1
|
Liu S, Bush WS, Akinyemi RO, Byrd GS, Caban-Holt AM, Rajabli F, Reitz C, Kunkle BW, Tosto G, Vance JM, Pericak-Vance M, Haines JL, Williams SM, Crawford DC. Alzheimer disease is (sometimes) highly heritable: Drivers of variation in heritability estimates for binary traits, a systematic review. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.29.25326648. [PMID: 40343016 PMCID: PMC12060970 DOI: 10.1101/2025.04.29.25326648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
Estimating heritability has been fundamental in understanding the genetic contributions to complex disorders like late-onset Alzheimer's disease (LOAD), and provides a rationale for identifying genetic factors associated with disease susceptibility. While numerous studies have established substantial genetic contribution for LOAD, the interpretation of heritability estimates remains challenging. These challenges are further complicated by the binary nature of LOAD status, where estimation and interpretation require additional considerations. Through a systematic review, we identified LOAD heritability estimates from 6 twin studies and 17 genome-wide association studies, all conducted in populations of European ancestry. We demonstrate that these heritability estimates for LOAD vary considerably. The variation reflects not only differences in study design and methodological approaches but also the underlying study population characteristics. Our findings indicate that commonly cited heritability estimates, often treated as universal values, should be interpreted within specific population contexts and methodological frameworks.
Collapse
Affiliation(s)
- Shiying Liu
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - William S Bush
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Rufus Olusola Akinyemi
- Neuroscience and Ageing Research Unit, Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Oyo, Nigeria
| | - Goldie S Byrd
- Maya Angelou Center for Health Equity, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Allison Mercedes Caban-Holt
- Department of Behavioral Science, College of Medicine and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Farid Rajabli
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Christiane Reitz
- Gertrude H. Sergievsky Center, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Department of Neurology Columbia University, Department of Epidemiology, Columbia University, New York, NY, USA
| | - Brian W Kunkle
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Giuseppe Tosto
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, Columbia University; Department of Neurology, Columbia University Irving Medical Center, Columbia University; The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University Irving Medical Center, Columbia University, New York, NY USA
| | - Jeffery M Vance
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Margaret Pericak-Vance
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jonathan L Haines
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Scott M Williams
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Dana C Crawford
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| |
Collapse
|
2
|
Ye C, Li K, Sun W, Jiang Y, Zhang W, Zhang P, Hu YJ, Han Y, Li L. Biological Prior Knowledge-Embedded Deep Neural Network for Plant Genomic Prediction. Genes (Basel) 2025; 16:411. [PMID: 40282370 PMCID: PMC12027452 DOI: 10.3390/genes16040411] [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: 02/24/2025] [Revised: 03/23/2025] [Accepted: 03/26/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: Genomic prediction is a powerful approach that predicts phenotypic traits from genotypic information, enabling the acceleration of trait improvement in plant breeding. Traditional genomic prediction methods have primarily relied on linear mixed models, such as Genomic Best Linear Unbiased Prediction (GBLUP), and conventional machine learning methods like Support Vector Regression (SVR). Traditional methods are limited in handling high-dimensional data and nonlinear relationships. Thus, deep learning methods have also been applied to genomic prediction in recent years. Methods: We proposed iADEP, Integrated Additive, Dominant, and Epistatic Prediction model based on deep learning. Specifically, single nucleotide polymorphism (SNP) data integrating latent genetic interactions and genome-wide association study results as biological prior knowledge are fused to an SNP embedding block, which is then input to a local encoder. The local encoder is fused with an omic-data-incorporated global decoder through a multi-head attention mechanism, followed by multilayer perceptrons. Results: Firstly, we demonstrated through experiments on four datasets that iADEP outperforms existing methods in genotype-to-phenotype prediction. Secondly, we validated the effectiveness of SNP embedding through ablation experiments. Third, we provided an available module for combining other omics data in iADEP and propose a novel method for fusing them. Fourthly, we explored the impact of feature selection on iADEP performance and conclude that utilizing the full set of SNPs generally provides optimal results. Finally, by altering the partition of training and testing sets, we investigated the differences between transductive learning and inductive learning. Conclusions: iADEP provides a new approach for AI breeding, a promising method that integrates biological prior knowledge and enables combination with other omics data.
Collapse
Affiliation(s)
- Chonghang Ye
- Agricultural Bioinformatics Key Laboratory of Hubei Province, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China; (C.Y.); (K.L.); (W.S.); (Y.J.); (P.Z.)
- Hubei Hongshan Laboratory, Wuhan 430070, China;
| | - Kai Li
- Agricultural Bioinformatics Key Laboratory of Hubei Province, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China; (C.Y.); (K.L.); (W.S.); (Y.J.); (P.Z.)
| | - Weicheng Sun
- Agricultural Bioinformatics Key Laboratory of Hubei Province, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China; (C.Y.); (K.L.); (W.S.); (Y.J.); (P.Z.)
| | - Yiwei Jiang
- Agricultural Bioinformatics Key Laboratory of Hubei Province, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China; (C.Y.); (K.L.); (W.S.); (Y.J.); (P.Z.)
| | - Weihan Zhang
- Hubei Hongshan Laboratory, Wuhan 430070, China;
- State Key Laboratory of Plant Diversity and Specialty Crops, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan 430074, China
| | - Ping Zhang
- Agricultural Bioinformatics Key Laboratory of Hubei Province, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China; (C.Y.); (K.L.); (W.S.); (Y.J.); (P.Z.)
- School of Computer, BaoJi University of Arts and Sciences, Baoji 721016, China
| | - Yi-Juan Hu
- Department of Biostatistics, School of Public Health, Peking University, Beijing 100191, China;
- Beijing International Center for Mathematical Research, Peking University, Beijing 100871, China
| | - Yuepeng Han
- Hubei Hongshan Laboratory, Wuhan 430070, China;
- State Key Laboratory of Plant Diversity and Specialty Crops, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan 430074, China
| | - Li Li
- Agricultural Bioinformatics Key Laboratory of Hubei Province, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China; (C.Y.); (K.L.); (W.S.); (Y.J.); (P.Z.)
- Hubei Hongshan Laboratory, Wuhan 430070, China;
| |
Collapse
|
3
|
Yuan M, Goovaerts S, Lee MK, Devine J, Richmond S, Walsh S, Shriver MD, Shaffer JR, Marazita ML, Peeters H, Weinberg SM, Claes P. Optimized phenotyping of complex morphological traits: enhancing discovery of common and rare genetic variants. Brief Bioinform 2025; 26:bbaf090. [PMID: 40062617 PMCID: PMC11891655 DOI: 10.1093/bib/bbaf090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 02/03/2025] [Accepted: 02/18/2025] [Indexed: 05/13/2025] Open
Abstract
Genotype-phenotype (G-P) analyses for complex morphological traits typically utilize simple, predetermined anatomical measures or features derived via unsupervised dimension reduction techniques (e.g. principal component analysis (PCA) or eigen-shapes). Despite the popularity of these approaches, they do not necessarily reveal axes of phenotypic variation that are genetically relevant. Therefore, we introduce a framework to optimize phenotyping for G-P analyses, such as genome-wide association studies (GWAS) of common variants or rare variant association studies (RVAS) of rare variants. Our strategy is two-fold: (i) we construct a multidimensional feature space spanning a wide range of phenotypic variation, and (ii) within this feature space, we use an optimization algorithm to search for directions or feature combinations that are genetically enriched. To test our approach, we examine human facial shape in the context of GWAS and RVAS. In GWAS, we optimize for phenotypes exhibiting high heritability, estimated from either family data or genomic relatedness measured in unrelated individuals. In RVAS, we optimize for the skewness of phenotype distributions, aiming to detect commingled distributions that suggest single or few genomic loci with major effects. We compare our approach with eigen-shapes as baseline in GWAS involving 8246 individuals of European ancestry and in gene-based tests of rare variants with a subset of 1906 individuals. After applying linkage disequilibrium score regression to our GWAS results, heritability-enriched phenotypes yielded the highest SNP heritability, followed by eigen-shapes, while commingling-based traits displayed the lowest SNP heritability. Heritability-enriched phenotypes also exhibited higher discovery rates, identifying the same number of independent genomic loci as eigen-shapes with a smaller effective number of traits. For RVAS, commingling-based traits resulted in more genes passing the exome-wide significance threshold than eigen-shapes, while heritability-enriched phenotypes lead to only a few associations. Overall, our results demonstrate that optimized phenotyping allows for the extraction of genetically relevant traits that can specifically enhance discovery efforts of common and rare variants, as evidenced by their increased power in facial GWAS and RVAS.
Collapse
Affiliation(s)
- Meng Yuan
- Department of Electrical Engineering, ESAT/PSI, KU Leuven, Oude Markt 13, 3000 Leuven, Belgium
- Department of Human Genetics, KU Leuven, Oude Markt 13, 3000 Leuven, Belgium
- Medical Imaging Research Center, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Seppe Goovaerts
- Department of Human Genetics, KU Leuven, Oude Markt 13, 3000 Leuven, Belgium
- Medical Imaging Research Center, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Myoung K Lee
- Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, University of Pittsburgh, Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA 15260, United States
| | - Jay Devine
- Department of Electrical Engineering, ESAT/PSI, KU Leuven, Oude Markt 13, 3000 Leuven, Belgium
- Department of Human Genetics, KU Leuven, Oude Markt 13, 3000 Leuven, Belgium
- Medical Imaging Research Center, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Stephen Richmond
- Applied Clinical Research and Public Health, School of Dentistry, Cardiff University, Cardiff CF10 3AT, United Kingdom
| | - Susan Walsh
- Department of Biology, Indiana University Indianapolis, 420 University Blvd, Indianapolis 46202, IN, United States
| | - Mark D Shriver
- Department of Anthropology, Pennsylvania State University, 201 Old Main, University Park, PA 16802, United States
| | - John R Shaffer
- Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, University of Pittsburgh, Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA 15260, United States
- Department of Human Genetics, University of Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA 15260, United States
| | - Mary L Marazita
- Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, University of Pittsburgh, Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA 15260, United States
- Department of Human Genetics, University of Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA 15260, United States
| | - Hilde Peeters
- Department of Human Genetics, KU Leuven, Oude Markt 13, 3000 Leuven, Belgium
| | - Seth M Weinberg
- Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, University of Pittsburgh, Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA 15260, United States
- Department of Human Genetics, University of Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA 15260, United States
| | - Peter Claes
- Department of Electrical Engineering, ESAT/PSI, KU Leuven, Oude Markt 13, 3000 Leuven, Belgium
- Department of Human Genetics, KU Leuven, Oude Markt 13, 3000 Leuven, Belgium
- Medical Imaging Research Center, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
- Murdoch Children's Research Institute, 50 Flemington Rd, Parkville VIC 3052, Australia
| |
Collapse
|
4
|
Harris L, McDonagh EM, Zhang X, Fawcett K, Foreman A, Daneck P, Sergouniotis PI, Parkinson H, Mazzarotto F, Inouye M, Hollox EJ, Birney E, Fitzgerald T. Genome-wide association testing beyond SNPs. Nat Rev Genet 2025; 26:156-170. [PMID: 39375560 DOI: 10.1038/s41576-024-00778-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2024] [Indexed: 10/09/2024]
Abstract
Decades of genetic association testing in human cohorts have provided important insights into the genetic architecture and biological underpinnings of complex traits and diseases. However, for certain traits, genome-wide association studies (GWAS) for common SNPs are approaching signal saturation, which underscores the need to explore other types of genetic variation to understand the genetic basis of traits and diseases. Copy number variation (CNV) is an important source of heritability that is well known to functionally affect human traits. Recent technological and computational advances enable the large-scale, genome-wide evaluation of CNVs, with implications for downstream applications such as polygenic risk scoring and drug target identification. Here, we review the current state of CNV-GWAS, discuss current limitations in resource infrastructure that need to be overcome to enable the wider uptake of CNV-GWAS results, highlight emerging opportunities and suggest guidelines and standards for future GWAS for genetic variation beyond SNPs at scale.
Collapse
Affiliation(s)
- Laura Harris
- European Molecular Biology Laboratory (EMBL), European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, UK
| | - Ellen M McDonagh
- European Molecular Biology Laboratory (EMBL), European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, UK
| | - Xiaolei Zhang
- European Molecular Biology Laboratory (EMBL), European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, UK
| | - Katherine Fawcett
- European Molecular Biology Laboratory (EMBL), European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, UK
- Department of Population Health Sciences, University of Leicester, Leicester, UK
| | - Amy Foreman
- European Molecular Biology Laboratory (EMBL), European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, UK
| | - Petr Daneck
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Panagiotis I Sergouniotis
- European Molecular Biology Laboratory (EMBL), European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, UK
- Division of Evolution, Infection and Genomics, School of Biological Sciences, University of Manchester, Manchester, UK
| | - Helen Parkinson
- European Molecular Biology Laboratory (EMBL), European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, UK
| | - Francesco Mazzarotto
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Michael Inouye
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Edward J Hollox
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Ewan Birney
- European Molecular Biology Laboratory (EMBL), European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, UK
| | - Tomas Fitzgerald
- European Molecular Biology Laboratory (EMBL), European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, UK.
| |
Collapse
|
5
|
Canida T, Ke H, Chen S, Ye Z, Ma T. Multivariate Bayesian variable selection for multi-trait genetic fine mapping. J R Stat Soc Ser C Appl Stat 2025; 74:331-351. [PMID: 40092670 PMCID: PMC11905884 DOI: 10.1093/jrsssc/qlae055] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/21/2024] [Accepted: 10/01/2024] [Indexed: 03/19/2025]
Abstract
Genome-wide association studies (GWAS) have identified thousands of single-nucleotide polymorphisms (SNPs) associated with complex traits, but determining the underlying causal variants remains challenging. Fine mapping aims to pinpoint the potentially causal variants from a large number of correlated SNPs possibly with group structure in GWAS-enriched genomic regions using variable selection approaches. In multi-trait fine mapping, we are interested in identifying the causal variants for multiple related traits. Existing multivariate variable selection methods for fine mapping select variables for all responses without considering the possible heterogeneity across different responses. Here, we develop a novel multivariate Bayesian variable selection method for multi-trait fine mapping to select causal variants from a large number of grouped SNPs that target at multiple correlated and possibly heterogeneous traits. Our new method is featured by its selection at multiple levels, incorporation of prior biological knowledge to guide selection and identification of best subset of traits the variants target at. We showed the advantage of our method over existing methods via comprehensive simulations that mimic typical fine-mapping settings and a real-world fine-mapping example in UK Biobank, where we identified critical causal variants potentially targeting at different subsets of addictive behaviours and risk factors.
