Improved Imputation Accuracy of Rare and Low-Frequency Variants Using Population-Specific High-Coverage WGS-Based Imputation Reference Panel

Total Page:16

File Type:pdf, Size:1020Kb

Improved Imputation Accuracy of Rare and Low-Frequency Variants Using Population-Specific High-Coverage WGS-Based Imputation Reference Panel Improved imputation accuracy of rare and low- frequency variants using population-specific high- coverage WGS-based imputation reference panel The MIT Faculty has made this article openly available. Please share how this access benefits you. Your story matters. Citation Mitt, Mario et al. “Improved Imputation Accuracy of Rare and Low- Frequency Variants Using Population-Specific High-Coverage WGS- Based Imputation Reference Panel.” European Journal of Human Genetics 25, 7 (April 2017): 869–876 © 2017 The Author(s) As Published http://dx.doi.org/10.1038/EJHG.2017.51 Publisher Nature Publishing Group Version Final published version Citable link http://hdl.handle.net/1721.1/116697 Terms of Use Creative Commons Attribution-NonCommercial 4.0 International Detailed Terms http://creativecommons.org/licenses/by-nc/4.0/ European Journal of Human Genetics (2017) 25, 869–876 Official journal of The European Society of Human Genetics www.nature.com/ejhg ARTICLE Improved imputation accuracy of rare and low-frequency variants using population-specific high-coverage WGS-based imputation reference panel Mario Mitt1,2,7, Mart Kals1,3,7, Kalle Pärn1,4,7, Stacey B Gabriel5, Eric S Lander5, Aarno Palotie4,5, Samuli Ripatti4, Andrew P Morris1,6, Andres Metspalu1,2, Tõnu Esko1,5, Reedik Mägi1,8 and Priit Palta*,1,4,8 Genetic imputation is a cost-efficient way to improve the power and resolution of genome-wide association (GWA) studies. Current publicly accessible imputation reference panels accurately predict genotypes for common variants with minor allele frequency (MAF) ≥ 5% and low-frequency variants (0.5 ≤ MAFo5%) across diverse populations, but the imputation of rare variation (MAFo0.5%) is still rather limited. In the current study, we evaluate imputation accuracy achieved with reference panels from diverse populations with a population-specific high-coverage (30 ×) whole-genome sequencing (WGS) based reference panel, comprising of 2244 Estonian individuals (0.25% of adult Estonians). Although the Estonian-specific panel contains fewer haplotypes and variants, the imputation confidence and accuracy of imputed low-frequency and rare variants was significantly higher. The results indicate the utility of population-specific reference panels for human genetic studies. European Journal of Human Genetics (2017) 25, 869–876; doi:10.1038/ejhg.2017.51; published online 12 April 2017 INTRODUCTION (HRC)20 contains even more individuals (N = 32 488, mostly with Genotype imputation is a method for statistically inferring untyped European ancestry) and should therefore enable better imputation of genotypes in a sample of partially genotyped individuals, based on a both low-frequency and rare variants in European samples. reference panel of individuals who have been more densely genotyped Recently, several studies have demonstrated that the use of or sequenced. Imputation methods attempt to identify haplotype population-specific IRPs can further improve the imputation accuracy sharing between individuals in the sample and in an imputation of common and low-frequency variants, and improve the imputation reference panel (IRP), and use this information to infer the alleles at of rarer variants in the relevant population.21–24 By using an IRP untyped loci in the sample.1 Imputation allows geneticists to study composed of related Dutch individuals, Deelan et al.23 showed that it variants that have not been directly genotyped in a sample and thereby is possible to substantially improve the completeness and accuracy of to increase power and resolution of genome-wide association studies imputation of rare variants into a set of Dutch individuals. Gudb- (GWAS). Imputation is particularly useful for combining association jartsson et al used long-range haplotype phasing in combination with results across studies that used different genotyping arrays2 and imputation to increase imputation accuracy for rare variants down to facilitate fine-mapping to localise association signals by considering MAF of 0.1% in the Icelandic population.22 Sidore et al.reported all genetic variants in a region. several variants associated with circulating lipid levels in Sardinians Publicly available IRPs from the International