20,507 transcript-metabolite pairs. Resulting networks of highly connected (hub) transcripts and metabolites were leveraged to gain mechanistic insights into metabolic signaling pathways. In conclusion, here, we present the largest available multi-omics integration of genome-wide transcriptome data and metabolite data of amino acid and fatty acid metabolism and further leverage these findings to characterize potential mediation effects towards BMI proposing candidate mechanisms of obesity and related metabolic diseases. KEY MESSAGES: Thousands of associations of 72 amino acid and acylcarnitine metabolites and 8579 genes expand the knowledge of metabolome-transcriptome associations. A mediation analysis of effects on body mass index revealed large mediation networks of thousands of obesity-related gene-metabolite pairs. Highly connected, potentially mediating hub genes and metabolites enabled insight into obesity and related metabolic disease pathomechanisms.
Collapse
Affiliation(s)
- Carl Beuchel
- Institute for Medical Informatics, Statistics and Epidemiology, Leipzig University, Leipzig, Germany
| | - Julia Dittrich
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, Leipzig University, Leipzig, Germany
| | - Susen Becker
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, Leipzig University, Leipzig, Germany
- Department of Forensic Toxicology, Institute of Legal Medicine, University Leipzig, Leipzig, Germany
| | - Holger Kirsten
- Institute for Medical Informatics, Statistics and Epidemiology, Leipzig University, Leipzig, Germany
- LIFE - Leipzig Research Center for Civilization Diseases, Leipzig University, Leipzig, Germany
| | - Anke Tönjes
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University Hospital Leipzig, Leipzig, Germany
| | - Peter Kovacs
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University Hospital Leipzig, Leipzig, Germany
- Deutsches Zentrum für Diabetesforschung, Neuherberg, Germany
| | - Michael Stumvoll
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University Hospital Leipzig, Leipzig, Germany
| | - Markus Loeffler
- Institute for Medical Informatics, Statistics and Epidemiology, Leipzig University, Leipzig, Germany
- LIFE - Leipzig Research Center for Civilization Diseases, Leipzig University, Leipzig, Germany
| | | | - Joachim Thiery
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, Leipzig University, Leipzig, Germany
- LIFE - Leipzig Research Center for Civilization Diseases, Leipzig University, Leipzig, Germany
| | - Berend Isermann
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, Leipzig University, Leipzig, Germany
- LIFE - Leipzig Research Center for Civilization Diseases, Leipzig University, Leipzig, Germany
| | - Uta Ceglarek
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, Leipzig University, Leipzig, Germany
- LIFE - Leipzig Research Center for Civilization Diseases, Leipzig University, Leipzig, Germany
| | - Markus Scholz
- Institute for Medical Informatics, Statistics and Epidemiology, Leipzig University, Leipzig, Germany.
- LIFE - Leipzig Research Center for Civilization Diseases, Leipzig University, Leipzig, Germany.
| |
Collapse
|
43
|
Deflaux N, Selvaraj MS, Condon HR, Mayo K, Haidermota S, Basford MA, Lunt C, Philippakis AA, Roden DM, Denny JC, Musick A, Collins R, Allen N, Effingham M, Glazer D, Natarajan P, Bick AG. Demonstrating paths for unlocking the value of cloud genomics through cross cohort analysis. Nat Commun 2023; 14:5419. [PMID: 37669985 PMCID: PMC10480504 DOI: 10.1038/s41467-023-41185-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 08/24/2023] [Indexed: 09/07/2023] Open
Abstract
Recently, large scale genomic projects such as All of Us and the UK Biobank have introduced a new research paradigm where data are stored centrally in cloud-based Trusted Research Environments (TREs). To characterize the advantages and drawbacks of different TRE attributes in facilitating cross-cohort analysis, we conduct a Genome-Wide Association Study of standard lipid measures using two approaches: meta-analysis and pooled analysis. Comparison of full summary data from both approaches with an external study shows strong correlation of known loci with lipid levels (R2 ~ 83-97%). Importantly, 90 variants meet the significance threshold only in the meta-analysis and 64 variants are significant only in pooled analysis, with approximately 20% of variants in each of those groups being most prevalent in non-European, non-Asian ancestry individuals. These findings have important implications, as technical and policy choices lead to cross-cohort analyses generating similar, but not identical results, particularly for non-European ancestral populations.