Collapse
Affiliation(s)
- Travis Canida
- Department of Epidemiology and Biostatistics, University of Maryland, 4200 Valley Drive, College Park, MD 20742, USA
| | - Hongjie Ke
- Department of Epidemiology and Biostatistics, University of Maryland, 4200 Valley Drive, College Park, MD 20742, USA
| | - Shuo Chen
- Department of Epidemiology and Public Health, University of Maryland, 655 W. Baltimore Street, Baltimore, MD 21201, USA
| | - Zhenyao Ye
- Department of Epidemiology and Public Health, University of Maryland, 655 W. Baltimore Street, Baltimore, MD 21201, USA
| | - Tianzhou Ma
- Department of Epidemiology and Biostatistics, University of Maryland, 4200 Valley Drive, College Park, MD 20742, USA
| |
Collapse
|
6
|
Mostafaei Dehnavi M, Damerum A, Taheri S, Ebadi A, Panahi S, Hodgin G, Brandley B, Salami SA, Taylor G. Population genomics of a natural Cannabis sativa L. collection from Iran identifies novel genetic loci for flowering time, morphology, sex and chemotyping. BMC PLANT BIOLOGY 2025; 25:80. [PMID: 39838336 PMCID: PMC11748290 DOI: 10.1186/s12870-025-06045-4] [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: 07/31/2024] [Accepted: 01/01/2025] [Indexed: 01/23/2025]
Abstract
BACKGROUND Future breeding and selection of Cannabis sativa L. for both drug production and industrial purposes require a source of germplasm with wide genetic variation, such as that found in wild relatives and progenitors of highly cultivated plants. Limited directional selection and breeding have occurred in this crop, especially informed by molecular markers. RESULTS This study investigated the population genomics of a natural cannabis collection comprising male and female individuals from various climatic zones in Iran. Using Genotyping-By-Sequencing (GBS), we sequenced 228 individuals from 35 populations. The data obtained enabled an association analysis, linking genotypes with key phenotypes such as inflorescence characteristics, flowering time, plant morphology, tetrahydrocannabinol (THC) and cannabidiol (CBD) content, and sex. We detected approximately 23,266 significant high-quality Single Nucleotide Polymorphisms (SNPs), establishing associations between markers and traits. The population structure analysis revealed that Iranian cannabis plants fall into five distinct groups. Additionally, a comparison with global data suggested that the Iranian populations is distinctive and generally closer to marijuana than to hemp, with some populations showing a closer affinity to hemp. The GWAS identified novel genetic loci associated with sex, yield, and chemotype traits in cannabis, which had not been previously reported. CONCLUSION The study's findings highlight the distinct genetic structure of Iranian Cannabis populations. The identification of novel genetic loci associated with important traits suggests potential targets for future breeding programs. This research underscores the value of the Iranian cannabis germplasm as a resource for breeding and selection efforts aimed at improving Cannabis for various uses.
Collapse
Affiliation(s)
- Mahboubeh Mostafaei Dehnavi
- Department of Plant Sciences, University of California, Davis, CA, USA
- Department of Horticultural Science, Faculty of Agriculture, University of Tehran, Karaj, Iran
| | - Annabelle Damerum
- Department of Plant Sciences, University of California, Davis, CA, USA
- Present address, Zymo Research Corp, Irvine, CA, USA
| | - Sadegh Taheri
- Department of Animal Science, Faculty of Agriculture, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Ali Ebadi
- Department of Horticultural Science, Faculty of Agriculture, University of Tehran, Karaj, Iran
| | - Shadab Panahi
- Department of Horticultural Science, Faculty of Agriculture, University of Tehran, Karaj, Iran
| | - George Hodgin
- Biopharmaceutical Research Company, Castroville, CA, USA
| | - Brian Brandley
- Biopharmaceutical Research Company, Castroville, CA, USA
| | - Seyed Alireza Salami
- Department of Horticultural Science, Faculty of Agriculture, University of Tehran, Karaj, Iran.
- Industrial and Medical Cannabis Research Institute (IMCRI), Tehran, Iran.
| | - Gail Taylor
- Department of Plant Sciences, University of California, Davis, CA, USA.
| |
Collapse
|
7
|
Zhang H, Dai J, Mu Q, Zhao X, Lin Z, Wang K, Wang M, Sun D. Macrophage heterogeneity and oncogenic mechanisms in lung adenocarcinoma: insights from scRNA-seq analysis and predictive modeling. Front Immunol 2025; 15:1491872. [PMID: 39850883 PMCID: PMC11754191 DOI: 10.3389/fimmu.2024.1491872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 12/03/2024] [Indexed: 01/25/2025] Open
Abstract
Background Macrophages play a dual role in the tumor microenvironment(TME), capable of secreting pro-inflammatory factors to combat tumors while also promoting tumor growth through angiogenesis and immune suppression. This study aims to explore the characteristics of macrophages in lung adenocarcinoma (LUAD) and establish a prognostic model based on macrophage-related genes. Method We performed scRNA-seq analysis to investigate macrophage heterogeneity and their potential pseudotime evolutionary processes. Specifically, we used scRNA-seq data processing, intercellular communication analysis, pseudotime trajectory analysis, and transcription factor regulatory analysis to reveal the complexity of macrophage subpopulations. Data from The Cancer Genome Atlas (TCGA) was used to assess the impact of various macrophage subtypes on LUAD prognosis. Univariate Cox regression was applied to select prognostic-related genes from macrophage markers. We constructed a prognostic model using Lasso regression and multivariate Cox regression, categorizing LUAD patients into high and low-risk groups based on the median risk score. The model's performance was validated across multiple external datasets. We also examined differences between high and low-risk groups in terms of pathway enrichment, mutation information, tumor microenvironment(TME), and immunotherapy efficacy. Finally, RT-PCR confirmed the expression of model genes in LUAD, and cellular experiments explored the carcinogenic mechanism of COL5A1. Results We found that signals such as SPP1 and MIF were more active in tumor tissues, indicating potential oncogenic roles of macrophages. Using macrophage marker genes, we developed a robust prognostic model for LUAD that effectively predicts prognosis and immunotherapy efficacy. A nomogram was constructed to predict LUAD prognosis based on the model's risk score and other clinical features. Differences between high and low-risk groups in terms of TME, enrichment analysis, mutational landscape, and immunotherapy efficacy were systematically analyzed. RT-PCR and cellular experiments supported the oncogenic role of COL5A1. Conclusion Our study identified potential oncogenic mechanisms of macrophages and their impact on LUAD prognosis. We developed a prognostic model based on macrophage marker genes, demonstrating strong performance in predicting prognosis and immunotherapy efficacy. Finally, cellular experiments suggested COL5A1 as a potential therapeutic target for LUAD.
Collapse
Affiliation(s)
- Han Zhang
- Tianjin Chest Hospital, Tianjin University, Tianjin, China
| | | | - Qiuqiao Mu
- Clinical School of Thoracic, Tianjin Medical University, Tianjin, China
| | - Xiaojiang Zhao
- Clinical School of Thoracic, Tianjin Medical University, Tianjin, China
| | - Ziao Lin
- OmixScience Research Institute, OmixScience Co., Ltd., Hangzhou, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
| | - Kai Wang
- Tianjin Chest Hospital, Tianjin University, Tianjin, China
| | - Meng Wang
- Tianjin Chest Hospital, Tianjin University, Tianjin, China
- Clinical School of Thoracic, Tianjin Medical University, Tianjin, China
| | - Daqiang Sun
- Tianjin Chest Hospital, Tianjin University, Tianjin, China
| |
Collapse
|
8
|
Yan FY, Xu YF, Feng WR, He QH, Hua GA, Li WJ, Xu P, Zhou J, Tang YK. Genomic analysis of hypoxia-tolerant population of the Chinese mitten crab (Eriocheir sinensis). FISH & SHELLFISH IMMUNOLOGY 2024; 154:109931. [PMID: 39343063 DOI: 10.1016/j.fsi.2024.109931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 09/01/2024] [Accepted: 09/26/2024] [Indexed: 10/01/2024]
Abstract
Hypoxic stress, triggered by a multitude of factors, has inflicted significant economic repercussions on the aquaculture of Eriocheir sinensis. In this research, we sequenced a collective of 60 samples from both hypoxia-sensitive and hypoxia-resistant groups utilizing streamlined genome sequencing techniques. Subsequently, we delved into population evolution, scrutinized the selective sweep within these populations, and performed a genome-wide association study (GWAS) focused on the hypoxia tolerance traits within the population, all through the lens of SNPs molecular markers. This comprehensive analysis aimed to uncover the SNPs and pinpoint the pertinent candidate genes that influence the hypoxia tolerance capabilities of E. sinensis. The selective sweep analysis revealed that genes harboring potential genetic variations within the two populations were predominantly enriched in areas such as signaling molecules and interactions, energy metabolism, glycolipid metabolism, and immune response. In the genome-wide association study focusing on hypoxia tolerance traits, we identified four SNPs significantly associated with hypoxia resistance. Furthermore, one potential candidate gene, Dscam2, which is believed to influence hypoxia tolerance, was discovered within a 50 kb vicinity of these SNPs. These identified SNPs can serve as molecular markers for screening hypoxia tolerance, offering valuable insights for the genetic improvement of E. sinensis.
Collapse
Affiliation(s)
- Feng-Yuan Yan
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China; Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China
| | - Yuan-Feng Xu
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China
| | - Wen-Rong Feng
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China
| | - Qing-Hong He
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China; Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China
| | - Guo-An Hua
- Jiangsu Haorun Biological Industry Group Co., Ltd, Taizhou, 225500, China
| | - Wen-Jing Li
- Jiangsu Haorun Biological Industry Group Co., Ltd, Taizhou, 225500, China
| | - Pao Xu
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China
| | - Jun Zhou
- Freshwater Fisheries Research Institute of Jiangsu Province, Nanjing, 210017, China.
| | - Yong-Kai Tang
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China; Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China.
| |
Collapse
|
9
|
薛 恩, 陈 曦, 王 雪, 王 斯, 王 梦, 李 劲, 秦 雪, 武 轶, 李 楠, 李 静, 周 治, 朱 洪, 吴 涛, 陈 大, 胡 永. [Single nucleotide polymorphism heritability of non-syndromic cleft lip with or without cleft palate in Chinese population]. BEIJING DA XUE XUE BAO. YI XUE BAN = JOURNAL OF PEKING UNIVERSITY. HEALTH SCIENCES 2024; 56:775-780. [PMID: 39397453 PMCID: PMC11480541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Indexed: 10/15/2024]
Abstract
OBJECTIVE To delve into the intricate relationship between common genetic variations across the entire genome and the risk of non-syndromic cleft lip with or without cleft palate (NSCL/P). METHODS Utilizing summary statistics data from genome-wide association studies (GWAS), a thorough investigation to evaluate the impact of common variations on the genome were undertook. This involved assessing single nucleotide polymorphism (SNP) heritability across the entire genome, as well as within specific genomic regions. To ensure the robustness of our analysis, stringent quality control measures were applied to the GWAS summary statistics data. Criteria for inclusion encompassed the absence of missing values, a minor allele frequency ≥1%, P-values falling within the range of 0 to 1, and clear SNP strand orientation. SNP meeting these stringent criteria were then meticulously included in our analysis. The SNP heritability of NSCL/P was calculated using linkage disequilibrium score regression. Additionally, hierarchical linkage disequilibrium score regression to partition SNP heritability within coding regions, promoters, introns, enhancers, and super enhancers were employed, and the enrichment levels within different genomic regions using LDSC (v1.0.1) software were further elucidated. RESULTS Our study drew upon GWAS summary statistics data obtained from 806 NSCL/P trios, comprising a total of 2 418 individuals from the Chinese population. Following rigorous quality control procedures, 490 593 out of 492 993 SNP were deemed suitable for inclusion in SNP heritability calculations. The observed SNP heritability of NSCL/P was 0.55 (95%CI: 0.28-0.82). Adjusting for the elevated disease pre-valence within our sample, the SNP heritability scaled down to 0.37 (95%CI: 0.19-0.55) based on the prevalence observed in the general Chinese population. Notably, our enrichment analysis unveiled significant enrichment of SNP heritability within enhancer regions (15.70, P=0.04) and super enhancer regions (3.18, P=0.03). CONCLUSION Our study sheds light on the intricate interplay between common genetic variations and the risk of NSCL/P in the Chinese population. By elucidating the SNP heritability landscape across different genomic regions, we contribute valuable insights into the genetic basis of NSCL/P. The significant enrichment of SNP heritability within enhancer and super enhancer regions underscores the potential role of these regulatory elements in shaping the genetic susceptibility to NSCL/P. This paves the way for further research aimed at uncovering novel genetic pathogenic factors underlying NSCL/P pathogenesis.
Collapse
Affiliation(s)
- 恩慈 薛
- 北京大学公共卫生学院流行病与卫生统计学系,北京 100191Department of Epidemiology and Biostatistics, Peking University School of Public Health, Beijing 100191, China
| | - 曦 陈
- 北京大学公共卫生学院流行病与卫生统计学系,北京 100191Department of Epidemiology and Biostatistics, Peking University School of Public Health, Beijing 100191, China
| | - 雪珩 王
- 北京大学公共卫生学院流行病与卫生统计学系,北京 100191Department of Epidemiology and Biostatistics, Peking University School of Public Health, Beijing 100191, China
| | - 斯悦 王
- 北京大学公共卫生学院流行病与卫生统计学系,北京 100191Department of Epidemiology and Biostatistics, Peking University School of Public Health, Beijing 100191, China
| | - 梦莹 王
- 北京大学公共卫生学院流行病与卫生统计学系,北京 100191Department of Epidemiology and Biostatistics, Peking University School of Public Health, Beijing 100191, China
| | - 劲 李
- 北京大学公共卫生学院流行病与卫生统计学系,北京 100191Department of Epidemiology and Biostatistics, Peking University School of Public Health, Beijing 100191, China
| | - 雪英 秦
- 北京大学公共卫生学院流行病与卫生统计学系,北京 100191Department of Epidemiology and Biostatistics, Peking University School of Public Health, Beijing 100191, China
| | - 轶群 武
- 北京大学公共卫生学院流行病与卫生统计学系,北京 100191Department of Epidemiology and Biostatistics, Peking University School of Public Health, Beijing 100191, China
| | - 楠 李
- 北京大学口腔医学院口腔颌面外科,北京 100081Department of Oral and Maxillofacial Surgery, Peking University School of Stomatology, Beijing 100081, China
| | - 静 李
- 北京大学口腔医学院儿童口腔科,北京 100081Department of Pediatric Dentistry, Peking University School of Stomatology, Beijing 100081, China
| | - 治波 周
- 北京大学口腔医学院口腔颌面外科,北京 100081Department of Oral and Maxillofacial Surgery, Peking University School of Stomatology, Beijing 100081, China
| | - 洪平 朱
- 北京大学口腔医学院口腔颌面外科,北京 100081Department of Oral and Maxillofacial Surgery, Peking University School of Stomatology, Beijing 100081, China
| | - 涛 吴
- 北京大学公共卫生学院流行病与卫生统计学系,北京 100191Department of Epidemiology and Biostatistics, Peking University School of Public Health, Beijing 100191, China
| | - 大方 陈
- 北京大学公共卫生学院流行病与卫生统计学系,北京 100191Department of Epidemiology and Biostatistics, Peking University School of Public Health, Beijing 100191, China
| | - 永华 胡
- 北京大学公共卫生学院流行病与卫生统计学系,北京 100191Department of Epidemiology and Biostatistics, Peking University School of Public Health, Beijing 100191, China
| |
Collapse
|
10
|
Kember RL, Davis CN, Feuer KL, Kranzler HR. Considerations for the application of polygenic scores to clinical care of individuals with substance use disorders. J Clin Invest 2024; 134:e172882. [PMID: 39403926 PMCID: PMC11473164 DOI: 10.1172/jci172882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024] Open
Abstract
Substance use disorders (SUDs) are highly prevalent and associated with excess morbidity, mortality, and economic costs. Thus, there is considerable interest in the early identification of individuals who may be more susceptible to developing SUDs and in improving personalized treatment decisions for those who have SUDs. SUDs are known to be influenced by both genetic and environmental factors. Polygenic scores (PGSs) provide a single measure of genetic liability that could be used as a biomarker in predicting disease development, progression, and treatment response. Although PGSs are rapidly being integrated into clinical practice, there is little information to guide clinicians in their responsible use and interpretation. In this Review, we discuss the potential benefits and pitfalls of the use of PGSs in the clinical care of SUDs, highlighting current research. We also provide suggestions for important considerations prior to implementing the clinical use of PGSs and recommend future directions for research.