HapMap Project3,4 that were detected due to accurate imputation achieved by using a and 1000 Genomes Project (1000G)5 have been instrumental to the Sardinian WGS-based IRP; these authors showed that the variants discovery of thousands of loci affecting diseases and traits in individual would not have been identified if the analyses had been based on the GWAS and collaborative meta-analyses. The first wave of studies 1000G IRP.24 Similar results were obtained in the UK10K project, mostly used the HapMap II IRP, which used microarray-based where the British population-specific IRP combined with 1000G genotypes from 270 individuals at 3.1 million (M) variants.6–10 Later Project reference panel facilitated the discovery of several novel genetic studies used IRPs based on the 1000G project, which performed variants associated with medically relevant phenotypes.19,25,26 whole-genome sequencing (WGS) on a diverse set of populations, Studies have shown that the genetic structure of European countries with 2504 individuals and up to 84.4 M variants.11–16 Although the correlates closely with their geographic origin.27,28 The Estonian latter IRP allows robust imputation of common variants (minor allele population, being located in Northeast Europe, is genetically most frequency (MAF) ≥ 5%) and low-frequency variants (0.5 ≤ MAFo5%) similar to its neighbouring countries, including Finland, the North- 5 it has only limited imputation accuracy for rare (MAFo0.5%) western part of Russia, and other Baltic countries.28–30 Notwithstand- variants.17–19 A recent IRP from Haplotype Reference Consortium ing this overall genetic similarity, the Estonian population still has a 1Estonian Genome Center, University of Tartu, Tartu, Estonia; 2Department of Biotechnology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia; 3Institute of Mathematics and Statistics, University of Tartu, Tartu, Estonia; 4Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland; 5Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA; 6Department of Biostatistics, University of Liverpool, Liverpool, UK *Correspondence: Dr P Palta, Estonian Genome Center, University of Tartu, 23B Riia Street., 51010 Tartu, Estonia. Tel: +372 7375029; Fax: +372 7374060; E-mail: [email protected] 7These authors contributed equally to this work. 8These authors jointly supervised this work. Received 17 December 2016; revised 14 February 2017; accepted 25 February 2017; published online 12 April 2017 Imputation with population-specific reference MMittet al 870 substantial proportion of haplotypes that are not expected to be Multi-allelic SNVs were removed and we further excluded variants with call covered by the more diverse IRPs. Moreover, the population-specific rate o0.95, minor allele count ≤ 2, Hardy–Weinberg equilibrium test P- − differences are expected to increase as allele frequencies decrease. valueo1×10 6 and variants in low-complexity regions.35 fi In the current study, we first evaluated two most commonly used Genotype array data was ltered sample-wise by excluding on the basis of call o 4 ± phasing algorithms to create population-specific IRP based on high- rate ( 98%), extreme heterozygosity ( mean 3 SD), genotype and pheno- type sex discordance, cryptic relatedness (IBD420%) and outliers from the coverage (30 ×) WGS data from 2244 Estonian individuals. To impute fi European descent from the MDS plot in comparison with HapMap reference low-frequency and rare variants more accurately in a speci cpopula- samples. SNP quality filtering included call rate (o99%), MAF (o1%) and tion, one can take two approaches: (i) increase the size of IRPs extreme deviation from Hardy–Weinberg equilibrium (P-valueo1×10− 4). from diverse populations to capture more reference haplotypes or Non-autosomal SNPs were excluded from the analysis. (ii) employ population-specific IRPs. We assessed the utility of these approaches for improving imputation in Estonian samples by compar- Haplotype phasing ing the performance of (i) an Estonian-specific IRP, (ii) the commonly The EGCUT WGS data was phased with SHAPEIT2(ref. 36) (r837), using four used 1000G IRP, (iii) the much larger HRC IRP and (iv) combinations computer cores. Pre-phasing of genotype array data was made in similar of these panels. manner using SHAPEIT2 using four cores. As a separate test for pre-phasing accuracy, we used chromosome 20 sequence of 2244 full genomes, which fi MATERIALS AND METHODS were ltered beforehand to exclude any non-founder family members and individuals with a genome-wide PI_HAT