Collapse
Affiliation(s)
| | - Margaret Sunitha Selvaraj
- Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Henry Robert Condon
- Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kelsey Mayo
- Vanderbilt Institute for Clinical and Translational Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sara Haidermota
- Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Division of Cardiology, Massachusetts General Hospital, Boston, MA, USA
| | - Melissa A Basford
- Vanderbilt Institute for Clinical and Translational Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Chris Lunt
- All of Us Research Program, National Institutes of Health, Bethesda, MD, USA
| | | | - Dan M Roden
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Joshua C Denny
- All of Us Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Anjene Musick
- All of Us Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Rory Collins
- Nuffield Department of Population Health, University of Oxford, Oxford, Oxfordshire, UK
- UK Biobank, Cheadle, Stockport, UK
| | - Naomi Allen
- Nuffield Department of Population Health, University of Oxford, Oxford, Oxfordshire, UK
- UK Biobank, Cheadle, Stockport, UK
| | | | | | - Pradeep Natarajan
- Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Division of Cardiology, Massachusetts General Hospital, Boston, MA, USA
| | - Alexander G Bick
- Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
44
|
Cruz LA, Cooke Bailey JN, Crawford DC. Importance of Diversity in Precision Medicine: Generalizability of Genetic Associations Across Ancestry Groups Toward Better Identification of Disease Susceptibility Variants. Annu Rev Biomed Data Sci 2023; 6:339-356. [PMID: 37196357 PMCID: PMC10720270 DOI: 10.1146/annurev-biodatasci-122220-113250] [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] [Indexed: 05/19/2023]
Abstract
Genome-wide association studies (GWAS) revolutionized our understanding of common genetic variation and its impact on common human disease and traits. Developed and adopted in the mid-2000s, GWAS led to searchable genotype-phenotype catalogs and genome-wide datasets available for further data mining and analysis for the eventual development of translational applications. The GWAS revolution was swift and specific, including almost exclusively populations of European descent, to the neglect of the majority of the world's genetic diversity. In this narrative review, we recount the GWAS landscape of the early years that established a genotype-phenotype catalog that is now universally understood to be inadequate for a complete understanding of complex human genetics. We then describe approaches taken to augment the genotype-phenotype catalog, including the study populations, collaborative consortia, and study design approaches aimed to generalize and then ultimately discover genome-wide associations in non-European descent populations. The collaborations and data resources established in the efforts to diversify genomic findings undoubtedly provide the foundations of the next chapters of genetic association studies with the advent of budget-friendly whole-genome sequencing.
Collapse
Affiliation(s)
- Lauren A Cruz
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, USA;
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Jessica N Cooke Bailey
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, USA;
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, USA
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Dana C Crawford
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, USA;
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, USA
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
45
|
Cho SB. Molecular Mechanisms of Endometriosis Revealed Using Omics Data. Biomedicines 2023; 11:2210. [PMID: 37626707 PMCID: PMC10452455 DOI: 10.3390/biomedicines11082210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/22/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
Endometriosis is a gynecological disorder prevalent in women of reproductive age. The primary symptoms include dysmenorrhea, irregular menstruation, and infertility. However, the pathogenesis of endometriosis remains unclear. With the advent of high-throughput technologies, various omics experiments have been conducted to identify genes related to the pathophysiology of endometriosis. This review highlights the molecular mechanisms underlying endometriosis using omics. When genes identified in omics experiments were compared with endometriosis disease genes identified in independent studies, the number of overlapping genes was moderate. However, the characteristics of these genes were found to be equivalent when functional gene set enrichment analysis was performed using gene ontology and biological pathway information. These findings indicate that omics technology provides invaluable information regarding the pathophysiology of endometriosis. Moreover, the functional characteristics revealed using enrichment analysis provide important clues for discovering endometriosis disease genes in future research.