Collapse
|
11
|
Zhang X, Lin G, Zhang Q, Wu H, Xu W, Wang Z, He Z, Su L, Zhuang Y, Gong A. The rs3918188 and rs1799983 loci of eNOS gene are associated with susceptibility in patients with systemic lupus erythematosus in Northeast China. Sci Rep 2024; 14:20803. [PMID: 39242633 PMCID: PMC11379712 DOI: 10.1038/s41598-024-70711-0] [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: 10/20/2023] [Accepted: 08/20/2024] [Indexed: 09/09/2024] Open
Abstract
To investigate the association between single nucleotide polymorphism (SNP) at the rs3918188, rs1799983 and rs1007311 loci of the endothelial nitric oxide synthase (eNOS) gene and genetic susceptibility to systemic lupus erythematosus (SLE) in northeastern China. The base distribution of eNOS gene rs3918188, rs1799983 and rs1007311 in 1712 human peripheral blood samples from Northeast China was detected by SNaPshot sequencing technology. The correlation between genotype, allele and gene model of these loci of the eNOS gene and the genetic susceptibility to SLE was investigated by logistic regression analysis. The results of the differences in the frequency distribution of their gene models were visualised using R 4.3.2 software. Finally, HaploView 4.2 software was used to analyse the relationship between the haplotypes of the three loci mentioned above and the genetic susceptibility to SLE. A multifactor dimensionality reduction (MDR) analysis was used to determine the best SNP-SNP interaction model. The CC genotype and C allele at the rs3918188 locus may be a risk factor for SLE (CC vs AA: OR = 1.827, P < 0.05; C vs A: OR = 1.558, P < 0.001), and this locus increased the risk of SLE in the dominant model and the recessive model (AC + CC vs AA: OR = 1.542, P < 0.05; CC vs AA + AC: OR = 1.707, P < 0.001), while the risk of SLE was reduced in the overdominant model (AC vs AA + CC: OR = 0.628, P < 0.001). The GT genotype and T allele at locus rs1799983 may be a protective factor for SLE (GT vs GG: OR = 0.328, P < 0.001; T vs G: OR = 0.438, P < 0.001) and this locus reduced the risk of SLE in the overdominant model (GT vs GG + TT: OR = 0.385, P < 0.001). There is a strong linkage disequilibrium between the rs1007311 and rs1799983 loci of the eNOS gene. Among them, the formed haplotype AG increased the risk of SLE compared to GG. AT and GT decreased the risk of SLE compared to GG. In this study, the eNOS gene rs3918188 and rs1799983 loci were found to be associated with susceptibility to SLE. This helps to deeply explore the mechanism of eNOS gene and genetic susceptibility to SLE. It provides a certain research basis for the subsequent exploration of the molecular mechanism of these loci and SLE, as well as the early diagnosis, treatment and prognosis of SLE.
Collapse
Affiliation(s)
- Xuan Zhang
- College of Traditional Chinese Medicine, Hainan Medical University, Haikou, 571199, China
| | - Guiling Lin
- College of Traditional Chinese Medicine, Hainan Medical University, Haikou, 571199, China
| | - Qi Zhang
- Heilongjiang Academy of Chinese Medicine, Harbin, 150036, Heilongjiang, China
| | - Huitao Wu
- Heilongjiang Academy of Chinese Medicine, Harbin, 150036, Heilongjiang, China
| | - Wenlu Xu
- College of Traditional Chinese Medicine, Hainan Medical University, Haikou, 571199, China
| | - Zhe Wang
- College of Traditional Chinese Medicine, Hainan Medical University, Haikou, 571199, China
| | - Ziman He
- Heilongjiang Academy of Chinese Medicine, Harbin, 150036, Heilongjiang, China
| | - Linglan Su
- Heilongjiang Academy of Chinese Medicine, Harbin, 150036, Heilongjiang, China
| | - Yanping Zhuang
- International Research Center for Aging and Cancer, Hainan Medical University, Haikou, 571199, Hainan, China.
| | - Aimin Gong
- College of Traditional Chinese Medicine, Hainan Medical University, Haikou, 571199, China.
| |
Collapse
|
12
|
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
|
13
|
Lin WY. Gene-Environment Interactions and Gene-Gene Interactions on Two Biological Age Measures: Evidence from Taiwan Biobank Participants. Adv Biol (Weinh) 2024; 8:e2400149. [PMID: 38684452 DOI: 10.1002/adbi.202400149] [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: 03/16/2024] [Revised: 04/14/2024] [Indexed: 05/02/2024]
Abstract
PhenoAge and BioAge are two commonly used biological age (BA) measures. The author here searched for gene-environment interactions (GxE) and gene-gene interactions (GxG) on PhenoAgeAccel (age-adjusted PhenoAge) and BioAgeAccel (age-adjusted BioAge) of 111,996 Taiwan Biobank (TWB) participants, including a discovery set of 86,536 TWB2 individuals and a replication set of 25,460 TWB1 individuals. Searching for variance quantitative trait loci (vQTLs) provides a convenient way to evaluate GxE and GxG. A total of 4 nearly independent (linkage disequilibrium measure r2 < 0.01) PhenoAgeAccel-vQTLs are identified from 5,303,039 autosomal TWB2 SNPs (p < 5E-8), whereas no vQTLs are found from BioAgeAccel. These 4 PhenoAgeAccel-vQTLs (rs35276921, rs141927875, rs10903013, and rs76038336) are further replicated by TWB1 (p < 5E-8). They are located in the OR51B5, FAM234A, and AXIN1 genes. All 4 PhenoAgeAccel-vQTLs are significantly associated with PhenoAgeAccel (p < 5E-8). A phylogenetic heat map of the GxE analyses showed that smoking exacerbated the PhenoAgeAccel-vQTLs' aging effects, while higher educational attainment attenuated the PhenoAgeAccel-vQTLs' aging effects. Body mass index, chronological age, alcohol consumption, and sex do not prominently modulate PhenoAgeAccel-vQTLs' aging effects. Based on these vQTL results, rs141927875-rs35276921 interaction (p = 4.7E-61) and rs76038336-rs10903013 interaction (p = 3.3E-116) on PhenoAgeAccel are detected.
Collapse
Affiliation(s)
- Wan-Yu Lin
- Institute of Health Data Analytics and Statistics, College of Public Health, National Taiwan University, Taipei, 100, Taiwan
- Master of Public Health Degree Program, College of Public Health, National Taiwan University, Taipei, 100, Taiwan
| |
Collapse
|
14
|
Aghogho CI, Kayondo SI, Eleblu SJY, Ige A, Asante I, Offei SK, Parkes E, Egesi C, Mbanjo EGN, Shah T, Kulakow P, Rabbi IY. Genome-wide association study for yield and quality of granulated cassava processed product. THE PLANT GENOME 2024; 17:e20469. [PMID: 38880944 DOI: 10.1002/tpg2.20469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 04/27/2024] [Accepted: 05/03/2024] [Indexed: 06/18/2024]
Abstract
The starchy storage roots of cassava are commonly processed into a variety of products, including cassava granulated processed products (gari). The commercial value of cassava roots depends on the yield and quality of processed products, directly influencing the acceptance of new varieties by farmers, processors, and consumers. This study aims to estimate genetic advance through phenotypic selection and identify genomic regions associated and candidate genes linked with gari yield and quality. Higher single nucleotide polymorphism (SNP)-based heritability estimates compared to broad-sense heritability estimates were observed for most traits highlighting the influence of genetic factors on observed variation. Using genome-wide association analysis of 188 clones, genotyped using 53,150 genome-wide SNPs, nine SNPs located on seven chromosomes were significantly associated with peel loss, gari yield, color parameters for gari and eba, bulk density, swelling index, and textural properties of eba. Future research will focus on validating and understanding the functions of identified genes and their influence on gari yield and quality traits.
Collapse
Affiliation(s)
- Cynthia Idhigu Aghogho
- West Africa Centre for Crop Improvement (WACCI), College of Basic and Applied Sciences, University of Ghana, Legon, Ghana
- International Institute of Tropical Agriculture (IITA), Ibadan, Nigeria
| | - Siraj Ismail Kayondo
- International Institute of Tropical Agriculture (IITA), Eastern Africa Hub, Dar es Salaam, Tanzania
| | - Saviour J Y Eleblu
- West Africa Centre for Crop Improvement (WACCI), College of Basic and Applied Sciences, University of Ghana, Legon, Ghana
| | - Adenike Ige
- Department of Agronomy and Plant Genetics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Isaac Asante
- West Africa Centre for Crop Improvement (WACCI), College of Basic and Applied Sciences, University of Ghana, Legon, Ghana
| | - Samuel K Offei
- West Africa Centre for Crop Improvement (WACCI), College of Basic and Applied Sciences, University of Ghana, Legon, Ghana
| | - Elizabeth Parkes
- International Institute of Tropical Agriculture (IITA), Ibadan, Nigeria
| | - Chiedozie Egesi
- National Root Crops Research Institute, Umuahia, Nigeria
- Plant Breeding and Genetics Section, School of Integrative Plant Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, New York, USA
| | | | - Trushar Shah
- International Institute of Tropical Agriculture (IITA), c/o ILRI, Nairobi, Kenya
| | - Peter Kulakow
- International Institute of Tropical Agriculture (IITA), Ibadan, Nigeria
| | - Ismail Y Rabbi
- International Institute of Tropical Agriculture (IITA), Ibadan, Nigeria
| |
Collapse
|
15
|
Lu X, Suo L, Yan X, Li W, Su Y, Zhou B, Liu C, Yang L, Wang J, Ji D, Cuomu R, Cuoji A, Gui B, Wang Z, Jiang W, Wu Y, Su R. Genome-wide association analysis of fleece traits in Northwest Xizang white cashmere goat. Front Vet Sci 2024; 11:1409084. [PMID: 38872797 PMCID: PMC11171727 DOI: 10.3389/fvets.2024.1409084] [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: 03/29/2024] [Accepted: 05/07/2024] [Indexed: 06/15/2024] Open
Abstract
Northwest Xizang White Cashmere Goat (NXWCG) is the first new breed of cashmere goat in the Xizang Autonomous Region. It has significant characteristics of extremely high fineness, gloss, and softness. Genome-wide association analysis is an effective biological method used to measure the consistency and correlation of genotype changes between two molecular markers in the genome. In addition, it can screen out the key genes affecting the complex traits of biological individuals. The aim of this study was to analyze the genetic mechanism of cashmere trait variation in NXWCG and to discover SNP locus and key genes closely related to traits such as superfine cashmere. Additionally, the key genes near the obtained significant SNPs were analyzed by gene function annotation and biological function mining. In this study, the phenotype data of the four traits (cashmere length, fiber length, cashmere diameter, and cashmere production) were collected. GGP_Goat_70K SNP chip was used for genotyping the ear tissue DNA of the experimental group. Subsequently, the association of phenotype data and genotype data was performed using Gemma-0.98.1 software. A linear mixed model was used for the association study. The results showed that four fleece traits were associated with 18 significant SNPs at the genome level and 232 SNPs at the chromosome level, through gene annotated from Capra hircus genome using assembly ARS1. A total of 107 candidate genes related to fleece traits were obtained. Combined with Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analysis, we can find that CLNS1A, CCSER1, RPS6KC1, PRLR, KCNRG, KCNK9, and CLYBL can be used as important candidate genes for fleece traits of NXWCG. We used Sanger sequencing and suitability chi-square test to further verify the significant loci and candidate genes screened by GWAS, and the results show that the base mutations loci on the five candidate genes, CCSER1 (snp12579, 34,449,796, A → G), RPS6KC1 (snp41503, 69,173,527, A → G), KCNRG (snp41082, 67,134,820, G → A), KCNK9 (14:78472665, 78,472,665, G → A), and CLYBL (12: 9705753, 9,705,753, C → T), significantly affect the fleece traits of NXWCG. The results provide a valuable basis for future research and contribute to a better understanding of the genetic structure variation of the goat.