value above 0.5 (2195 individuals Cohort description remained) when compared to other individuals in the data set. To assess the 2304 geographically distributed individuals (selected randomly by county of efficiency of various approaches to phasing of WGS data, we applied two birth) from the Estonian Biobank of the Estonian Genome Center, University different tools: SHAPEIT2(ref. 36) and Eagle2.37,38 Both programs were engaged of Tartu (EGCUT) were selected for WGS. EGCUT is a population-based with the default parameters with varying number of cores (1, 2, 4, 8, 16, 24 and biobank, containing almost 52 000 samples of the adult population (aged ≥ 18 32). To verify the phasing accuracy for other data sets, the 1000G data was years), which
Recommended publications
  • Impact of Imputation Methods on the Amount of Genetic Variation Captured by a Single-Nucleotide Polymorphism Panel in Soybeans A
    View metadata, citation and similar papers at core.ac.uk brought to you by CORE provided by Purdue E-Pubs Purdue University Purdue e-Pubs Department of Agronomy Faculty Publications Department of Agronomy 2-2-2016 Impact of imputation methods on the amount of genetic variation captured by a single-nucleotide polymorphism panel in soybeans A. Xavier Purdue University William M. Muir Purdue University Katy M. Rainey Purdue Univeristy, [email protected] Follow this and additional works at: https://docs.lib.purdue.edu/agrypubs Recommended Citation Xavier, A.; Muir, William M.; and Rainey, Katy M., "Impact of imputation methods on the amount of genetic variation captured by a single-nucleotide polymorphism panel in soybeans" (2016). Department of Agronomy Faculty Publications. Paper 13. http://dx.doi.org/10.1186/s12859-016-0899-7 This document has been made available through Purdue e-Pubs, a service of the Purdue University Libraries. Please contact [email protected] for additional information. Xavier et al. BMC Bioinformatics (2016) 17:55 DOI 10.1186/s12859-016-0899-7 METHODOLOGY ARTICLE Open Access Impact of imputation methods on the amount of genetic variation captured by a single-nucleotide polymorphism panel in soybeans A. Xavier1, William M. Muir2 and Katy M. Rainey1* Abstract Background: Success in genome-wide association studies and marker-assisted selection depends on good phenotypic and genotypic data. The more complete this data is, the more powerful will be the results of analysis. Nevertheless, there are next-generation technologies that seek to provide genotypic information in spite of great proportions of missing data. The procedures these technologies use to impute genetic data, therefore, greatly affect downstream analyses.
    [Show full text]
  • Imputation-Based Assessment of Next Generation Rare Exome Variant Arrays
    IMPUTATION-BASED ASSESSMENT OF NEXT GENERATION RARE EXOME VARIANT ARRAYS ALICIA R. MARTIN* Department of Genetics & Biomedical Informatics Training Program, Stanford University Stanford, CA, 94305 Email: [email protected] GERARD TSE Department of Computer Science, Stanford University Stanford, CA, 94305 Email: [email protected] CARLOS D. BUSTAMANTE Department of Genetics, Stanford University Stanford, CA, 94305 Email: [email protected] EIMEAR E. KENNY* Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai New York, NY 10029 Email: [email protected] A striking finding from recent large-scale sequencing efforts is that the vast majority of variants in the human genome are rare and found within single populations or lineages. These observations hold important implications for the design of the next round of disease variant discovery efforts—if genetic variants that influence disease risk follow the same trend, then we expect to see population-specific disease associations that require large samples sizes for detection. To address this challenge, and due to the still prohibitive cost of sequencing large cohorts, researchers have developed a new generation of low-cost genotyping arrays that assay rare variation previously identified from large exome sequencing studies. Genotyping approaches rely not only on directly observing variants, but also on phasing and imputation methods that use publicly available reference panels to infer unobserved variants in a study cohort. Rare variant exome arrays are intentionally enriched for variants likely to be disease causing, and here we assay the ability of the first commercially available rare exome variant array (the Illumina Infinium HumanExome BeadChip) to also tag other potentially damaging variants not molecularly assayed.