Collapse
Affiliation(s)
- Seong Beom Cho
- Department of Biomedical Informatics, College of Medicine, Gachon University, 38-13, Dokgeom-ro 3 Street Namdon-gu, Incheon 21565, Republic of Korea
| |
Collapse
|
46
|
Fields A, Potel KN, Cabuhal R, Aziri B, Stewart ID, Schock BC. Mediators of systemic sclerosis-associated interstitial lung disease (SSc-ILD): systematic review and meta-analyses. Thorax 2023; 78:799-807. [PMID: 36261273 PMCID: PMC10359532 DOI: 10.1136/thorax-2022-219226] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 09/23/2022] [Indexed: 11/04/2022]
Abstract
Systemic sclerosis-associated interstitial lung disease (SSc-ILD) is rare, poorly understood, with heterogeneous characteristics resulting in difficult diagnosis. We aimed to systematically review evidence of soluble markers in peripheral blood or bronchoalveolar lavage fluid (BALF) as biomarkers in SSc-ILD. METHOD Five databases were screened for observational or interventional, peer-reviewed studies in adults published between January 2000 and September 2021 that assessed levels of biomarkers in peripheral blood or BALF of SSc-ILD patients compared with healthy controls. Qualitative assessment was performed using Critical Appraisal Skills Programme (CASP) checklists. Standardised mean difference (SMD) in biomarkers were combined in random-effects meta-analyses where multiple independent studies reported quantitative data. RESULTS 768 published studies were identified; 38 articles were included in the qualitative synthesis. Thirteen studies were included in the meta-analyses representing three biomarkers: KL6, SP-D and IL-8. Greater IL-8 levels were associated with SSc-ILD in both peripheral blood and BALF, overall SMD 0.88 (95% CI 0.61 to 1.15; I2=1%). Greater levels of SP-D and KL-6 were both estimated in SSc-ILD peripheral blood compared with healthy controls, at an SMD of 1.78 (95% CI 1.50 to 2.17; I2=8%) and 1.66 (95% CI 1.17 to 2.14; I2=76%), respectively. CONCLUSION We provide robust evidence that KL-6, SP-D and IL-8 have the potential to serve as reliable biomarkers in blood/BALF for supporting the diagnosis of SSc-ILD. However, while several other biomarkers have been proposed, the evidence of their independent value in diagnosis and prognosis is currently lacking and needs further investigation. PROSPERO REGISTRATION NUMBER CRD42021282452.
Collapse
Affiliation(s)
- Aislin Fields
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast Faculty of Medicine Health and Life Sciences, Belfast, UK
| | - Koray N Potel
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast Faculty of Medicine Health and Life Sciences, Belfast, UK
| | - Rhandel Cabuhal
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast Faculty of Medicine Health and Life Sciences, Belfast, UK
| | - Buena Aziri
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast Faculty of Medicine Health and Life Sciences, Belfast, UK
- Sarajevo Medical School, Sarajevo School of Science and Technology Sarajevo Medical School, Sarajevo, Bosnia and Herzegovina
| | - Iain D Stewart
- National Heart & Lung Institute, Imperial College London, London, UK
| | - Bettina C Schock
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast Faculty of Medicine Health and Life Sciences, Belfast, UK
| |
Collapse
|
47
|
Wang S, Kang Y, Qi F, Jin H. Genetics of hair graying with age. Ageing Res Rev 2023; 89:101977. [PMID: 37276979 DOI: 10.1016/j.arr.2023.101977] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 03/17/2023] [Accepted: 06/01/2023] [Indexed: 06/07/2023]
Abstract
Hair graying is an early and obvious phenotypic and physiological trait with age in humans. Several recent advances in molecular biology and genetics have increased our understanding of the mechanisms of hair graying, which elucidate genes related to the synthesis, transport, and distribution of melanin in hair follicles, as well as genes regulating these processes above. Therefore, we review these advances and examine the trends in the genetic aspects of hair graying from enrichment theory, Genome-Wide association studies, whole exome sequencing, gene expression studies, and animal models for hair graying with age, aiming to overview the changes in hair graying at the genetic level and establish the foundation for future research. Meanwhile, by summarizing the genetics, it's of great value to explore the possible mechanism, treatment, or even prevention of hair graying with age.