Collapse
Affiliation(s)
- Xiaotian Lu
- Institute of Animal Science, Xizang Academy of Agricultural and Animal Husbandry Science, Lhasa, China
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Langda Suo
- Institute of Animal Science, Xizang Academy of Agricultural and Animal Husbandry Science, Lhasa, China
- Sino-Arabian Joint Laboratory of Sheep and Goat Germplasm Innovation, Hohhot, China
| | - Xiaochun Yan
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Wenze Li
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Yixin Su
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Bohan Zhou
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Can Liu
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Lepu Yang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Jiayin Wang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - De Ji
- Institute of Animal Science, Xizang Academy of Agricultural and Animal Husbandry Science, Lhasa, China
- Key Laboratory of Animal Genetics and Breeding on Xizang Plateau, Ministry of Agriculture and Rural Affairs, Lhasa, China
| | - Renqing Cuomu
- Institute of Animal Science, Xizang Academy of Agricultural and Animal Husbandry Science, Lhasa, China
- Key Laboratory of Animal Genetics and Breeding on Xizang Plateau, Ministry of Agriculture and Rural Affairs, Lhasa, China
| | - Awang Cuoji
- Institute of Animal Science, Xizang Academy of Agricultural and Animal Husbandry Science, Lhasa, China
- Key Laboratory of Animal Genetics and Breeding on Xizang Plateau, Ministry of Agriculture and Rural Affairs, Lhasa, China
| | - Ba Gui
- Institute of Animal Science, Xizang Academy of Agricultural and Animal Husbandry Science, Lhasa, China
- Key Laboratory of Animal Genetics and Breeding on Xizang Plateau, Ministry of Agriculture and Rural Affairs, Lhasa, China
| | - Zhiying Wang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Wei Jiang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Yujiang Wu
- Institute of Animal Science, Xizang Academy of Agricultural and Animal Husbandry Science, Lhasa, China
- Key Laboratory of Animal Genetics and Breeding on Xizang Plateau, Ministry of Agriculture and Rural Affairs, Lhasa, China
| | - Rui Su
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| |
Collapse
|
16
|
Fong WJ, Tan HM, Garg R, Teh AL, Pan H, Gupta V, Krishna B, Chen ZH, Purwanto NY, Yap F, Tan KH, Chan KYJ, Chan SY, Goh N, Rane N, Tan ESE, Jiang Y, Han M, Meaney M, Wang D, Keppo J, Tan GCY. Comparing feature selection and machine learning approaches for predicting CYP2D6 methylation from genetic variation. Front Neuroinform 2024; 17:1244336. [PMID: 38449836 PMCID: PMC10915285 DOI: 10.3389/fninf.2023.1244336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 10/18/2023] [Indexed: 03/08/2024] Open
Abstract
Introduction Pharmacogenetics currently supports clinical decision-making on the basis of a limited number of variants in a few genes and may benefit paediatric prescribing where there is a need for more precise dosing. Integrating genomic information such as methylation into pharmacogenetic models holds the potential to improve their accuracy and consequently prescribing decisions. Cytochrome P450 2D6 (CYP2D6) is a highly polymorphic gene conventionally associated with the metabolism of commonly used drugs and endogenous substrates. We thus sought to predict epigenetic loci from single nucleotide polymorphisms (SNPs) related to CYP2D6 in children from the GUSTO cohort. Methods Buffy coat DNA methylation was quantified using the Illumina Infinium Methylation EPIC beadchip. CpG sites associated with CYP2D6 were used as outcome variables in Linear Regression, Elastic Net and XGBoost models. We compared feature selection of SNPs from GWAS mQTLs, GTEx eQTLs and SNPs within 2 MB of the CYP2D6 gene and the impact of adding demographic data. The samples were split into training (75%) sets and test (25%) sets for validation. In Elastic Net model and XGBoost models, optimal hyperparameter search was done using 10-fold cross validation. Root Mean Square Error and R-squared values were obtained to investigate each models' performance. When GWAS was performed to determine SNPs associated with CpG sites, a total of 15 SNPs were identified where several SNPs appeared to influence multiple CpG sites. Results Overall, Elastic Net models of genetic features appeared to perform marginally better than heritability estimates and substantially better than Linear Regression and XGBoost models. The addition of nongenetic features appeared to improve performance for some but not all feature sets and probes. The best feature set and Machine Learning (ML) approach differed substantially between CpG sites and a number of top variables were identified for each model. Discussion The development of SNP-based prediction models for CYP2D6 CpG methylation in Singaporean children of varying ethnicities in this study has clinical application. With further validation, they may add to the set of tools available to improve precision medicine and pharmacogenetics-based dosing.
Collapse
Affiliation(s)
- Wei Jing Fong
- Computational Biology, National University of Singapore, Singapore, Singapore
| | - Hong Ming Tan
- Computational Biology, National University of Singapore, Singapore, Singapore
| | - Rishabh Garg
- Computational Biology, National University of Singapore, Singapore, Singapore
| | - Ai Ling Teh
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Hong Pan
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Varsha Gupta
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Bernadus Krishna
- Computational Biology, National University of Singapore, Singapore, Singapore
| | - Zou Hui Chen
- Computational Biology, National University of Singapore, Singapore, Singapore
| | | | - Fabian Yap
- KK Women's and Children's Hospital, Singapore, Singapore
| | - Kok Hian Tan
- KK Women's and Children's Hospital, Singapore, Singapore
- Duke NUS Medical School, Singapore, Singapore
| | - Kok Yen Jerry Chan
- KK Women's and Children's Hospital, Singapore, Singapore
- Duke NUS Medical School, Singapore, Singapore
| | - Shiao-Yng Chan
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- National University Hospital, Singapore, Singapore
| | | | - Nikita Rane
- Institute of Mental Health,Singapore, Singapore
| | | | | | - Mei Han
- Computational Biology, National University of Singapore, Singapore, Singapore
| | - Michael Meaney
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Dennis Wang
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Jussi Keppo
- Computational Biology, National University of Singapore, Singapore, Singapore
| | - Geoffrey Chern-Yee Tan
- Computational Biology, National University of Singapore, Singapore, Singapore
- Institute of Mental Health,Singapore, Singapore
| |
Collapse
|
17
|
Sofer T, Granot-Hershkovitz E, Tarraf W, Filigrana P, Isasi CR, Suglia SF, Kaplan R, Taylor K, Daviglus ML, Testai FD, Zeng D, Cai J, Fornage M, González HM, DeCarli C. Intracranial Volume Is Driven by Both Genetics and Early Life Exposures: The SOL-INCA-MRI Study. Ethn Dis 2024; 34:103-112. [PMID: 38973806 PMCID: PMC11223032 DOI: 10.18865/ed.34.2.103] [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: 07/09/2024] Open
Abstract
Intracranial volume (ICV) reflects maximal brain development and is associated with later-life cognitive abilities. We quantified ICV among first- and second-generation Hispanic and Latino adults from the Study of Latinos-Investigation of Cognitive Aging - MRI (SOL-INCA-MRI), estimated ICV heritability, and tested its associations with previously reported genetic variants, both individually and as a genetic risk score (GRS). We also estimated the association of ICV with early life environmental measures: nativity or age of immigration and parental education. The estimated heritability of ICV was 19% (95% CI, 0.1%-56%) in n=1781 unrelated SOL-INCA-MRI individuals. Four of 10 tested genetic variants were associated with ICV and an increase of 1 SD of the ICV-GRS was associated with an increase of 10.37 cm3 in the ICV (95% CI, 5.29-15.45). Compared to being born in the continental United States, immigrating to the United States at age 11 years or older was associated with 24 cm3 smaller ICV (95% CI, -39.97 to -8.06). Compared to both parents having less than high-school education, at least 1 parent completing high-school education was associated with 15.4 cm3 greater ICV (95% CI, 4.46-26.39). These data confirm the importance of early life health on brain development.
Collapse
Affiliation(s)
- Tamar Sofer
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Boston, MA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Einat Granot-Hershkovitz
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Wassim Tarraf
- Institute of Gerontology, Wayne State University, Detroit, MI
| | - Paola Filigrana
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Carmen R. Isasi
- Department of Epidemiology & Population Health, Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY
| | - Shakira F. Suglia
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Robert Kaplan
- Department of Epidemiology & Population Health, Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle WA
| | - Kent Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Martha L. Daviglus
- Department of Medicine, Institute for Minority Health Research, University of Illinois at Chicago, IL
| | - Fernando D. Testai
- Department of Neurology, University of Illinois at Chicago College of Medicine, Chicago, IL
| | - Donglin Zeng
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC
| | - Jianwen Cai
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC
| | - Myriam Fornage
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX
| | - Hector M. González
- Department of Neurosciences and Shiley-Marcos Alzheimer’s Disease Center, University of California, San Diego, La Jolla, CA
| | - Charles DeCarli
- Alzheimer’s Disease Research Center, Department of Neurology, University of California, Davis, Sacramento, CA
| |
Collapse
|
18
|
So HC, Xue X, Ma Z, Sham PC. SumVg: Total Heritability Explained by All Variants in Genome-Wide Association Studies Based on Summary Statistics with Standard Error Estimates. Int J Mol Sci 2024; 25:1347. [PMID: 38279346 PMCID: PMC10816209 DOI: 10.3390/ijms25021347] [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: 10/13/2023] [Revised: 01/15/2024] [Accepted: 01/16/2024] [Indexed: 01/28/2024] Open
Abstract
Genome-wide association studies (GWAS) are commonly employed to study the genetic basis of complex traits/diseases, and a key question is how much heritability could be explained by all single nucleotide polymorphisms (SNPs) in GWAS. One widely used approach that relies on summary statistics only is linkage disequilibrium score regression (LDSC); however, this approach requires certain assumptions about the effects of SNPs (e.g., all SNPs contribute to heritability and each SNP contributes equal variance). More flexible modeling methods may be useful. We previously developed an approach recovering the "true" effect sizes from a set of observed z-statistics with an empirical Bayes approach, using only summary statistics. However, methods for standard error (SE) estimation are not available yet, limiting the interpretation of our results and the applicability of the approach. In this study, we developed several resampling-based approaches to estimate the SE of SNP-based heritability, including two jackknife and three parametric bootstrap methods. The resampling procedures are performed at the SNP level as it is most common to estimate heritability from GWAS summary statistics alone. Simulations showed that the delete-d-jackknife and parametric bootstrap approaches provide good estimates of the SE. In particular, the parametric bootstrap approaches yield the lowest root-mean-squared-error (RMSE) of the true SE. We also explored various methods for constructing confidence intervals (CIs). In addition, we applied our method to estimate the SNP-based heritability of 12 immune-related traits (levels of cytokines and growth factors) to shed light on their genetic architecture. We also implemented the methods to compute the sum of heritability explained and the corresponding SE in an R package SumVg. In conclusion, SumVg may provide a useful alternative tool for calculating SNP heritability and estimating SE/CI, which does not rely on distributional assumptions of SNP effects.
Collapse
Affiliation(s)
- Hon-Cheong So
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China; (X.X.); (Z.M.)
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research of Common Diseases, Kunming Institute of Zoology and The Chinese University of Hong Kong, Shatin, Hong Kong, China
- Department of Psychiatry, The Chinese University of Hong Kong, Shatin, Hong Kong, China
- CUHK Shenzhen Research Institute, Shenzhen 518057, China
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism, The Chinese University of Hong Kong, Shatin, Hong Kong, China
- Hong Kong Branch of the Chinese Academy of Sciences Center for Excellence in Animal Evolution and Genetics, The Chinese University of Hong Kong, Shatin, Hong Kong, China
- Brain and Mind Institute, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Xiao Xue
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China; (X.X.); (Z.M.)
| | - Zhijie Ma
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China; (X.X.); (Z.M.)
| | - Pak-Chung Sham
- Department of Psychiatry, The University of Hong Kong, Pokfulam, Hong Kong, China;
| |
Collapse
|
19
|
Li H, Mazumder R, Lin X. Accurate and efficient estimation of local heritability using summary statistics and the linkage disequilibrium matrix. Nat Commun 2023; 14:7954. [PMID: 38040712 PMCID: PMC10692177 DOI: 10.1038/s41467-023-43565-9] [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: 04/17/2023] [Accepted: 11/14/2023] [Indexed: 12/03/2023] Open
Abstract
Existing SNP-heritability estimators that leverage summary statistics from genome-wide association studies (GWAS) are much less efficient (i.e., have larger standard errors) than the restricted maximum likelihood (REML) estimators which require access to individual-level data. We introduce a new method for local heritability estimation-Heritability Estimation with high Efficiency using LD and association Summary Statistics (HEELS)-that significantly improves the statistical efficiency of summary-statistics-based heritability estimator and attains comparable statistical efficiency as REML (with a relative statistical efficiency >92%). Moreover, we propose representing the empirical LD matrix as the sum of a low-rank matrix and a banded matrix. We show that this way of modeling the LD can not only reduce the storage and memory cost, but also improve the computational efficiency of heritability estimation. We demonstrate the statistical efficiency of HEELS and the advantages of our proposed LD approximation strategies both in simulations and through empirical analyses of the UK Biobank data.
Collapse
Affiliation(s)
- Hui Li
- Harvard T.H. Chan School of Public Health, Department of Biostatistics, Boston, MA, USA
| | - Rahul Mazumder
- Massachusetts Institute of Technology, Operations Research and Statistics group, Cambridge, MA, USA
| | - Xihong Lin
- Harvard T.H. Chan School of Public Health, Department of Biostatistics, Boston, MA, USA.
- Harvard University, Department of Statistics, Cambridge, MA, USA.
| |
Collapse
|
20
|
Chen R, Liu J, Zhang Y, Cai W, Zhang X, Xu Y, Dou X, Wang Z, Han D, Wang J, Lin G, Wang L, Sun Y, Bai Z, Gu M, Wang Z. Association analysis between reproduction genes INHA, PGR, RARG with lamb and other traits of Liaoning cashmere goats. Anim Biotechnol 2023; 34:2094-2105. [PMID: 35622393 DOI: 10.1080/10495398.2022.2077212] [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] [Indexed: 11/01/2022]
Abstract
Reproductive traits have a high economic value in goat breeding, and increasing the number of lambs produced by ewes is of great importance to improve the production efficiency of goat farming. Lambing traits in goats are low heritability traits, but their genetic basis is ultimately determined by genes. This study aimed to investigate the relationship between INHA, RARG, and PGR gene polymorphisms and production performance, such as lambing, cashmere production, milk production, and body size in Liaoning cashmere goats. A total of six single nucleotide polymorphisms (SNPs) loci were identified in these three genes, G144A and T504C on the INHA gene, A56G, G144A, G490C on the RARG gene, and G109519T on the PGR gene. For lambing and cashmere production traits, the AA genotype of G144A on the INHA gene, TT on the T504C genotype, GG genotype of G144A on the INHA gene, A56G, G144A, and T504C on RARG and G109519T on PGR gene are dominant genotypes. AATT is a dominant haplotype combination. Allele G can be used as a molecular marker for lambing, cashmere, and milk production traits in Liaoning cashmere goats. Marker-assisted selection can be used for early selection to achieve improvement of genetic traits in Liaoning cashmere goats.
Collapse
Affiliation(s)
- Rui Chen
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Jichang Liu
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Yurou Zhang
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Weidong Cai
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Xinjiang Zhang
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Yanan Xu
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Xingtang Dou
- Liaoning Province Modern Agricultural Production Base Construction Engineering Center, Shenyang, China
| | - Zhanhong Wang
- Liaoning Province Modern Agricultural Production Base Construction Engineering Center, Shenyang, China
| | - Di Han
- Liaoning Province Modern Agricultural Production Base Construction Engineering Center, Shenyang, China
| | - Jiaming Wang
- Liaoning Province Modern Agricultural Production Base Construction Engineering Center, Shenyang, China
| | - Guangyu Lin
- Liaoning Province Modern Agricultural Production Base Construction Engineering Center, Shenyang, China
| | - Lingling Wang
- Liaoning Province Modern Agricultural Production Base Construction Engineering Center, Shenyang, China
| | - Yinggang Sun
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Zhixian Bai
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Ming Gu
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Zeying Wang
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| |
Collapse
|
21
|
Sallam AM, Abou-Souliman I, Reyer H, Wimmers K, Rabee AE. New insights into the genetic predisposition of brucellosis and its effect on the gut and vaginal microbiota in goats. Sci Rep 2023; 13:20086. [PMID: 37973848 PMCID: PMC10654701 DOI: 10.1038/s41598-023-46997-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 11/07/2023] [Indexed: 11/19/2023] Open
Abstract
Goats contribute significantly to the global food security and industry. They constitute a main supplier of meat and milk for large proportions of people in Egypt and worldwide. Brucellosis is a zoonotic infectious disease that causes a significant economic loss in animal production. A case-control genome-wide association analysis (GWAS) was conducted using the infectious status of the animal as a phenotype. The does that showed abortion during the last third period of pregnancy and which were positive to both rose bengal plate and serum tube agglutination tests, were considered as cases. Otherwise, they were considered as controls. All animals were genotyped using the Illumina 65KSNP BeadChip. Additionally, the diversity and composition of vaginal and fecal microbiota in cases and controls were investigated using PCR-amplicone sequencing of the V4 region of 16S rDNA. After applying quality control criteria, 35,818 markers and 66 does were available for the GWAS test. The GWAS revealed a significantly associated SNP (P = 5.01 × 10-7) located on Caprine chromosome 15 at 29 megabases. Four other markers surpassed the proposed threshold (P = 2.5 × 10-5). Additionally, fourteen genomic regions accounted for more than 0.1% of the variance explained by all genome windows. Corresponding markers were located within or in close vicinity to several candidate genes, such as ARRB1, RELT, ATG16L2, IGSF21, UBR4, ULK1, DCN, MAPB1, NAIP, CD26, IFIH1, NDFIP2, DOK4, MAF, IL2RB, USP18, ARID5A, ZAP70, CNTN5, PIK3AP1, DNTT, BLNK, and NHLRC3. These genes play important roles in the regulation of immune responses to the infections through several biological pathways. Similar vaginal bacterial community was observed in both cases and controls while the fecal bacterial composition and diversity differed between the groups (P < 0.05). Faeces from the control does showed a higher relative abundance of the phylum Bacteroidota compared to cases (P < 0.05), while the latter showed more Firmicutes, Spirochaetota, Planctomycetota, and Proteobacteria. On the genus level, the control does exhibited higher abundances of Rikenellaceae RC9 gut group and Christensenellaceae R-7 group (P < 0.05), while the infected does revealed higher Bacteroides, Alistipes, and Prevotellaceae UCG-003 (P < 0.05). This information increases our understanding of the genetics of the susceptibility to Brucella in goats and may be useful in breeding programs and selection schemes that aim at controlling the disease in livestock.