    [Show full text]
  • Allele Imputation and Haplotype Determination from Databases Composed of Nuclear Families by Nathan Medina-Rodríguez and Ángelo Santana
    CONTRIBUTED RESEARCH ARTICLE 35 Allele Imputation and Haplotype Determination from Databases Composed of Nuclear Families by Nathan Medina-Rodríguez and Ángelo Santana Abstract The alleHap package is designed for imputing genetic missing data and reconstruct non- recombinant haplotypes from pedigree databases in a deterministic way. When genotypes of related individuals are available in a number of linked genetic markers, the program starts by identifying haplotypes compatible with the observed genotypes in those markers without missing values. If haplotypes are identified in parents or offspring, missing alleles can be imputed in subjects containing missing values. Several scenarios are analyzed: family completely genotyped, children partially genotyped and parents completely genotyped, children fully genotyped and parents containing entirely or partially missing genotypes, and founders and their offspring both only partially genotyped. The alleHap package also has a function to simulate pedigrees including all these scenarios. This article describes in detail how our package works for the desired applications, including illustrated explanations and easily reproducible examples. Introduction The knowledge about human genetic variation has been growing exponentially over the last decade. Collaborative efforts of international projects such as HapMap (Consortium and others, 2005) and 1000 Genomes (Consortium and others, 2012) have contributed to improving the discovery about human genetic diversity. Genotype imputation and haplotype reconstruction have achieved an important role in Genome- Wide Association Studies (GWAS) during recent years. Estimation methods are frequently used to infer missing genotypes as well as haplotypes from databases containing related and/or unrelated subjects. The majority of these analyses have been developed using several statistical methods (Browning and Browning, 2011a) which are able to impute genotypes as well as perform haplotype phasing (also known as haplotype estimation) of the corresponding genomic regions.
    [Show full text]
  • Identity-By-Descent Estimation with Population- and Pedigree-Based Imputation in Admixed Family Data Mohamad Saad
    Marshall University Marshall Digital Scholar Biochemistry and Microbiology Faculty Research 2016 Identity-by-descent estimation with population- and pedigree-based imputation in admixed family data Mohamad Saad Alejandro Q. Nato Jr. Marshall University, [email protected] Fiona L. Grimson Steven M. Lewis Lisa A. Brown See next page for additional authors Follow this and additional works at: https://mds.marshall.edu/sm_bm Part of the Genetic Phenomena Commons, Genetic Processes Commons, and the Genetic Structures Commons Recommended Citation Saad M, Nato AQ, Grimson FL, et al. Identity-by-descent estimation with population- and pedigree-based imputation in admixed family data. BMC Proceedings. 2016;10(Suppl 7):295-301. doi:10.1186/s12919-016-0046-5. This Conference Proceeding is brought to you for free and open access by the Faculty Research at Marshall Digital Scholar. It has been accepted for inclusion in Biochemistry and Microbiology by an authorized administrator of Marshall Digital Scholar. For more information, please contact [email protected], [email protected]. Authors Mohamad Saad, Alejandro Q. Nato Jr., Fiona L. Grimson, Steven M. Lewis, Lisa A. Brown, Elizabeth M. Blue, Timothy A. Thornton, Elizabeth A. Thompson, and Ellen M. Wijsman This conference proceeding is available at Marshall Digital Scholar: https://mds.marshall.edu/sm_bm/238 The Author(s) BMC Proceedings 2016, 10(Suppl 7):7 DOI 10.1186/s12919-016-0046-5 BMC Proceedings PROCEEDINGS Open Access Identity-by-descent estimation with population- and pedigree-based imputation in admixed family data Mohamad Saad1,2, Alejandro Q. Nato Jr2, Fiona L. Grimson3, Steven M. Lewis3, Lisa A. Brown1, Elizabeth M.
    [Show full text]
  • Genotype Imputation Via Matrix Completion
    Downloaded from genome.cshlp.org on September 27, 2021 - Published by Cold Spring Harbor Laboratory Press Method Genotype imputation via matrix completion Eric C. Chi,1,6 Hua Zhou,2 Gary K. Chen,3 Diego Ortega Del Vecchyo,4 and Kenneth Lange5 1Department of Human Genetics, University of California, Los Angeles, California 90095, USA; 2Department of Statistics, North Carolina State University, Raleigh, North Carolina 27695-8203, USA; 3Department of Preventive Medicine, University of Southern California, Los Angeles, California 90089, USA; 4Interdepartmental Program in Bioinformatics, University of California, Los Angeles, California 90095, USA; 5Department of Biomathematics, Department of Human Genetics, and Department of Statistics, University of California, Los Angeles, California 90095, USA Most current genotype imputation methods are model-based and computationally intensive, taking days to impute one chromosome pair on 1000 people. We describe an efficient genotype imputation method based on matrix completion. Our matrix completion method is implemented in MATLAB and tested on real data from HapMap 3, simulated pedigree data, and simulated low-coverage sequencing data derived from the 1000 Genomes Project. Compared with leading im- putation programs, the matrix completion algorithm embodied in our program MENDEL-IMPUTE achieves comparable imputation accuracy while reducing run times significantly. Implementation in a lower-level language such as Fortran or C is apt to further improve computational efficiency. [Supplemental material is available for this article.] Modern genomics studies almost invariably deal with massive the spirit of Occam’s razor, it seeks the simplest matrix consistent amounts of data. Data sets collected on single nucleotide poly- with the observed entries. This criterion conveniently translates morphisms (SNPs), next-generation sequencing (NGS), copy num- into searching for a low rank matrix with a small squared error ber variations (CNV), and RNA-seq all fall into this category.