Collapse
Affiliation(s)
- Sifan Wang
- Department of Dermatology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing 100730, China
| | - Yuanbo Kang
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Shuaifuyuan1#, Dongcheng District, Beijing 100730, P.R.China
| | - Fei Qi
- Department of Dermatology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing 100730, China
| | - Hongzhong Jin
- Department of Dermatology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing 100730, China.
| |
Collapse
|
48
|
Sinha D, Maurya AK, Abdi G, Majeed M, Agarwal R, Mukherjee R, Ganguly S, Aziz R, Bhatia M, Majgaonkar A, Seal S, Das M, Banerjee S, Chowdhury S, Adeyemi SB, Chen JT. Integrated Genomic Selection for Accelerating Breeding Programs of Climate-Smart Cereals. Genes (Basel) 2023; 14:1484. [PMID: 37510388 PMCID: PMC10380062 DOI: 10.3390/genes14071484] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/14/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Rapidly rising population and climate changes are two critical issues that require immediate action to achieve sustainable development goals. The rising population is posing increased demand for food, thereby pushing for an acceleration in agricultural production. Furthermore, increased anthropogenic activities have resulted in environmental pollution such as water pollution and soil degradation as well as alterations in the composition and concentration of environmental gases. These changes are affecting not only biodiversity loss but also affecting the physio-biochemical processes of crop plants, resulting in a stress-induced decline in crop yield. To overcome such problems and ensure the supply of food material, consistent efforts are being made to develop strategies and techniques to increase crop yield and to enhance tolerance toward climate-induced stress. Plant breeding evolved after domestication and initially remained dependent on phenotype-based selection for crop improvement. But it has grown through cytological and biochemical methods, and the newer contemporary methods are based on DNA-marker-based strategies that help in the selection of agronomically useful traits. These are now supported by high-end molecular biology tools like PCR, high-throughput genotyping and phenotyping, data from crop morpho-physiology, statistical tools, bioinformatics, and machine learning. After establishing its worth in animal breeding, genomic selection (GS), an improved variant of marker-assisted selection (MAS), has made its way into crop-breeding programs as a powerful selection tool. To develop novel breeding programs as well as innovative marker-based models for genetic evaluation, GS makes use of molecular genetic markers. GS can amend complex traits like yield as well as shorten the breeding period, making it advantageous over pedigree breeding and marker-assisted selection (MAS). It reduces the time and resources that are required for plant breeding while allowing for an increased genetic gain of complex attributes. It has been taken to new heights by integrating innovative and advanced technologies such as speed breeding, machine learning, and environmental/weather data to further harness the GS potential, an approach known as integrated genomic selection (IGS). This review highlights the IGS strategies, procedures, integrated approaches, and associated emerging issues, with a special emphasis on cereal crops. In this domain, efforts have been taken to highlight the potential of this cutting-edge innovation to develop climate-smart crops that can endure abiotic stresses with the motive of keeping production and quality at par with the global food demand.