Collapse
Affiliation(s)
- Ahmed M Sallam
- Animal and Poultry Breeding Department, Desert Research Center, Cairo, Egypt.
| | | | - Henry Reyer
- Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Klaus Wimmers
- Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Alaa Emara Rabee
- Animal and Poultry Nutrition Department, Desert Research Center, Cairo, Egypt
| |
Collapse
|
22
|
Stamp J, DenAdel A, Weinreich D, Crawford L. Leveraging the genetic correlation between traits improves the detection of epistasis in genome-wide association studies. G3 (BETHESDA, MD.) 2023; 13:jkad118. [PMID: 37243672 PMCID: PMC10484060 DOI: 10.1093/g3journal/jkad118] [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: 01/11/2023] [Revised: 01/11/2023] [Accepted: 05/23/2023] [Indexed: 05/29/2023]
Abstract
Epistasis, commonly defined as the interaction between genetic loci, is known to play an important role in the phenotypic variation of complex traits. As a result, many statistical methods have been developed to identify genetic variants that are involved in epistasis, and nearly all of these approaches carry out this task by focusing on analyzing one trait at a time. Previous studies have shown that jointly modeling multiple phenotypes can often dramatically increase statistical power for association mapping. In this study, we present the "multivariate MArginal ePIstasis Test" (mvMAPIT)-a multioutcome generalization of a recently proposed epistatic detection method which seeks to detect marginal epistasis or the combined pairwise interaction effects between a given variant and all other variants. By searching for marginal epistatic effects, one can identify genetic variants that are involved in epistasis without the need to identify the exact partners with which the variants interact-thus, potentially alleviating much of the statistical and computational burden associated with conventional explicit search-based methods. Our proposed mvMAPIT builds upon this strategy by taking advantage of correlation structure between traits to improve the identification of variants involved in epistasis. We formulate mvMAPIT as a multivariate linear mixed model and develop a multitrait variance component estimation algorithm for efficient parameter inference and P-value computation. Together with reasonable model approximations, our proposed approach is scalable to moderately sized genome-wide association studies. With simulations, we illustrate the benefits of mvMAPIT over univariate (or single-trait) epistatic mapping strategies. We also apply mvMAPIT framework to protein sequence data from two broadly neutralizing anti-influenza antibodies and approximately 2,000 heterogeneous stock of mice from the Wellcome Trust Centre for Human Genetics. The mvMAPIT R package can be downloaded at https://github.com/lcrawlab/mvMAPIT.
Collapse
Affiliation(s)
- Julian Stamp
- Center for Computational Molecular Biology, Brown University, Providence, RI 02906, USA
| | - Alan DenAdel
- Center for Computational Molecular Biology, Brown University, Providence, RI 02906, USA
| | - Daniel Weinreich
- Center for Computational Molecular Biology, Brown University, Providence, RI 02906, USA
- Department of Ecology, Evolution, and Organismal Biology, Brown University, Providence, RI 02906, USA
| | - Lorin Crawford
- Center for Computational Molecular Biology, Brown University, Providence, RI 02906, USA
- Department of Biostatistics, Brown University, Providence, RI 02903, USA
- Microsoft Research New England, Cambridge, MA 02142, USA
| |
Collapse
|
23
|
Yang J, Wu J. Discovery of potential biomarkers for osteoporosis diagnosis by individual omics and multi-omics technologies. Expert Rev Mol Diagn 2023:1-16. [PMID: 37140363 DOI: 10.1080/14737159.2023.2208750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
INTRODUCTION Global aging has made osteoporosis an increasingly serious public health problem. Osteoporotic fractures seriously affect the quality of life of patients and increase disability and mortality rates. Early diagnosis is important for timely intervention. The continuous development of individual- and multi-omics methods is helpful for the exploration and discovery of biomarkers for the diagnosis of osteoporosis. AREAS COVERED In this review, we first introduce the epidemiological status of osteoporosis and then describe the pathogenesis of osteoporosis. Furthermore, the latest progress in individual- and multi-omics technologies for exploring biomarkers for osteoporosis diagnosis is summarized. Moreover, we clarify the advantages and disadvantages of the application of osteoporosis biomarkers obtained using the omics method. Finally, we put forward valuable views on the future research direction of diagnostic biomarkers of osteoporosis. EXPERT OPINION Omics methods undoubtedly provide greatly contribute to the exploration of diagnostic biomarkers of osteoporosis; however, in the future, the clinical validity and clinical utility of the obtained potential biomarkers should be thoroughly examined. In addition, the improvement and optimization of the detection methods for different types of biomarkers and standardization of the detection process guarantee the reliability and accuracy of the detection results.
Collapse
Affiliation(s)
- Jing Yang
- Department of Clinical Laboratory Medicine, Beijing Jishuitan Hospital, Peking University, Beijing, China
| | - Jun Wu
- Department of Clinical Laboratory Medicine, Beijing Jishuitan Hospital, Peking University, Beijing, China
| |
Collapse
|
24
|
Zimmermann E, Distl O. SNP-Based Heritability of Osteochondrosis Dissecans in Hanoverian Warmblood Horses. Animals (Basel) 2023; 13:ani13091462. [PMID: 37174498 PMCID: PMC10177438 DOI: 10.3390/ani13091462] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/24/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Before the genomics era, heritability estimates were performed using pedigree data. Data collection for pedigree analysis is time consuming and holds the risk of incorrect or incomplete data. With the availability of SNP-based arrays, heritability can now be estimated based on genotyping data. We used SNP array and 1.6 million imputed genotype data with different minor allele frequency restrictions to estimate heritabilities for osteochondrosis dissecans in the fetlock, hock and stifle joints of 446 Hanoverian warmblood horses. SNP-based heritabilities were estimated using a genomic restricted maximum likelihood (GREML) method and accounting for patterns of regional linkage disequilibrium in the equine genome. In addition, we employed GREML for family data to account for different degrees of relatedness in the study population. Our results indicate that we were able to capture a larger proportion of additive genetic variance compared to pedigree-based estimates in the same population of Hanoverian horses. Heritability estimates on the linear scale for fetlock-, hock- and stifle-osteochondrosis dissecans were 0.41-0.43, 0.62-0.63, and 0.23-0.25, respectively, with standard errors of 0.11-0.14. Accounting for linkage disequilibrium patterns had an upward effect on the imputed data and a downward impact on the SNP array genotype data. GREML for family data resulted in higher heritability estimates for fetlock-osteochondrosis dissecans and slightly higher estimates for hock-osteochondrosis dissecans, but had no effect on stifle-osteochondrosis dissecans. The largest and most consistent heritability estimates were obtained when we employed GREML for family data with genomic relationship matrices weighted through patterns of regional linkage disequilibrium. Estimation of SNP-based heritability should be recommended for traits that can only be phenotyped in smaller samples or are cost-effective.
Collapse
Affiliation(s)
- Elisa Zimmermann
- Institute for Animal Breeding and Genetics, University of Veterinary Medicine Hannover (Foundation), 30559 Hannover, Germany
| | - Ottmar Distl
- Institute for Animal Breeding and Genetics, University of Veterinary Medicine Hannover (Foundation), 30559 Hannover, Germany
| |
Collapse
|
25
|
Barry CJS, Walker VM, Cheesman R, Davey Smith G, Morris TT, Davies NM. How to estimate heritability: a guide for genetic epidemiologists. Int J Epidemiol 2023; 52:624-632. [PMID: 36427280 PMCID: PMC10114051 DOI: 10.1093/ije/dyac224] [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: 05/03/2022] [Accepted: 11/14/2022] [Indexed: 11/27/2022] Open
Abstract
Traditionally, heritability has been estimated using family-based methods such as twin studies. Advancements in molecular genomics have facilitated the development of methods that use large samples of (unrelated or related) genotyped individuals. Here, we provide an overview of common methods applied in genetic epidemiology to estimate heritability, i.e. the proportion of phenotypic variation explained by genetic variation. We provide a guide to key genetic concepts required to understand heritability estimation methods from family-based designs (twin and family studies), genomic designs based on unrelated individuals [linkage disequilibrium score regression, genomic relatedness restricted maximum-likelihood (GREML) estimation] and family-based genomic designs (sibling regression, GREML-kinship, trio-genome-wide complex trait analysis, maternal-genome-wide complex trait analysis, relatedness disequilibrium regression). We describe how heritability is estimated for each method and the assumptions underlying its estimation, and discuss the implications when these assumptions are not met. We further discuss the benefits and limitations of estimating heritability within samples of unrelated individuals compared with samples of related individuals. Overall, this article is intended to help the reader determine the circumstances when each method would be appropriate and why.
Collapse
Affiliation(s)
- Ciarrah-Jane S Barry
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Venexia M Walker
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, USA
| | - Rosa Cheesman
- PROMENTA Research Center, Department of Psychology, University of Oslo, Oslo, Norway
| | - George Davey Smith
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Tim T Morris
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Neil M Davies
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
26
|
Abstract
Prior to the development of genome-wide arrays and whole genome sequencing technologies, heritability estimation mainly relied on the study of related individuals. Over the past decade, various approaches have been developed to estimate SNP-based narrow-sense heritability (h SNP 2 ${\rm{h}}_{{\rm{SNP}}}^2$ ) in unrelated individuals. These latter approaches use either individual-level genetic variations or summary results from genome-wide association studies (GWAS). Recently, several studies compared these approaches using extensive simulations and empirical datasets. However, sparse information on hands-on training necessitates revisiting these approaches from the perspective of a stepwise guide for practical applications. Here, we provide an overview of the commonly used SNP-heritability estimation approaches utilizing genome-wide array, imputed or whole genome data from unrelated individuals, or summary results. We not only discuss these approaches based on their statistical concepts, utility, advantages, and limitations, but also provide step-by-step protocols to apply these approaches. For illustration purposes, we estimateh SNP 2 ${\rm{h}}_{{\rm{SNP}}}^2$ of height and BMI utilizing individual-level data from The Northern Finland Birth Cohort (NFBC) and summary results from the Genetic Investigation of ANthropometric Traits (GIANT;) consortium. We present this review as a template for the researchers who estimate and use heritability in their studies and as a reference for geneticists who develop or extend heritability estimation approaches. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: GREML (GCTA) Alternate Protocol 1: Stratified GREML Basic Protocol 2: LDAK Alternate Protocol 2: Stratified LDAK Basic Protocol 3: Threshold GREML Basic Protocol 4: LD score (LDSC) regression Basic Protocol 5: SumHer.
Collapse
Affiliation(s)
- Amit K. Srivastava
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, USA; The Center for Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children’s Hospital Medical Center, USA; March of Dimes Prematurity Research Center Ohio Collaborative, USA; Department of Pediatrics, University of Cincinnati College of Medicine, USA
| | - Scott M. Williams
- Department of Population and Quantitative Health Sciences, School of Medicine, Case Western Reserve University, USA; Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, USA; Institute of Computational Biology, Case Western Reserve University, USA
| | - Ge Zhang
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, USA; The Center for Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children’s Hospital Medical Center, USA; March of Dimes Prematurity Research Center Ohio Collaborative, USA; Department of Pediatrics, University of Cincinnati College of Medicine, USA
| |
Collapse
|
27
|
Li H, Mazumder R, Lin X. Accurate and Efficient Estimation of Local Heritability using Summary Statistics and LD Matrix. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.08.527759. [PMID: 36798290 PMCID: PMC9934676 DOI: 10.1101/2023.02.08.527759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Existing SNP-heritability estimation methods that leverage GWAS summary statistics produce estimators that are less efficient than the restricted maximum likelihood (REML) estimator using individual-level data under linear mixed models (LMMs). Increasing the precision of a heritability estimator is particularly important for regional analyses, as local genetic variances tend to be small. We introduce a new estimator for local heritability, "HEELS", which attains comparable statistical efficiency as REML (\emph{i.e.} relative efficiency greater than 92%) but only requires summary-level statistics -- Z-scores from the marginal association tests plus the empirical LD matrix. HEELS significantly improves the statistical efficiency of the existing summary-statistics-based heritability estimators-- for instance, HEELS produces heritability estimates that are more than 3-fold and 7-times less variable than GRE and LDSC, respectively. Moreover, we introduce a unified framework to evaluate and compare the performance of different LD approximation strategies. We propose representing the empirical LD as the sum of a low-rank matrix and a banded matrix. This approximation not only reduces the storage and memory cost of using the LD matrix, but also improves the computational efficiency of the HEELS estimation. We demonstrate the statistical efficiency of HEELS and the advantages of our proposed LD approximation strategies both in simulations and through empirical analyses of the UK Biobank data.