    [Show full text]
  • Impact of Imputation Methods on the Amount of Genetic Variation Captured by a Single-Nucleotide Polymorphism Panel in Soybeans A
    Xavier et al. BMC Bioinformatics (2016) 17:55 DOI 10.1186/s12859-016-0899-7 METHODOLOGY ARTICLE Open Access Impact of imputation methods on the amount of genetic variation captured by a single-nucleotide polymorphism panel in soybeans A. Xavier1, William M. Muir2 and Katy M. Rainey1* Abstract Background: Success in genome-wide association studies and marker-assisted selection depends on good phenotypic and genotypic data. The more complete this data is, the more powerful will be the results of analysis. Nevertheless, there are next-generation technologies that seek to provide genotypic information in spite of great proportions of missing data. The procedures these technologies use to impute genetic data, therefore, greatly affect downstream analyses. This study aims to (1) compare the genetic variance in a single-nucleotide polymorphism panel of soybean with missing data imputed using various methods, (2) evaluate the imputation accuracy and post-imputation quality associated with these methods, and (3) evaluate the impact of imputation method on heritability and the accuracy of genome-wide prediction of soybean traits. The imputation methods we evaluated were as follows: multivariate mixed model, hidden Markov model, logical algorithm, k-nearest neighbor, single value decomposition, and random forest. We used raw genotypes from the SoyNAM project and the following phenotypes: plant height, days to maturity, grain yield, and seed protein composition. Results: We propose an imputation method based on multivariate mixed models using pedigree information. Our methods comparison indicate that heritability of traits can be affected by the imputation method. Genotypes with missing values imputed with methods that make use of genealogic information can favor genetic analysis of highly polygenic traits, but not genome-wide prediction accuracy.
    [Show full text]
  • References for Haplotype Imputation in the Big Data
    ogy iol : Op r B e a n l A u c c c e l e s Li et al., Mol Biol 2015, 4:4 o s M Molecular Biology: Open Access DOI: 10.4172/2168-9547.1000143 ISSN: 2168-9547 Review Article Open Access References for Haplotype Imputation in the Big Data Era Wenzhi Li1,2#, Wei Xu1,2#, Qiling Li1, Li Ma2,3* and Qing Song1,2,3* 1Center of Big Data and Bioinformatics, First Affiliated Hospital of Medicine School, Xi’an Jiaotong University, Xi’an, Shaanxi, China 2Cardiovascular Research Institute and Department of Medicine, Morehouse School of Medicine, Atlanta, Georgia, USA 34DGenome Inc, Atlanta, Georgia, USA #Equal contribution Abstract Imputation is a powerful in silico approach to fill in those missing values in the big datasets. This process requires a reference panel, which is a collection of big data from which the missing information can be extracted and imputed. Haplotype imputation requires ethnicity-matched references; a mismatched reference panel will significantly reduce the quality of imputation. However, currently existing big datasets cover only a small number of ethnicities, there is a lack of ethnicity-matched references for many ethnic populations in the world, which has hampered the data imputation of haplotypes and its downstream applications. To solve this issue, several approaches have been proposed and explored, including the mixed reference panel, the internal reference panel and genotype-converted reference panel. This review article provides the information and comparison between these approaches. Increasing evidence showed that not just one or two genetic elements dictate the gene activity and functions; instead, cis- interactions of multiple elements dictate gene activity.