Collapse
Affiliation(s)
- Dwaipayan Sinha
- Department of Botany, Government General Degree College, Mohanpur 721436, India
| | - Arun Kumar Maurya
- Department of Botany, Multanimal Modi College, Modinagar, Ghaziabad 201204, India
| | - Gholamreza Abdi
- Department of Biotechnology, Persian Gulf Research Institute, Persian Gulf University, Bushehr 75169, Iran
| | - Muhammad Majeed
- Department of Botany, University of Gujrat, Punjab 50700, Pakistan
| | - Rachna Agarwal
- Applied Genomics Section, Bhabha Atomic Research Centre, Mumbai 400085, India
| | - Rashmi Mukherjee
- Research Center for Natural and Applied Sciences, Department of Botany (UG & PG), Raja Narendralal Khan Women's College, Gope Palace, Midnapur 721102, India
| | - Sharmistha Ganguly
- Department of Dravyaguna, Institute of Post Graduate Ayurvedic Education and Research, Kolkata 700009, India
| | - Robina Aziz
- Department of Botany, Government, College Women University, Sialkot 51310, Pakistan
| | - Manika Bhatia
- TERI School of Advanced Studies, New Delhi 110070, India
| | - Aqsa Majgaonkar
- Department of Botany, St. Xavier's College (Autonomous), Mumbai 400001, India
| | - Sanchita Seal
- Department of Botany, Polba Mahavidyalaya, Polba 712148, India
| | - Moumita Das
- V. Sivaram Research Foundation, Bangalore 560040, India
| | - Swastika Banerjee
- Department of Botany, Kairali College of +3 Science, Champua, Keonjhar 758041, India
| | - Shahana Chowdhury
- Department of Biotechnology, Faculty of Engineering Sciences, German University Bangladesh, TNT Road, Telipara, Chandona Chowrasta, Gazipur 1702, Bangladesh
| | - Sherif Babatunde Adeyemi
- Ethnobotany/Phytomedicine Laboratory, Department of Plant Biology, Faculty of Life Sciences, University of Ilorin, Ilorin P.M.B 1515, Nigeria
| | - Jen-Tsung Chen
- Department of Life Sciences, National University of Kaohsiung, Kaohsiung 811, Taiwan
| |
Collapse
|
49
|
Chang L, Zhou G, Xia J. mGWAS-Explorer 2.0: Causal Analysis and Interpretation of Metabolite-Phenotype Associations. Metabolites 2023; 13:826. [PMID: 37512533 PMCID: PMC10384390 DOI: 10.3390/metabo13070826] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 06/23/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023] Open
Abstract
Metabolomics-based genome-wide association studies (mGWAS) are key to understanding the genetic regulations of metabolites in complex phenotypes. We previously developed mGWAS-Explorer 1.0 to link single-nucleotide polymorphisms (SNPs), metabolites, genes and phenotypes for hypothesis generation. It has become clear that identifying potential causal relationships between metabolites and phenotypes, as well as providing deep functional insights, are crucial for further downstream applications. Here, we introduce mGWAS-Explorer 2.0 to support the causal analysis between >4000 metabolites and various phenotypes. The results can be interpreted within the context of semantic triples and molecular quantitative trait loci (QTL) data. The underlying R package is released for reproducible analysis. Using two case studies, we demonstrate that mGWAS-Explorer 2.0 is able to detect potential causal relationships between arachidonic acid and Crohn's disease, as well as between glycine and coronary heart disease.
Collapse
Affiliation(s)
- Le Chang
- Department of Human Genetics, McGill University, Montreal, QC H3A 0C7, Canada
| | - Guangyan Zhou
- Institute of Parasitology, McGill University, Montreal, QC H9X 3V9, Canada
| | - Jianguo Xia
- Department of Human Genetics, McGill University, Montreal, QC H3A 0C7, Canada
- Institute of Parasitology, McGill University, Montreal, QC H9X 3V9, Canada
| |
Collapse
|
50
|
Kolaski K, Logan LR, Ioannidis JPA. Improving systematic reviews: guidance on guidance and other options and challenges. J Clin Epidemiol 2023; 159:266-273. [PMID: 37196861 DOI: 10.1016/j.jclinepi.2023.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/07/2023] [Accepted: 05/09/2023] [Indexed: 05/19/2023]
Affiliation(s)
- Kat Kolaski
- Department of Orthopaedic Surgery, Wake Forest School of Medicine, Winston-Salem, NC, USA; Department of Pediatrics, Wake Forest School of Medicine, Winston-Salem, NC, USA; Department of Neurology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Lynne Romeiser Logan
- Department of Physical Medicine and Rehabilitation, SUNY Upstate Medical University, Syracuse, NY, USA
| | - John P A Ioannidis
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA; Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA, USA; Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA; Department of Statistics, Stanford University School of Humanities and Sciences, Stanford, CA, USA; Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, CA, USA.
| |
Collapse
|