Collapse
|
28
|
Rostamzadeh Mahdabi E, Tian R, Li Y, Wang X, Zhao M, Li H, Yang D, Zhang H, Li S, Esmailizadeh A. Genomic heritability and correlation between carcass traits in Japanese Black cattle evaluated under different ceilings of relatedness among individuals. Front Genet 2023; 14:1053291. [PMID: 36816045 PMCID: PMC9928846 DOI: 10.3389/fgene.2023.1053291] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 01/18/2023] [Indexed: 02/04/2023] Open
Abstract
The investigation of carcass traits to produce meat with high efficiency has been in focus on Japanese Black cattle since 1972. To implement a successful breeding program in carcass production, a comprehensive understanding of genetic characteristics and relationships between the traits is of paramount importance. In this study, genomic heritability and genomic correlation between carcass traits, including carcass weight (CW), rib eye area (REA), rib thickness (RT), subcutaneous fat thickness (SFT), yield rate (YI), and beef marbling score (BMS) were estimated using the genomic data of 9,850 Japanese Black cattle (4,142 heifers and 5,708 steers). In addition, we investigated the effect of genetic relatedness degree on the estimation of genetic parameters of carcass traits in sub-populations created based on different GRM-cutoff values. Genome-based restricted maximum likelihood (GREML) analysis was applied to estimate genetic parameters. Using all animal data, the heritability values for carcass traits were estimated as moderate to relatively high magnitude, ranging from 0.338 to 0.509 with standard errors, ranging from 0.014 to 0.015. The genetic correlations were obtained low and negative between SFT and REA [-0.198 (0.034)] and between SFT and BMS [-0.096 (0.033)] traits, and high and negative between SFT and YI [-0.634 (0.022)]. REA trait was genetically highly correlated with YI and BMS [0.811 (0.012) and 0.625 (0.022), respectively]. In sub-populations created based on the genetic-relatedness ceiling, the heritability estimates ranged from 0.212 (0.131) to 0.647 (0.066). At the genetic-relatedness ceiling of 0.15, the correlation values between most traits with low genomic correlation were overestimated while the correlations between the traits with relatively moderate to high correlations, ranging from 0.380 to 0.811, were underestimated. The values were steady at the ceilings of 0.30-0.95 (sample size of 5,443-9,850) for most of the highly correlated traits. The results demonstrated that there is considerable genetic variation and also favorable genomic correlations between carcass traits. Therefore, the genetic improvement for the traits can be simultaneously attained through genomic selection. In addition, we observed that depending on the degree of relationship between individuals and sample size, the genomic heritability and correlation estimates for carcass traits may be different.
Collapse
Affiliation(s)
| | - Rugang Tian
- Institute of Animal Husbandry, Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Hohhot, China
| | - Yuan Li
- Institute of Animal Husbandry, Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Hohhot, China
| | - Xiao Wang
- Institute of Animal Husbandry, Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Hohhot, China
| | - Meng Zhao
- Institute of Animal Husbandry, Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Hohhot, China
| | - Hui Li
- Institute of Animal Husbandry, Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Hohhot, China
| | - Ding Yang
- Institute of Animal Husbandry, Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Hohhot, China
| | - Hao Zhang
- Institute of Animal Husbandry, Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Hohhot, China
| | - SuFan Li
- Institute of Animal Husbandry, Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Hohhot, China
| | - Ali Esmailizadeh
- Department of Animal Science, Faculty of Agriculture, Shahid Bahonar University of Kerman, Kerman, Iran
| |
Collapse
|
29
|
Genetic Polymorphisms Associated with Prothrombin Time and Activated Partial Thromboplastin Time in Chinese Healthy Population. Genes (Basel) 2022; 13:genes13101867. [PMID: 36292752 PMCID: PMC9602091 DOI: 10.3390/genes13101867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 09/30/2022] [Accepted: 10/12/2022] [Indexed: 11/04/2022] Open
Abstract
(1) Background: The purpose of this study was to evaluate the effect of gene polymorphisms on prothrombin time (PT) and activated partial thromboplastin time (APTT) in a healthy Chinese population. (2) Methods: A total of 403 healthy volunteers from a series of novel oral anticoagulants (NOACs) bioequivalence trials in China were included. Coagulation tests for PT and APTT were performed in the central lab at Peking University First Hospital. Whole-exome sequencing (WES) and genome-wide association analysis were performed. (3) Results: In the correlation analysis of PT, 105 SNPs from 84 genes reached the genome-wide significance threshold (p < 1 × 10−5). Zinc Finger Protein 594 (ZNF594) rs184838268 (p = 4.50 × 10−19) was most significantly related to PT, and Actinin Alpha 1 (ACTN1) was found to interact most with other candidate genes. Significant associations with previously reported candidate genes Aurora Kinase B (AURKB), Complement C5(C5), Clock Circadian Regulator (CLOCK), and Histone Deacetylase 9(HDAC9) were detected in our dataset (p < 1 × 10−5). PiggyBac Transposable Element Derived 2(PGBD2) rs75935520 (p = 4.49 × 10−6), Bromodomain Adjacent To Zinc Finger Domain 2A(BAZ2A) rs199970765 (p = 5.69 × 10−6) and Protogenin (PRTG) rs80064850 (p = 8.69 × 10−6) were significantly correlated with APTT (p < 1 × 10−5). The heritability values of PT and APTT were 0.83 and 0.64, respectively; (4) Conclusion: The PT and APTT of healthy populations are affected by genetic polymorphisms. ZNF594 and ACTN1 variants could be novel genetic markers of PT, while PRTG polymorphisms might be associated with APTT levels. The findings could be attributed to ethnic differences, and need further investigation.
Collapse
|
30
|
Olasege BS, Porto-Neto LR, Tahir MS, Gouveia GC, Cánovas A, Hayes BJ, Fortes MRS. Correlation scan: identifying genomic regions that affect genetic correlations applied to fertility traits. BMC Genomics 2022; 23:684. [PMID: 36195838 PMCID: PMC9533527 DOI: 10.1186/s12864-022-08898-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 09/19/2022] [Indexed: 11/10/2022] Open
Abstract
Although the genetic correlations between complex traits have been estimated for more than a century, only recently we have started to map and understand the precise localization of the genomic region(s) that underpin these correlations. Reproductive traits are often genetically correlated. Yet, we don't fully understand the complexities, synergism, or trade-offs between male and female fertility. In this study, we used reproductive traits in two cattle populations (Brahman; BB, Tropical Composite; TC) to develop a novel framework termed correlation scan (CS). This framework was used to identify local regions associated with the genetic correlations between male and female fertility traits. Animals were genotyped with bovine high-density single nucleotide polymorphisms (SNPs) chip assay. The data used consisted of ~1000 individual records measured through frequent ovarian scanning for age at first corpus luteum (AGECL) and a laboratory assay for serum levels of insulin growth hormone (IGF1 measured in bulls, IGF1b, or cows, IGF1c). The methodology developed herein used correlations of 500-SNP effects in a 100-SNPs sliding window in each chromosome to identify local genomic regions that either drive or antagonize the genetic correlations between traits. We used Fisher's Z-statistics through a permutation method to confirm which regions of the genome harboured significant correlations. About 30% of the total genomic regions were identified as driving and antagonizing genetic correlations between male and female fertility traits in the two populations. These regions confirmed the polygenic nature of the traits being studied and pointed to genes of interest. For BB, the most important chromosome in terms of local regions is often located on bovine chromosome (BTA) 14. However, the important regions are spread across few different BTA's in TC. Quantitative trait loci (QTLs) and functional enrichment analysis revealed many significant windows co-localized with known QTLs related to milk production and fertility traits, especially puberty. In general, the enriched reproductive QTLs driving the genetic correlations between male and female fertility are the same for both cattle populations, while the antagonizing regions were population specific. Moreover, most of the antagonizing regions were mapped to chromosome X. These results suggest regions of chromosome X for further investigation into the trade-offs between male and female fertility. We compared the CS with two other recently proposed methods that map local genomic correlations. Some genomic regions were significant across methods. Yet, many significant regions identified with the CS were overlooked by other methods.
Collapse
Affiliation(s)
- Babatunde S Olasege
- The University of Queensland, School of Chemistry and Molecular Biosciences, Saint Lucia Campus, Brisbane, QLD, 4072, Australia.,CSIRO Agriculture and Food, Saint Lucia, QLD, 4067, Australia
| | | | - Muhammad S Tahir
- The University of Queensland, School of Chemistry and Molecular Biosciences, Saint Lucia Campus, Brisbane, QLD, 4072, Australia.,CSIRO Agriculture and Food, Saint Lucia, QLD, 4067, Australia
| | - Gabriela C Gouveia
- Animal Science Department, Veterinary School, Federal University of Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Angela Cánovas
- Department of Animal Biosciences, Centre for Genetic Improvement of Livestock, University of Guelph, 50 Stone Rd E, Guelph, ON, N1G 2W1, Canada
| | - Ben J Hayes
- The University of Queensland, Queensland Alliance for Agriculture and Food Innovation (QAAFI), Saint Lucia Campus, Brisbane, QLD, 4072, Australia
| | - Marina R S Fortes
- The University of Queensland, School of Chemistry and Molecular Biosciences, Saint Lucia Campus, Brisbane, QLD, 4072, Australia. .,The University of Queensland, Queensland Alliance for Agriculture and Food Innovation (QAAFI), Saint Lucia Campus, Brisbane, QLD, 4072, Australia.
| |
Collapse
|
31
|
Pattee J, Vanderlinden LA, Mahaffey S, Hoffman P, Tabakoff B, Saba LM. Evaluation and characterization of expression quantitative trait analysis methods in the Hybrid Rat Diversity Panel. Front Genet 2022; 13:947423. [PMID: 36186443 PMCID: PMC9515987 DOI: 10.3389/fgene.2022.947423] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/26/2022] [Indexed: 01/07/2023] Open
Abstract
The Hybrid Rat Diversity Panel (HRDP) is a stable and well-characterized set of more than 90 inbred rat strains that can be leveraged for systems genetics approaches to understanding the genetic and genomic variation associated with complex disease. The HRDP exhibits substantial between-strain diversity while retaining substantial within-strain isogenicity, allowing for the precise mapping of genetic variation associated with complex phenotypes and providing statistical power to identify associated variants. In order to robustly identify associated genetic variants, it is important to account for the population structure induced by inbreeding. To this end, we investigate the performance of four plausible approaches towards modeling quantitative traits in the HRDP and quantify their operating characteristics. In particular, we investigate three approaches based on genome-wide mixed model analysis, and one approach based on ordinary least squares linear regression. Towards facilitating study planning and design, we conduct extensive simulations to investigate the power of genetic association analyses in the HRDP, and characterize the impressive attained power. In simulation of eQTL data in the HRDP, we find that a mixed model approach that leverages leave-one-chromosome-out kinship estimation attains the highest power while controlling type I error.
Collapse
Affiliation(s)
- Jack Pattee
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Lauren A. Vanderlinden
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Spencer Mahaffey
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Paula Hoffman
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States,Department of Pharmacology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Boris Tabakoff
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Laura M. Saba
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States,*Correspondence: Laura M. Saba,
| |
Collapse
|
32
|
Dao C, Jiang J, Paul D, Zhao H. Variance Estimation and Confidence Intervals from Genome-wide Association Studies Through High-dimensional Misspecified Mixed Model Analysis. J Stat Plan Inference 2022; 220:15-23. [PMID: 37089275 PMCID: PMC10121196 DOI: 10.1016/j.jspi.2022.01.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
We study variance estimation and associated confidence intervals for parameters characterizing genetic effects from genome-wide association studies (GWAS) in misspecified mixed model analysis. Previous studies have shown that, in spite of the model misspecification, certain quantities of genetic interests are consistently estimable, and consistent estimators of these quantities can be obtained using the restricted maximum likelihood (REML) method under a misspecified linear mixed model. However, the asymptotic variance of such a REML estimator is complicated and not ready to be implemented for practical use. In this paper, we develop practical and computationally convenient methods for estimating such asymptotic variances and constructing the associated confidence intervals. Performance of the proposed methods is evaluated empirically based on Monte-Carlo simulations and real-data application.
Collapse
|
33
|
Moradi MH, Mahmodi R, Farahani AHK, Karimi MO. Genome-wide evaluation of copy gain and loss variations in three Afghan sheep breeds. Sci Rep 2022; 12:14286. [PMID: 35996004 PMCID: PMC9395407 DOI: 10.1038/s41598-022-18571-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 08/16/2022] [Indexed: 11/23/2022] Open
Abstract
Copy number variation (CNV) is one of the main sources of variation between different individuals that has recently attracted much researcher interest as a major source for heritable variation in complex traits. The aim of this study was to identify CNVs in Afghan indigenous sheep consisting of three Arab, Baluchi, and Gadik breeds using genomic arrays containing 53,862 single nucleotide polymorphism (SNP) markers. Data were analyzed using the Hidden Markov Model (HMM) of PennCNV software. In this study, out of 45 sheep studied, 97.8% (44 animals) have shown CNVs. In total, 411 CNVs were observed for autosomal chromosomes and the entire sequence length of around 144 Mb was identified across the genome. The average number of CNVs per each sheep was 9.13. The identified CNVs for Arab, Baluchi, and Gadik breeds were 306, 62, and 43, respectively. After merging overlapped regions, a total of 376 copy number variation regions (CNVR) were identified, which are 286, 50, and 40 for Arab, Baluchi, and Gadik breeds, respectively. Bioinformatics analysis was performed to identify the genes and QTLs reported in these regions and the biochemical pathways involved by these genes. The results showed that many of these CNVRs overlapped with the genes or QTLs that are associated with various pathways such as immune system development, growth, reproduction, and environmental adaptions. Furthermore, to determine a genome-wide pattern of selection signatures in Afghan sheep breeds, the unbiased estimates of FST was calculated and the results indicated that 37 of the 376 CNVRs (~ 10%) have been also under selection signature, most of those overlapped with the genes influencing production, reproduction and immune system. Finally, the statistical methods used in this study was applied in an external dataset including 96 individuals of the Iranian sheep breed. The results indicated that 20 of the 114 CNVRs (18%) identified in Iranian sheep breed were also identified in our study, most of those overlapped with the genes influencing production, reproduction and immune system. Overall, this is the first attempts to develop the genomic map of loss and gain variation in the genome of Afghan indigenous sheep breeds, and may be important to shed some light on the genomic regions associated with some economically important traits in these breeds.
Collapse
Affiliation(s)
- Mohammad Hossein Moradi
- Department of Animal Science, Faculty of Agriculture and Natural Resources, Arak University, Arak, 38156-8-8349, Iran.
| | - Roqiah Mahmodi
- Department of Animal Science, Faculty of Agriculture and Natural Resources, Arak University, Arak, 38156-8-8349, Iran
| | | | - Mohammad Osman Karimi
- Department of Animal Science, Faculty of Agriculture and Natural Resources, Herat University, Herat, Afghanistan
| |
Collapse
|
34
|
Wang Y, Tsuo K, Kanai M, Neale BM, Martin AR. Challenges and Opportunities for Developing More Generalizable Polygenic Risk Scores. Annu Rev Biomed Data Sci 2022; 5:293-320. [PMID: 35576555 PMCID: PMC9828290 DOI: 10.1146/annurev-biodatasci-111721-074830] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Polygenic risk scores (PRS) estimate an individual's genetic likelihood of complex traits and diseases by aggregating information across multiple genetic variants identified from genome-wide association studies. PRS can predict a broad spectrum of diseases and have therefore been widely used in research settings. Some work has investigated their potential applications as biomarkers in preventative medicine, but significant work is still needed to definitively establish and communicate absolute risk to patients for genetic and modifiable risk factors across demographic groups. However, the biggest limitation of PRS currently is that they show poor generalizability across diverse ancestries and cohorts. Major efforts are underway through methodological development and data generation initiatives to improve their generalizability. This review aims to comprehensively discuss current progress on the development of PRS, the factors that affect their generalizability, and promising areas for improving their accuracy, portability, and implementation.