    [Show full text]
  • Accurate Genotype Imputation in Multiparental Populations
    Genetics: Early Online, published on July 25, 2018 as 10.1534/genetics.118.300885 1 Accurate genotype imputation in multiparental populations 2 from low-coverage sequence ∗ 3 Chaozhi Zheng , Martin P. Boer, and Fred A. van Eeuwijk 4 Biometris, Wageningen University and Research, Wageningen, The Netherlands 5 July 25, 2018 6 Running head: Genotype imputation 7 Key words: Genotype imputation, genotyping by sequencing, hidden Markov model, cross 8 pollinated (CP) population, multiparent advanced generation intercross (MAGIC) 9 *Corresponding author: 10 Chaozhi Zheng 11 Biometris 12 Wageningen University and Research 13 PO Box 16, 6700AA Wageningen 14 The Netherlands 15 Email: [email protected] 1 Copyright 2018. 16 Abstract 17 Many different types of multiparental populations have recently been produced to increase ge- 18 netic diversity and resolution in quantitative trait loci (QTL) mapping. Low coverage geno- 19 typing by sequencing (GBS) technology has become a cost effective tool in these populations, 20 despite large amounts of missing data in offspring and founders. In this work, we present a 21 general statistical framework for genotype imputation in such experimental crosses from low 22 coverage GBS data. Generalizing a previously developed hidden Markov model for calculating 23 ancestral origins of offspring DNA, we present an imputation algorithm that doesn’t require 24 parental data and that is applicable to bi- and multiparental populations. Our imputation al- 25 gorithm allows heterozygosity of parents and offspring as well as error correction in observed 26 genotypes. Further, our approach can combine imputation and genotype calling from sequenc- 27 ing reads, and it also applies to called genotypes from single nucleotide polymorphism (SNP) 28 array data.
    [Show full text]
  • 1000 Genomes-Based Imputation Identifies Novel and Refined
    European Journal of Human Genetics (2012) 20, 801–805 & 2012 Macmillan Publishers Limited All rights reserved 1018-4813/12 www.nature.com/ejhg SHORT REPORT 1000 Genomes-based imputation identifies novel and refined associations for the Wellcome Trust Case Control Consortium phase 1 Data Jie Huang1, David Ellinghaus2, Andre Franke2, Bryan Howie3 and Yun Li*,4 We hypothesize that imputation based on data from the 1000 Genomes Project can identify novel association signals on a genome-wide scale due to the dense marker map and the large number of haplotypes. To test the hypothesis, the Wellcome Trust Case Control Consortium (WTCCC) Phase I genotype data were imputed using 1000 genomes as reference (20100804 EUR), and seven case/control association studies were performed using imputed dosages. We observed two ‘missed’ disease- associated variants that were undetectable by the original WTCCC analysis, but were reported by later studies after the 2007 WTCCC publication. One is within the IL2RA gene for association with type 1 diabetes and the other in proximity with the CDKN2B gene for association with type 2 diabetes. We also identified two refined associations. One is SNP rs11209026 in exon 9 of IL23R for association with Crohn’s disease, which is predicted to be probably damaging by PolyPhen2. The other refined variant is in the CUX2 gene region for association with type 1 diabetes, where the newly identified top SNP rs1265564 has an association P-value of 1.68Â10À16. The new lead SNP for the two refined loci provides a more plausible explanation for the disease association. We demonstrated that 1000 Genomes-based imputation could indeed identify both novel (in our case, ‘missed’ because they were detected and replicated by studies after 2007) and refined signals.
    [Show full text]
  • The Use of Genomic Data and Imputation Methods in Dairy Cattle Breeding
    Review Czech Journal of Animal Science, 65, 2020 (12): 445–453 https://doi.org/10.17221/83/2020-CJAS The use of genomic data and imputation methods in dairy cattle breeding Anita Klímová1,2*, Eva Kašná2, Karolína Machová1, Michaela Brzáková2, Josef Přibyl2, Luboš Vostrý1 1Department of Genetics and Breeding, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Prague – Suchdol, Czech Republic 2Department of Genetics and Breeding of Farm Animals, Institute of Animal Science, Prague – Uhříněves, Czech Republic *Corresponding author: [email protected] Citation: Klímová A., Kašná E., Machová K., Brzáková M., Přibyl J., Vostrý L. (2020): The use of genomic data and im- putation methods in dairy cattle breeding. Czech J. Anim. Sci., 65: 445–453. Abstract: The inclusion of animal genotype data has contributed to the development of genomic selection. Ani- mals are selected not only based on pedigree and phenotypic data but also on the basis of information about their genotypes. Genomic information helps to increase the accuracy of selection of young animals and thus enables a reduction of the generation interval. Obtaining information about genotypes in the form of SNPs (single nucleotide polymorphisms) has led to the development of new chips for genotyping. Several methods of genomic comparison have been developed as a result. One of the methods is data imputation, which allows the missing SNPs to be calculated using low-density chips to high-density chips. Through imputations, it is possible to com- bine information from diverse sets of chips and thus obtain more information about genotypes at a lower cost.