Collapse
Affiliation(s)
- Ying Wang
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, Massachusetts, USA;
- Stanley Center for Psychiatric Research and Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | - Kristin Tsuo
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, Massachusetts, USA;
- Stanley Center for Psychiatric Research and Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
- Biological and Biomedical Sciences, Harvard Medical School, Boston, Massachusetts, USA
| | - Masahiro Kanai
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, Massachusetts, USA;
- Stanley Center for Psychiatric Research and Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts, USA
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Benjamin M Neale
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, Massachusetts, USA;
- Stanley Center for Psychiatric Research and Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | - Alicia R Martin
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, Massachusetts, USA;
- Stanley Center for Psychiatric Research and Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| |
Collapse
|
35
|
Fabbri C. Genetics in psychiatry: Methods, clinical applications and future perspectives. PCN REPORTS : PSYCHIATRY AND CLINICAL NEUROSCIENCES 2022; 1:e6. [PMID: 38868637 PMCID: PMC11114394 DOI: 10.1002/pcn5.6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/18/2022] [Accepted: 03/02/2022] [Indexed: 06/14/2024]
Abstract
Psychiatric disorders and related traits have a demonstrated genetic component, with heritability estimated by twin studies generally between 80% and 40%. Their pathogenesis is complex and multi-determined: environmental factors interact with a polygenic architecture, making difficult the development of models able to stratify patients or predict mental health outcomes. Despite this difficult challenge, relevant progress has been made in the field of psychiatric genetics in recent years. This review aims to present the main current methods in psychiatric genetics, their output, limitations, clinical applications, and possible future developments. Genome-wide association studies (GWASs) performed in increasingly large samples have led to the identification of replicated genetic loci associated with the risk of major psychiatric disorders, including schizophrenia and mood disorders. Statistical and biological approaches have been developed to improve our understanding of the etiopathogenetic mechanisms behind genome-wide significant associations, as well as for estimating the cumulative effect of risk variants at the individual level and the genetic overlap between different disorders, as pleiotropy is the rule rather than the exception. Clinical applications are available in the pharmacogenetics field. The main issues that remain to be addressed include improving ethnic diversity in genetic studies and the optimization of statistical power through methodological improvements, such as the definition of dimensional phenotypes with specific biological correlates and the integration of different types of omics data.
Collapse
Affiliation(s)
- Chiara Fabbri
- Department of Biomedical and Neuromotor SciencesUniversity of BolognaBolognaItaly
- Institute of Psychiatry, Psychology & NeuroscienceKing's College LondonLondonUK
| |
Collapse
|
36
|
Cowling SB, Treeintong P, Ferguson J, Soltani H, Swarup R, Mayes S, Murchie EH. Out of Africa: characterizing the natural variation in dynamic photosynthetic traits in a diverse population of African rice (Oryza glaberrima). JOURNAL OF EXPERIMENTAL BOTANY 2022. [PMID: 34657157 DOI: 10.5281/zenodo.5555931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
African rice (Oryza glaberrima) has adapted to challenging environments and is a promising source of genetic variation. We analysed dynamics of photosynthesis and morphology in a reference set of 155 O. glaberrima accessions. Plants were grown in an agronomy glasshouse to late tillering stage. Photosynthesis induction from darkness and the decrease in low light was measured by gas exchange and chlorophyll fluorescence along with root and shoot biomass, stomatal density, and leaf area. Steady-state and kinetic responses were modelled. We describe extensive natural variation in O. glaberrima for steady-state, induction, and reduction responses of photosynthesis that has value for gene discovery and crop improvement. Principal component analyses indicated key clusters of plant biomass, kinetics of photosynthesis (CO2 assimilation, A), and photoprotection induction and reduction (measured by non-photochemical quenching, NPQ), consistent with diverse adaptation. Accessions also clustered according to countries with differing water availability, stomatal conductance (gs), A, and NPQ, indicating that dynamic photosynthesis has adaptive value in O. glaberrima. Kinetics of NPQ, A, and gs showed high correlation with biomass and leaf area. We conclude that dynamic photosynthetic traits and NPQ are important within O. glaberrima, and we highlight NPQ kinetics and NPQ under low light.
Collapse
Affiliation(s)
- Sophie B Cowling
- Division of Plant and Crop Science, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| | - Pracha Treeintong
- Division of Plant and Crop Science, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| | - John Ferguson
- Division of Plant and Crop Science, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, UK
- Department of Plant Sciences, University of Cambridge, Cambridge, UK
| | - Hamidreza Soltani
- Advanced Data Analysis Centre, University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| | - Ranjan Swarup
- Division of Plant and Crop Science, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| | - Sean Mayes
- Division of Plant and Crop Science, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| | - Erik H Murchie
- Division of Plant and Crop Science, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| |
Collapse
|
37
|
Cowling SB, Treeintong P, Ferguson J, Soltani H, Swarup R, Mayes S, Murchie EH. Out of Africa: characterizing the natural variation in dynamic photosynthetic traits in a diverse population of African rice (Oryza glaberrima). JOURNAL OF EXPERIMENTAL BOTANY 2022; 73:3283-3298. [PMID: 34657157 PMCID: PMC9126740 DOI: 10.1093/jxb/erab459] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 10/15/2021] [Indexed: 05/15/2023]
Abstract
African rice (Oryza glaberrima) has adapted to challenging environments and is a promising source of genetic variation. We analysed dynamics of photosynthesis and morphology in a reference set of 155 O. glaberrima accessions. Plants were grown in an agronomy glasshouse to late tillering stage. Photosynthesis induction from darkness and the decrease in low light was measured by gas exchange and chlorophyll fluorescence along with root and shoot biomass, stomatal density, and leaf area. Steady-state and kinetic responses were modelled. We describe extensive natural variation in O. glaberrima for steady-state, induction, and reduction responses of photosynthesis that has value for gene discovery and crop improvement. Principal component analyses indicated key clusters of plant biomass, kinetics of photosynthesis (CO2 assimilation, A), and photoprotection induction and reduction (measured by non-photochemical quenching, NPQ), consistent with diverse adaptation. Accessions also clustered according to countries with differing water availability, stomatal conductance (gs), A, and NPQ, indicating that dynamic photosynthesis has adaptive value in O. glaberrima. Kinetics of NPQ, A, and gs showed high correlation with biomass and leaf area. We conclude that dynamic photosynthetic traits and NPQ are important within O. glaberrima, and we highlight NPQ kinetics and NPQ under low light.
Collapse
Affiliation(s)
- Sophie B Cowling
- Division of Plant and Crop Science, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| | - Pracha Treeintong
- Division of Plant and Crop Science, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| | - John Ferguson
- Division of Plant and Crop Science, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, UK
- Department of Plant Sciences, University of Cambridge, Cambridge, UK
| | - Hamidreza Soltani
- Advanced Data Analysis Centre, University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| | - Ranjan Swarup
- Division of Plant and Crop Science, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| | - Sean Mayes
- Division of Plant and Crop Science, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| | - Erik H Murchie
- Division of Plant and Crop Science, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, UK
- Correspondence:
| |
Collapse
|
38
|
Song S, Jiang W, Zhang Y, Hou L, Zhao H. Leveraging LD eigenvalue regression to improve the estimation of SNP heritability and confounding inflation. Am J Hum Genet 2022; 109:802-811. [PMID: 35421325 PMCID: PMC9118121 DOI: 10.1016/j.ajhg.2022.03.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/18/2022] [Indexed: 12/17/2022] Open
Abstract
Heritability is a fundamental concept in genetic studies, measuring the genetic contribution to complex traits and bringing insights about disease mechanisms. The advance of high-throughput technologies has provided many resources for heritability estimation. Linkage disequilibrium (LD) score regression (LDSC) estimates both heritability and confounding biases, such as cryptic relatedness and population stratification, among single-nucleotide polymorphisms (SNPs) by using only summary statistics released from genome-wide association studies. However, only partial information in the LD matrix is utilized in LDSC, leading to loss in precision. In this study, we propose LD eigenvalue regression (LDER), an extension of LDSC, by making full use of the LD information. Compared to state-of-the-art heritability estimating methods, LDER provides more accurate estimates of SNP heritability and better distinguishes the inflation caused by polygenicity and confounding effects. We demonstrate the advantages of LDER both theoretically and with extensive simulations. We applied LDER to 814 complex traits from UK Biobank, and LDER identified 363 significantly heritable phenotypes, among which 97 were not identified by LDSC.
Collapse
Affiliation(s)
- Shuang Song
- Center for Statistical Science, Department of Industrial Engineering, Tsinghua University, Beijing 100084, China
| | - Wei Jiang
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06510, USA
| | - Yiliang Zhang
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06510, USA
| | - Lin Hou
- Center for Statistical Science, Department of Industrial Engineering, Tsinghua University, Beijing 100084, China; MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Hongyu Zhao
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06510, USA.
| |
Collapse
|
39
|
Speed D, Kaphle A, Balding DJ. SNP-based heritability and selection analyses: Improved models and new results. Bioessays 2022; 44:e2100170. [PMID: 35279859 DOI: 10.1002/bies.202100170] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 03/02/2022] [Accepted: 03/03/2022] [Indexed: 01/15/2023]
Abstract
Complex-trait genetics has advanced dramatically through methods to estimate the heritability tagged by SNPs, both genome-wide and in genomic regions of interest such as those defined by functional annotations. The models underlying many of these analyses are inadequate, and consequently many SNP-heritability results published to date are inaccurate. Here, we review the modelling issues, both for analyses based on individual genotype data and association test statistics, highlighting the role of a low-dimensional model for the heritability of each SNP. We use state-of-art models to present updated results about how heritability is distributed with respect to functional annotations in the human genome, and how it varies with allele frequency, which can reflect purifying selection. Our results give finer detail to the picture that has emerged in recent years of complex trait heritability widely dispersed across the genome. Confounding due to population structure remains a problem that summary statistic analyses cannot reliably overcome. Also see the video abstract here: https://youtu.be/WC2u03V65MQ.
Collapse
Affiliation(s)
- Doug Speed
- Quantitative Genetics and Genomics, Aarhus University, Aarhus, Denmark.,Aarhus Institute of Advanced Studies, Aarhus University, Aarhus, Denmark.,UCL Genetics Institute, University College London, London, UK
| | - Anubhav Kaphle
- Melbourne Integrative Genomics, School of BioSciences and School of Mathematics and Statistics, University of Melbourne, Victoria, Australia
| | - David J Balding
- UCL Genetics Institute, University College London, London, UK.,Melbourne Integrative Genomics, School of BioSciences and School of Mathematics and Statistics, University of Melbourne, Victoria, Australia
| |
Collapse
|
40
|
Yuan Z, Liu L, Guo P, Yan R, Xue F, Zhou X. Likelihood-based Mendelian randomization analysis with automated instrument selection and horizontal pleiotropic modeling. SCIENCE ADVANCES 2022; 8:eabl5744. [PMID: 35235357 PMCID: PMC8890724 DOI: 10.1126/sciadv.abl5744] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 01/05/2022] [Indexed: 05/03/2023]
Abstract
Mendelian randomization (MR) is a common tool for identifying causal risk factors underlying diseases. Here, we present a method, MR with automated instrument determination (MRAID), for effective MR analysis. MRAID borrows ideas from fine-mapping analysis to model an initial set of candidate single-nucleotide polymorphisms that are in potentially high linkage disequilibrium with each other and automatically selects among them the suitable instruments for causal inference. MRAID also explicitly models both uncorrelated and correlated horizontal pleiotropic effects that are widespread for complex trait analysis. MRAID achieves both tasks through a joint likelihood framework and relies on a scalable sampling-based algorithm to compute calibrated P values. Comprehensive and realistic simulations show that MRAID can provide calibrated type I error control and reduce false positives while being more powerful than existing approaches. We illustrate the benefits of MRAID for an MR screening analysis across 645 trait pairs in U.K. Biobank, identifying multiple lifestyle causal risk factors of cardiovascular disease-related traits.
Collapse
Affiliation(s)
- Zhongshang Yuan
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Lu Liu
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Ping Guo
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Ran Yan
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Fuzhong Xue
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Xiang Zhou
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA
- Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
41
|
Cavedon M, vonHoldt B, Hebblewhite M, Hegel T, Heppenheimer E, Hervieux D, Mariani S, Schwantje H, Steenweg R, Watters M, Musiani M. Selection of both habitat and genes in specialized and endangered caribou. CONSERVATION BIOLOGY : THE JOURNAL OF THE SOCIETY FOR CONSERVATION BIOLOGY 2022; 36. [PMID: 35146809 DOI: 10.1111/cobi.13900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 01/26/2022] [Accepted: 01/31/2022] [Indexed: 06/14/2023]
Abstract
Genetic mechanisms determining habitat selection and specialization of individuals within species have been hypothesized, but not tested at the appropriate individual level in nature. In this work, we analyzed habitat selection for 139 GPS-collared caribou belonging to three declining ecotypes sampled throughout Northwestern Canada. We used Resource Selection Functions (RSFs) comparing resources at used and available locations. We found that the three caribou ecotypes differed in their use of habitat suggesting specialization. On expected grounds, we also found differences in habitat selection between summer and winter, but also, originally, among the individuals within an ecotype. We next obtained Single Nucleotide Polymorphisms (SNPs) for the same caribou individuals, we detected those associated to habitat selection, and then identified genes linked to these SNPs. These genes had functions related in other organisms to habitat and dietary specializations, and climatic adaptations. We therefore suggest that individual variation in habitat selection was based on genotypic variation in the SNPs of individual caribou, indicating that genetic forces underlie habitat and diet selection in the species. We also suggest that the associations between habitat and genes that we detected may lead to lack of resilience in the species, thus contributing to caribou endangerment. Our work emphasizes that similar mechanisms may exist for other specialized, endangered species. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Maria Cavedon
- Faculty of Environmental Design, University of Calgary, Calgary, AB, T2N 1N4, Canada
| | - Bridgett vonHoldt
- Department of Ecology & Evolutionary Biology, Princeton University, 106A Guyot Hall, Princeton, NJ, 08544-2016, USA
| | - Mark Hebblewhite
- Wildlife Biology Program, Department of Ecosystem and Conservation Sciences, College of Forestry and Conservation, University of Montana, Montana, MT, 59812, USA
| | - Troy Hegel
- Yukon Department of Environment, Whitehorse, Yukon, Y1A 2C6, Canada
- Fish and Wildlife Stewardship Branch, Alberta Environment and Parks, 4999 98 Ave., Edmonton, AB, T6B 2×3, Canada
| | - Elizabeth Heppenheimer
- Department of Ecology & Evolutionary Biology, Princeton University, 106A Guyot Hall, Princeton, NJ, 08544-2016, USA
| | - Dave Hervieux
- Fish and Wildlife Stewardship Branch, Alberta Environment and Parks, Grande Prairie, AB, T8V 6J4, Canada
| | - Stefano Mariani
- School of Natural Sciences and Psychology, Liverpool John Moores University, Liverpool, UK
| | - Helen Schwantje
- Wildlife and Habitat Branch, Ministry of Forests, Lands, Natural Resource Operations and Rural Development, Government of British Columbia, 2080 Labieux Road, Nanaimo, BC, V9T 6J 9, Canada
| | - Robin Steenweg
- Pacific Region, Canadian Wildlife Service, Environment and Climate Change Canada, 5421 Robertson Road, Delta, BC, V4K 3N2, Canada
| | - Megan Watters
- Land and Resource Specialist, 300 - 10003 110th Avenue Fort, St. John, BC, V1J 6M7, Canada
| | - Marco Musiani
- Dept. of Biological Sciences, Faculty of Science and Veterinary Medicine (Joint Appointment), University of Calgary, Calgary, AB, T2N 1N4, Canada
| |
Collapse
|
42
|
Livne I, Azriel D, Goldberg Y. Improved estimators for semi-supervised high-dimensional regression model. Electron J Stat 2022. [DOI: 10.1214/22-ejs2070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- Ilan Livne
- The Faculty of Industrial Engineering and Management, Technion, Israel
| | - David Azriel
- The Faculty of Industrial Engineering and Management, Technion, Israel
| | - Yair Goldberg
- The Faculty of Industrial Engineering and Management, Technion, Israel
| |
Collapse
|
43
|
Peng H, Wu X, Wen Y, Ao Y, Li Y, Guan W, Lin J, Li C, Liang H, He J, Liang W. Univariable and Multivariable Two-Sample Mendelian Randomization Investigating the Effects of Leisure Sedentary Behaviors on the Risk of Lung Cancer. Front Genet 2021; 12:742718. [PMID: 34899835 PMCID: PMC8651878 DOI: 10.3389/fgene.2021.742718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 10/22/2021] [Indexed: 11/13/2022] Open
Abstract
Leisure sedentary behaviors (LSB) are widespread, and observational studies have provided emerging evidence that LSB play a role in the development of lung cancer (LC). However, the causal inference between LSB and LC remains unknown. Methods: We utilized univariable (UVMR) and multivariable two-sample Mendelian randomization (MVMR) analysis to disentangle the effects of LSB on the risk of LC. MR analysis was conducted with genetic variants from genome-wide association studies of LSB (408,815 persons from UK Biobank), containing 152 single-nucleotide polymorphisms (SNPs) for television (TV) watching, 37 SNPs for computer use, and four SNPs for driving, and LC from the International Lung Cancer Consortium (11,348 cases and 15,861 controls). Multiple sensitivity analyses were further performed to verify the causality. Results: UVMR demonstrated that genetically predisposed 1.5-h increase in LSB spent on watching TV increased the odds of LC by 90% [odds ratio (OR), 1.90; 95% confidence interval (CI), 1.44-2.50; p < 0.001]. Similar trends were observed for squamous cell lung cancer (OR, 1.97; 95%CI, 1.31-2.94; p = 0.0010) and lung adenocarcinoma (OR, 1.64; 95%CI 1.12-2.39; p = 0.0110). The causal effects remained significant after adjusting for education (OR, 1.97; 95%CI, 1.44-2.68; p < 0.001) and body mass index (OR, 1.86; 95%CI, 1.36-2.54; p < 0.001) through MVMR approach. No association was found between prolonged LSB spent on computer use and driving and LC risk. Genetically predisposed prolonged LSB was additionally correlated with smoking (OR, 1.557; 95%CI, 1.287-1.884; p < 0.001) and alcohol consumption (OR, 1.010; 95%CI, 1.004-1.016; p = 0.0016). Consistency of results across complementary sensitivity MR methods further strengthened the causality. Conclusion: Robust evidence was demonstrated for an independent, causal effect of LSB spent on watching TV in increasing the risk of LC. Further work is necessary to investigate the potential mechanisms.
Collapse
Affiliation(s)
- Haoxin Peng
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Nanshan School, Guangzhou Medical University, Guangzhou, China
| | - Xiangrong Wu
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Nanshan School, Guangzhou Medical University, Guangzhou, China
| | - Yaokai Wen
- School of Medicine, Tongji University, Shanghai, China.,Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Yiyuan Ao
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Nanshan School, Guangzhou Medical University, Guangzhou, China
| | - Yutian Li
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenhui Guan
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jinsheng Lin
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Caichen Li
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hengrui Liang
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jianxing He
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenhua Liang
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Medical Oncology, the First People's Hospital of Zhaoqing, Zhaoqing, China
| |
Collapse
|
44
|
“Fast” women? The effects of childhood environments on women's developmental timing, mating strategies, and reproductive outcomes. EVOL HUM BEHAV 2021. [DOI: 10.1016/j.evolhumbehav.2021.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
45
|
Where the genome meets the connectome: Understanding how genes shape human brain connectivity. Neuroimage 2021; 244:118570. [PMID: 34508898 DOI: 10.1016/j.neuroimage.2021.118570] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 08/10/2021] [Accepted: 09/07/2021] [Indexed: 02/07/2023] Open
Abstract
The integration of modern neuroimaging methods with genetically informative designs and data can shed light on the molecular mechanisms underlying the structural and functional organization of the human connectome. Here, we review studies that have investigated the genetic basis of human brain network structure and function through three complementary frameworks: (1) the quantification of phenotypic heritability through classical twin designs; (2) the identification of specific DNA variants linked to phenotypic variation through association and related studies; and (3) the analysis of correlations between spatial variations in imaging phenotypes and gene expression profiles through the integration of neuroimaging and transcriptional atlas data. We consider the basic foundations, strengths, limitations, and discoveries associated with each approach. We present converging evidence to indicate that anatomical connectivity is under stronger genetic influence than functional connectivity and that genetic influences are not uniformly distributed throughout the brain, with phenotypic variation in certain regions and connections being under stronger genetic control than others. We also consider how the combination of imaging and genetics can be used to understand the ways in which genes may drive brain dysfunction in different clinical disorders.
Collapse
|
46
|
Liu TY, Lin CF, Wu HT, Wu YL, Chen YC, Liao CC, Chou YP, Chao D, Chang YS, Lu HF, Chang JG, Hsu KC, Tsai FJ. Comparison of multiple imputation algorithms and verification using whole-genome sequencing in the CMUH genetic biobank. Biomedicine (Taipei) 2021; 11:57-65. [PMID: 35223420 PMCID: PMC8823485 DOI: 10.37796/2211-8039.1302] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 03/09/2021] [Accepted: 04/08/2021] [Indexed: 11/13/2022] Open
Abstract
A genome-wide association study (GWAS) can be conducted to systematically analyze the contributions of genetic factors to a wide variety of complex diseases. Nevertheless, existing GWASs have provided highly ethnic specific data. Accordingly, to provide data specific to Taiwan, we established a large-scale genetic database in a single medical institution at the China Medical University Hospital. With current technological limitations, microarray analysis can detect only a limited number of single-nucleotide polymorphisms (SNPs) with a minor allele frequency of >1%. Nevertheless, imputation represents a useful alternative means of expanding data. In this study, we compared four imputation algorithms in terms of various metrics. We observed that among the compared algorithms, Beagle5.2 achieved the fastest calculation speed, smallest storage space, highest specificity, and highest number of high-quality variants. We obtained 15,277,414 high-quality variants in 175,871 people by using Beagle5.2. In our internal verification process, Beagle5.2 exhibited an accuracy rate of up to 98.75%. We also conducted external verification. Our imputed variants had a 79.91% mapping rate and 90.41% accuracy. These results will be combined with clinical data in future research. We have made the results available for researchers to use in formulating imputation algorithms, in addition to establishing a complete SNP database for GWAS and PRS researchers. We believe that these data can help improve overall medical capabilities, particularly precision medicine, in Taiwan.
Collapse
Affiliation(s)
- Ting-Yuan Liu
- Center for Precision Medicine, China Medical University Hospital, Taichung, 40447,
Taiwan
| | - Chih-Fan Lin
- Artificial Intelligence Center for Medical Diagnosis, China Medical University Hospital, Taichung, 40447,
Taiwan
| | - Hsing-Tsung Wu
- Artificial Intelligence Center for Medical Diagnosis, China Medical University Hospital, Taichung, 40447,
Taiwan
| | - Ya-Lun Wu
- Artificial Intelligence Center for Medical Diagnosis, China Medical University Hospital, Taichung, 40447,
Taiwan
| | - Yu-Chia Chen
- Center for Precision Medicine, China Medical University Hospital, Taichung, 40447,
Taiwan
| | - Chi-Chou Liao
- Center for Precision Medicine, China Medical University Hospital, Taichung, 40447,
Taiwan
| | - Yu-Pao Chou
- Center for Precision Medicine, China Medical University Hospital, Taichung, 40447,
Taiwan
| | - Dysan Chao
- Center for Precision Medicine, China Medical University Hospital, Taichung, 40447,
Taiwan
| | - Ya-Sian Chang
- Center for Precision Medicine, China Medical University Hospital, Taichung, 40447,
Taiwan
- Epigenome Research Center, China Medical University Hospital, Taichung, 40447,
Taiwan
| | - Hsing-Fang Lu
- Million-person Precision Medicine Initiative, China Medical University Hospital, Taichung, 40447,
Taiwan
| | - Jan-Gowth Chang
- Center for Precision Medicine, China Medical University Hospital, Taichung, 40447,
Taiwan
- Epigenome Research Center, China Medical University Hospital, Taichung, 40447,
Taiwan
| | - Kai-Cheng Hsu
- Artificial Intelligence Center for Medical Diagnosis, China Medical University Hospital, Taichung, 40447,
Taiwan
- Department of Medicine, China Medical University, Taichung,
Taiwan
- Department of Neurology, China Medical University Hospital, Taichung,
Taiwan
| | - Fuu-Jen Tsai
- Department of Medical Research, China Medical University Hospital, Taichung, 40402,
Taiwan
- School of Chinese Medicine, China Medical University, Taichung, 40402,
Taiwan
- Division of Pediatric Genetics, Children’s Hospital of China Medical University, Taichung, 40447,
Taiwan
- Department of Biotechnology and Bioinformatics, Asia University, Taichung, 41354,
Taiwan
| |
Collapse
|
47
|
Ma Y, Zhou X. Genetic prediction of complex traits with polygenic scores: a statistical review. Trends Genet 2021; 37:995-1011. [PMID: 34243982 PMCID: PMC8511058 DOI: 10.1016/j.tig.2021.06.004] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/31/2021] [Accepted: 06/03/2021] [Indexed: 01/03/2023]
Abstract
Accurate genetic prediction of complex traits can facilitate disease screening, improve early intervention, and aid in the development of personalized medicine. Genetic prediction of complex traits requires the development of statistical methods that can properly model polygenic architecture and construct a polygenic score (PGS). We present a comprehensive review of 46 methods for PGS construction. We connect the majority of these methods through a multiple linear regression framework which can be instrumental for understanding their prediction performance for traits with distinct genetic architectures. We discuss the practical considerations of PGS analysis as well as challenges and future directions of PGS method development. We hope our review serves as a useful reference both for statistical geneticists who develop PGS methods and for data analysts who perform PGS analysis.
Collapse
Affiliation(s)
- Ying Ma
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Xiang Zhou
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA; Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
48
|
Wang L, Gao B, Fan Y, Xue F, Zhou X. Mendelian randomization under the omnigenic architecture. Brief Bioinform 2021; 22:6347949. [PMID: 34379090 DOI: 10.1093/bib/bbab322] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/22/2021] [Accepted: 07/24/2021] [Indexed: 11/15/2022] Open
Abstract
Mendelian randomization (MR) is a common analytic tool for exploring the causal relationship among complex traits. Existing MR methods require selecting a small set of single nucleotide polymorphisms (SNPs) to serve as instrument variables. However, selecting a small set of SNPs may not be ideal, as most complex traits have a polygenic or omnigenic architecture and are each influenced by thousands of SNPs. Here, motivated by the recent omnigenic hypothesis, we present an MR method that uses all genome-wide SNPs for causal inference. Our method uses summary statistics from genome-wide association studies as input, accommodates the commonly encountered horizontal pleiotropy effects and relies on a composite likelihood framework for scalable computation. We refer to our method as the omnigenic Mendelian randomization, or OMR. We examine the power and robustness of OMR through extensive simulations including those under various modeling misspecifications. We apply OMR to several real data applications, where we identify multiple complex traits that potentially causally influence coronary artery disease (CAD) and asthma. The identified new associations reveal important roles of blood lipids, blood pressure and immunity underlying CAD as well as important roles of immunity and obesity underlying asthma.
Collapse
Affiliation(s)
- Lu Wang
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA.,Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Boran Gao
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA.,Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yue Fan
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA.,Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA.,School of Public Health, Health Science Center of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Fuzhong Xue
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiang Zhou
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA.,Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
49
|
McGowan MT, Zhang Z, Ficklin SP. Chromosomal characteristics of salt stress heritable gene expression in the rice genome. BMC Genom Data 2021; 22:17. [PMID: 34044788 PMCID: PMC8162008 DOI: 10.1186/s12863-021-00970-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 05/06/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Gene expression is potentially an important heritable quantitative trait that mediates between genetic variation and higher-level complex phenotypes through time and condition-dependent regulatory interactions. Therefore, we sought to explore both the genomic and condition-specific characteristics of gene expression heritability within the context of chromosomal structure. RESULTS Heritability was estimated for biological gene expression using a diverse, 84-line, Oryza sativa (rice) population under optimal and salt-stressed conditions. Overall, 5936 genes were found to have heritable expression regardless of condition and 1377 genes were found to have heritable expression only during salt stress. These genes with salt-specific heritable expression are enriched for functional terms associated with response to stimulus and transcription factor activity. Additionally, we discovered that highly and lowly expressed genes, and genes with heritable expression are distributed differently along the chromosomes in patterns that follow previously identified high-throughput chromosomal conformation capture (Hi-C) A/B chromatin compartments. Furthermore, multiple genomic hot-spots enriched for genes with salt-specific heritability were identified on chromosomes 1, 4, 6, and 8. These hotspots were found to contain genes functionally enriched for transcriptional regulation and overlaps with a previously identified major QTL for salt-tolerance in rice. CONCLUSIONS Investigating the heritability of traits, and in-particular gene expression traits, is important towards developing a basic understanding of how regulatory networks behave across a population. This work provides insights into spatial patterns of heritable gene expression at the chromosomal level.
Collapse
Affiliation(s)
- Matthew T McGowan
- Molecular Plant Sciences Program, Washington State University, French Ad 324G, Pullman, WA, 99164, USA.
| | - Zhiwu Zhang
- Molecular Plant Sciences Program, Washington State University, French Ad 324G, Pullman, WA, 99164, USA.,Department of Crops and Soils, Washington State University, 105 Johnson Hall, Pullman, WA, 99164, USA
| | - Stephen P Ficklin
- Molecular Plant Sciences Program, Washington State University, French Ad 324G, Pullman, WA, 99164, USA.,Department of Horticulture, Washington State University, 149 Johnson Hall, Pullman, WA, 99164, USA
| |
Collapse
|