    [Show full text]
  • Imputation Accuracy from Low to Moderate Density Single Nucleotide Polymorphism Chips in a Thai Multibreed Dairy Cattle Population
    464 Open Access Asian Australas. J. Anim. Sci. Vol. 29, No. 4 : 464-470 April 2016 http://dx.doi.org/10.5713/ajas.15.0291 www.ajas.info pISSN 1011-2367 eISSN 1976-5517 Imputation Accuracy from Low to Moderate Density Single Nucleotide Polymorphism Chips in a Thai Multibreed Dairy Cattle Population Danai Jattawa, Mauricio A. Elzo1, Skorn Koonawootrittriron*, and Thanathip Suwanasopee Department of Animal Science, Faculty of Agriculture, Kasetsart University, Bangkok 10900, Thailand ABSTRACT: The objective of this study was to investigate the accuracy of imputation from low density (LDC) to moderate density SNP chips (MDC) in a Thai Holstein-Other multibreed dairy cattle population. Dairy cattle with complete pedigree information (n = 1,244) from 145 dairy farms were genotyped with GeneSeek GGP20K (n = 570), GGP26K (n = 540) and GGP80K (n = 134) chips. After checking for single nucleotide polymorphism (SNP) quality, 17,779 SNP markers in common between the GGP20K, GGP26K, and GGP80K were used to represent MDC. Animals were divided into two groups, a reference group (n = 912) and a test group (n = 332). The SNP markers chosen for the test group were those located in positions corresponding to GeneSeek GGP9K (n = 7,652). The LDC to MDC genotype imputation was carried out using three different software packages, namely Beagle 3.3 (population-based algorithm), FImpute 2.2 (combined family- and population-based algorithms) and Findhap 4 (combined family- and population-based algorithms). Imputation accuracies within and across chromosomes were calculated as ratios of correctly imputed SNP markers to overall imputed SNP markers. Imputation accuracy for the three software packages ranged from 76.79% to 93.94%.
    [Show full text]
  • Odyssey: a Semi-Automated Pipeline for Phasing, Imputation, and Analysis of Genome-Wide Genetic Data Ryan J
    Eller et al. BMC Bioinformatics (2019) 20:364 https://doi.org/10.1186/s12859-019-2964-5 METHODOLOGY ARTICLE Open Access Odyssey: a semi-automated pipeline for phasing, imputation, and analysis of genome-wide genetic data Ryan J. Eller1* , Sarath C. Janga2,3 and Susan Walsh1 Abstract Background: Genome imputation, admixture resolution and genome-wide association analyses are timely and computationally intensive processes with many composite and requisite steps. Analysis time increases further when building and installing the run programs required for these analyses. For scientists that may not be as versed in programing language, but want to perform these operations hands on, there is a lengthy learning curve to utilize the vast number of programs available for these analyses. Results: In an effort to streamline the entire process with easy-to-use steps for scientists working with big data, the Odyssey pipeline was developed. Odyssey is a simplified, efficient, semi-automated genome-wide imputation and analysis pipeline, which prepares raw genetic data, performs pre-imputation quality control, phasing, imputation, post-imputation quality control, population stratification analysis, and genome-wide association with statistical data analysis, including result visualization. Odyssey is a pipeline that integrates programs such as PLINK, SHAPEIT, Eagle, IMPUTE, Minimac, and several R packages, to create a seamless, easy-to-use, and modular workflow controlled via a single user-friendly configuration file. Odyssey was built with compatibility in mind, and thus utilizes the Singularity container solution, which can be run on Linux, MacOS, and Windows platforms. It is also easily scalable from a simple desktop to a High-Performance System (HPS).
    [Show full text]